Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 70
Filter
Add more filters

Country/Region as subject
Publication year range
1.
N Engl J Med ; 384(13): 1216-1226, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33789010

ABSTRACT

BACKGROUND: Primary hyperoxaluria type 1 (PH1) is a rare genetic disease caused by hepatic overproduction of oxalate that leads to kidney stones, nephrocalcinosis, kidney failure, and systemic oxalosis. Lumasiran, an investigational RNA interference (RNAi) therapeutic agent, reduces hepatic oxalate production by targeting glycolate oxidase. METHODS: In this double-blind, phase 3 trial, we randomly assigned (in a 2:1 ratio) patients with PH1 who were 6 years of age or older to receive subcutaneous lumasiran or placebo for 6 months (with doses given at baseline and at months 1, 2, 3, and 6). The primary end point was the percent change in 24-hour urinary oxalate excretion from baseline to month 6 (mean percent change across months 3 through 6). Secondary end points included the percent change in the plasma oxalate level from baseline to month 6 (mean percent change across months 3 through 6) and the percentage of patients with 24-hour urinary oxalate excretion no higher than 1.5 times the upper limit of the normal range at month 6. RESULTS: A total of 39 patients underwent randomization; 26 were assigned to the lumasiran group and 13 to the placebo group. The least-squares mean difference in the change in 24-hour urinary oxalate excretion (lumasiran minus placebo) was -53.5 percentage points (P<0.001), with a reduction in the lumasiran group of 65.4% and an effect seen as early as month 1. The between-group differences for all hierarchically tested secondary end points were significant. The difference in the percent change in the plasma oxalate level (lumasiran minus placebo) was -39.5 percentage points (P<0.001). In the lumasiran group, 84% of patients had 24-hour urinary oxalate excretion no higher than 1.5 times the upper limit of the normal range at month 6, as compared with 0% in the placebo group (P<0.001). Mild, transient injection-site reactions were reported in 38% of lumasiran-treated patients. CONCLUSIONS: Lumasiran reduced urinary oxalate excretion, the cause of progressive kidney failure in PH1. The majority of patients who received lumasiran had normal or near-normal levels after 6 months of treatment. (Funded by Alnylam Pharmaceuticals; ILLUMINATE-A ClinicalTrials.gov number, NCT03681184.).


Subject(s)
Hyperoxaluria, Primary/drug therapy , Oxalates/urine , RNA, Small Interfering/therapeutic use , RNAi Therapeutics , Adolescent , Adult , Child , Creatinine/urine , Double-Blind Method , Female , Glomerular Filtration Rate , Humans , Hyperoxaluria, Primary/blood , Hyperoxaluria, Primary/complications , Hyperoxaluria, Primary/urine , Kidney Calculi/prevention & control , Male , Middle Aged , Oxalates/blood , Oxalates/metabolism , RNA, Small Interfering/adverse effects , Young Adult
2.
Am J Hum Genet ; 103(5): 808-816, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30388404

ABSTRACT

Over the last decades, a growing spectrum of monogenic disorders of human magnesium homeostasis has been clinically characterized, and genetic studies in affected individuals have identified important molecular components of cellular and epithelial magnesium transport. Here, we describe three infants who are from non-consanguineous families and who presented with a disease phenotype consisting of generalized seizures in infancy, severe hypomagnesemia, and renal magnesium wasting. Seizures persisted despite magnesium supplementation and were associated with significant intellectual disability. Whole-exome sequencing and conventional Sanger sequencing identified heterozygous de novo mutations in the catalytic Na+, K+-ATPase α1 subunit (ATP1A1). Functional characterization of mutant Na+, K+-ATPase α1 subunits in heterologous expression systems revealed not only a loss of Na+, K+-ATPase function but also abnormal cation permeabilities, which led to membrane depolarization and possibly aggravated the effect of the loss of physiological pump activity. These findings underline the indispensable role of the α1 isoform of the Na+, K+-ATPase for renal-tubular magnesium handling and cellular ion homeostasis, as well as maintenance of physiologic neuronal activity.


Subject(s)
Intellectual Disability/genetics , Mutation/genetics , Renal Tubular Transport, Inborn Errors/genetics , Seizures/genetics , Sodium-Potassium-Exchanging ATPase/genetics , Child , Child, Preschool , Female , Germ Cells , Heterozygote , Homeostasis/genetics , Humans , Infant , Infant, Newborn , Kidney/pathology , Magnesium/metabolism , Male , Phenotype , Protein Isoforms/genetics
3.
N Engl J Med ; 378(21): 1987-1998, 2018 05 24.
Article in English | MEDLINE | ID: mdl-29791829

ABSTRACT

BACKGROUND: X-linked hypophosphatemia is characterized by increased secretion of fibroblast growth factor 23 (FGF-23), which leads to hypophosphatemia and consequently rickets, osteomalacia, and skeletal deformities. We investigated burosumab, a monoclonal antibody that targets FGF-23, in patients with X-linked hypophosphatemia. METHODS: In an open-label, phase 2 trial, we randomly assigned 52 children with X-linked hypophosphatemia, in a 1:1 ratio, to receive subcutaneous burosumab either every 2 weeks or every 4 weeks; the dose was adjusted to achieve a serum phosphorus level at the low end of the normal range. The primary end point was the change from baseline to weeks 40 and 64 in the Thacher rickets severity total score (ranging from 0 to 10, with higher scores indicating greater disease severity). In addition, the Radiographic Global Impression of Change was used to evaluate rachitic changes from baseline to week 40 and to week 64. Additional end points were changes in pharmacodynamic markers, linear growth, physical ability, and patient-reported outcomes and the incidence of adverse events. RESULTS: The mean Thacher rickets severity total score decreased from 1.9 at baseline to 0.8 at week 40 with every-2-week dosing and from 1.7 at baseline to 1.1 at week 40 with every-4-week dosing (P<0.001 for both comparisons); these improvements persisted at week 64. The mean serum phosphorus level increased after the first dose in both groups, and more than half the patients in both groups had levels within the normal range (3.2 to 6.1 mg per deciliter [1.0 to 2.0 mmol per liter]) by week 6. Stable serum phosphorus levels were maintained through week 64 with every-2-week dosing. Renal tubular phosphate reabsorption increased from baseline in both groups, with an overall mean increase of 0.98 mg per deciliter (0.32 mmol per liter). The mean dose of burosumab at week 40 was 0.98 mg per kilogram of body weight with every-2-week dosing and 1.50 mg per kilogram with every-4-week dosing. Across both groups, the mean serum alkaline phosphatase level decreased from 459 U per liter at baseline to 369 U per liter at week 64. The mean standing-height z score increased in both groups, with greater improvement seen at all time points with every-2-week dosing (an increase from baseline of 0.19 at week 64) than with every-4-week dosing (an increase from baseline of 0.12 at week 64). Physical ability improved and pain decreased. Nearly all the adverse events were mild or moderate in severity. CONCLUSIONS: In children with X-linked hypophosphatemia, treatment with burosumab improved renal tubular phosphate reabsorption, serum phosphorus levels, linear growth, and physical function and reduced pain and the severity of rickets. (Funded by Ultragenyx Pharmaceutical and Kyowa Hakko Kirin; ClinicalTrials.gov number, NCT02163577 ; EudraCT number, 2014-000406-35 ).


Subject(s)
Antibodies, Monoclonal/therapeutic use , Familial Hypophosphatemic Rickets/drug therapy , Fibroblast Growth Factors/antagonists & inhibitors , Genetic Diseases, X-Linked/drug therapy , Alkaline Phosphatase/blood , Antibodies, Monoclonal, Humanized , Child , Child, Preschool , Familial Hypophosphatemic Rickets/metabolism , Familial Hypophosphatemic Rickets/physiopathology , Female , Fibroblast Growth Factor-23 , Genetic Diseases, X-Linked/metabolism , Genetic Diseases, X-Linked/physiopathology , Growth/drug effects , Humans , Kidney Tubules/metabolism , Knee Joint/diagnostic imaging , Male , Pain Management , Phosphorus/blood , Radiography , Severity of Illness Index
4.
Pediatr Transplant ; 23(4): e13411, 2019 06.
Article in English | MEDLINE | ID: mdl-30973673

ABSTRACT

BACKGROUND: In current practice, pediatric kidney transplant recipients receive large volumes of intravenous fluid intraoperatively to establish allograft perfusion, and further fluid to replace urinary and insensible losses postoperatively. Acute electrolyte imbalance can result, with potential for neurological sequelae. We aimed to determine the incidence and severity of postoperative plasma electrolyte imbalance in pediatric kidney transplant recipients managed with the current standard intravenous crystalloid regimen. METHODS: A retrospective analysis of plasma electrolytes in the first 72 hours post-kidney transplant in 76 children transplanted between January 1, 2015, and January 31, 2018, managed with a standard intravenous fluid strategy used in most UK pediatric transplant centers. RESULTS: Of 76 pediatric transplant recipients of median age 9.9 (range 2.2-17.9) years predominantly managed with 0.45% sodium chloride 5% glucose, 45 (59%) developed acute hyponatremia, 23 (30%) hyperkalemia, and 43 (57%) non-anion-gap acidosis in the postoperative period. Hyperglycemia occurred in 74 (97%) patients. Hyperkalemia was more prevalent in deceased than live donor recipients (P = 0.003) and was significantly associated with non-anion-gap acidosis (P < 0.001). Recipient weight was not associated with overt electrolyte imbalance. CONCLUSION: Postoperative plasma electrolyte imbalance is common in pediatric kidney transplant recipients. Current clinical care strategies mitigate the associated risks of neurological sequelae to some degree. Further studies to optimize intravenous fluid therapy and minimize electrolyte disturbance in this group of patients are needed.


Subject(s)
Electrolytes/blood , Kidney Failure, Chronic/blood , Kidney Failure, Chronic/surgery , Kidney Transplantation , Acid-Base Equilibrium , Adolescent , Allografts , Child , Child, Preschool , Glucose/administration & dosage , Humans , Incidence , Infusions, Intravenous , Perfusion , Postoperative Complications/therapy , Postoperative Period , Retrospective Studies , Sodium Chloride/administration & dosage
5.
J Am Soc Nephrol ; 29(3): 1041-1048, 2018 03.
Article in English | MEDLINE | ID: mdl-29242249

ABSTRACT

Maintenance of the composition of inner ear fluid and regulation of electrolytes and acid-base homeostasis in the collecting duct system of the kidney require an overlapping set of membrane transport proteins regulated by the forkhead transcription factor FOXI1. In two unrelated consanguineous families, we identified three patients with novel homozygous missense mutations in FOXI1 (p.L146F and p.R213P) predicted to affect the highly conserved DNA binding domain. Patients presented with early-onset sensorineural deafness and distal renal tubular acidosis. In cultured cells, the mutations reduced the DNA binding affinity of FOXI1, which hence, failed to adequately activate genes crucial for normal inner ear function and acid-base regulation in the kidney. A substantial proportion of patients with a clinical diagnosis of inherited distal renal tubular acidosis has no identified causative mutations in currently known disease genes. Our data suggest that recessive mutations in FOXI1 can explain the disease in a subset of these patients.


Subject(s)
Acidosis, Renal Tubular/genetics , Deafness/genetics , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Cells, Cultured , Child , Consanguinity , DNA/metabolism , Deafness/complications , Female , Hearing Loss, Central/genetics , Homozygote , Humans , Infant , Kidney Tubules, Distal/metabolism , Male , Mutation, Missense , Pedigree
6.
Kidney Int ; 93(4): 961-967, 2018 04.
Article in English | MEDLINE | ID: mdl-29398133

ABSTRACT

The clinical diagnosis of inherited renal tubulopathies can be challenging as they are rare and characterized by significant phenotypic variability. Advances in sequencing technologies facilitate the establishment of a molecular diagnosis. Therefore, we determined the diagnostic yield of a next generation sequencing panel assessing relevant disease genes in children followed through three national networks with a clinical diagnosis of a renal tubulopathy. DNA was amplified with a kit provided by the European Consortium for High-Throughput Research in Rare Kidney Diseases with nine multiplex PCR reactions. This kit produced 571 amplicons covering 37 genes associated with tubulopathies followed by massive parallel sequencing and bioinformatic interpretation. Identified mutations were confirmed by Sanger sequencing. Overall, 384 index patients and 16 siblings were assessed. Most common clinical diagnoses were 174 patients with Bartter/Gitelman syndrome and 76 with distal renal tubular acidosis. A total of 269 different variants were identified in 27 genes, of which 95 variants were considered likely, 136 definitely pathogenic and 100 had not been described at annotation. These mutations established a genetic diagnosis in 245 of the index patients. Genetic testing changed the clinical diagnosis in 16 cases and provided insights into the phenotypic spectrum of the respective disorders. Our results demonstrate a high diagnostic yield of genetic testing in children with a clinical diagnosis of a renal tubulopathy, consistent with a predominantly genetic etiology in known disease genes. Thus, genetic testing helped establish a definitive diagnosis in almost two-thirds of patients thereby informing prognosis, management and genetic counseling.


Subject(s)
DNA Mutational Analysis/methods , High-Throughput Nucleotide Sequencing , Mutation , Renal Tubular Transport, Inborn Errors/genetics , Acidosis, Renal Tubular/diagnosis , Acidosis, Renal Tubular/genetics , Adolescent , Age Factors , Bartter Syndrome/diagnosis , Bartter Syndrome/genetics , Case-Control Studies , Child , Child, Preschool , Europe , Female , Genetic Markers , Genetic Predisposition to Disease , Gitelman Syndrome/diagnosis , Gitelman Syndrome/genetics , Heredity , Humans , Infant , Infant, Newborn , Male , Multiplex Polymerase Chain Reaction , Pedigree , Phenotype , Predictive Value of Tests , Reagent Kits, Diagnostic , Renal Tubular Transport, Inborn Errors/diagnosis , Risk Factors
7.
Hum Mol Genet ; 24(18): 5079-92, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26082470

ABSTRACT

The adaptor protein-2 sigma subunit (AP2σ2) is pivotal for clathrin-mediated endocytosis of plasma membrane constituents such as the calcium-sensing receptor (CaSR). Mutations of the AP2σ2 Arg15 residue result in familial hypocalciuric hypercalcaemia type 3 (FHH3), a disorder of extracellular calcium (Ca(2+) o) homeostasis. To elucidate the role of AP2σ2 in Ca(2+) o regulation, we investigated 65 FHH probands, without other FHH-associated mutations, for AP2σ2 mutations, characterized their functional consequences and investigated the genetic mechanisms leading to FHH3. AP2σ2 mutations were identified in 17 probands, comprising 5 Arg15Cys, 4 Arg15His and 8 Arg15Leu mutations. A genotype-phenotype correlation was observed with the Arg15Leu mutation leading to marked hypercalcaemia. FHH3 probands harboured additional phenotypes such as cognitive dysfunction. All three FHH3-causing AP2σ2 mutations impaired CaSR signal transduction in a dominant-negative manner. Mutational bias was observed at the AP2σ2 Arg15 residue as other predicted missense substitutions (Arg15Gly, Arg15Pro and Arg15Ser), which also caused CaSR loss-of-function, were not detected in FHH probands, and these mutations were found to reduce the numbers of CaSR-expressing cells. FHH3 probands had significantly greater serum calcium (sCa) and magnesium (sMg) concentrations with reduced urinary calcium to creatinine clearance ratios (CCCR) in comparison with FHH1 probands with CaSR mutations, and a calculated index of sCa × sMg/100 × CCCR, which was ≥ 5.0, had a diagnostic sensitivity and specificity of 83 and 86%, respectively, for FHH3. Thus, our studies demonstrate AP2σ2 mutations to result in a more severe FHH phenotype with genotype-phenotype correlations, and a dominant-negative mechanism of action with mutational bias at the Arg15 residue.


Subject(s)
Adaptor Protein Complex 2/genetics , Adaptor Protein Complex sigma Subunits/genetics , Codon , Genes, Dominant , Genetic Association Studies , Hypercalcemia/congenital , Mutation , Adaptor Protein Complex 2/chemistry , Adaptor Protein Complex sigma Subunits/chemistry , Adolescent , Adult , Amino Acid Substitution , Biomarkers , Cell Line , Child , Child, Preschool , Diagnosis, Differential , Female , Gene Expression , Humans , Hypercalcemia/diagnosis , Hypercalcemia/genetics , Infant , Male , Middle Aged , Models, Molecular , Pedigree , Phenotype , Protein Conformation , Structure-Activity Relationship , Young Adult
8.
Pediatr Nephrol ; 32(5): 781-790, 2017 05.
Article in English | MEDLINE | ID: mdl-27924398

ABSTRACT

BACKGROUND: Primary hyperoxaluria (PH) is a rare, genetic disorder which involves the overproduction of endogenous oxalate, leading to hyperoxaluria, recurrent urolithiasis and/or progressive nephrocalcinosis and eventually resulting in kidney failure and systemic oxalosis. The aim of this trial was to investigate whether treatment involving an oxalate-metabolising bacterium (Oxalobacter formigenes) could reduce urinary oxalate excretion in PH patients. METHODS: The efficacy and safety of O. formigenes (Oxabact® OC5; OxThera AB, Stockholm, Sweden) was evaluated in a randomised, placebo-controlled, double-blind study for 8 weeks. The primary objective was reduction in urinary oxalate excretion (Uox). Secondary objectives included faecal O. formigenes count and decrease in plasma oxalate concentration (Pox). RESULTS: Twenty-eight patients randomised 1:1 to the treatment group (OC5) or the placebo group completed the study. After 8 weeks of treatment, there was no significant difference in the change in Uox (mmol/24 h/1.73 m2) between the groups (OC5: +0.042, placebo: -0.140). Post-hoc analysis showed a statistically significant increase in Uox per urinary creatinine excretion in the OC5 group (OC5: +5.41, placebo: -15.96; p = 0.030). Change in Pox from baseline was not significantly different between groups (p = 0.438). The O. formigenes cell count was significantly increased in OC5-treated patients (p < 0.001) versus placebo. The treatment response to O. formigenes was related to individual stage of kidney deterioration, and Pox was directly correlated to kidney function, even for early-stage patients (chronic kidney disease stage 1). No safety issues were observed. CONCLUSIONS: Treatment with OC5 did not significantly reduce Uox or Pox over 8 weeks of treatment. The treatment was well tolerated and successfully delivered to the gastrointestinal tract.


Subject(s)
Hyperoxaluria/therapy , Oxalobacter formigenes , Adolescent , Bacterial Load , Child , Child, Preschool , Double-Blind Method , Feces/microbiology , Female , Humans , Hyperoxaluria/physiopathology , Hyperoxaluria/urine , Kidney Function Tests , Male , Oxalic Acid/urine , Probiotics/administration & dosage , Probiotics/adverse effects , Probiotics/therapeutic use , Tablets, Enteric-Coated , Treatment Outcome , Young Adult
9.
Pediatr Nephrol ; 32(6): 987-996, 2017 06.
Article in English | MEDLINE | ID: mdl-28188436

ABSTRACT

BACKGROUND: Distal renal tubular acidosis (dRTA) is characterized by hyperchloraemic metabolic acidosis, hypokalaemia, hypercalciuria and nephrocalcinosis. It is due to reduced urinary acidification by the α-intercalated cells in the collecting duct and can be caused by mutations in genes that encode subunits of the vacuolar H+-ATPase (ATP6V1B1, ATP6V0A4) or the anion exchanger 1 (SLC4A1). Treatment with alkali is the mainstay of therapy. METHODS: This study is an analysis of clinical data from a long-term follow-up of 24 children with dRTA in a single centre, including a genetic analysis. RESULTS: Of the 24 children included in the study, genetic diagnosis was confirmed in 19 patients, with six children having mutations in ATP6V1B1, ten in ATP6V0A4 and three in SLC4A1; molecular diagnosis was not available for five children. Five novel mutations were detected (2 in ATP6V1B1 and 3 in ATP6V0A4). Two-thirds of patients presented with features of proximal tubular dysfunction leading to an erroneous diagnosis of renal Fanconi syndrome. The proximal tubulopathy disappeared after resolution of acidosis, indicating the importance of following proximal tubular function to establish the correct diagnosis. Growth retardation with a height below -2 standard deviation score was found in ten patients at presentation, but persisted in only three of these children once established on alkali treatment. Sensorineural hearing loss was found in five of the six patients with an ATP6V1B1 mutation. Only one patient with an ATP6V0A4 mutation had sensorineural hearing loss during childhood. Nine children developed medullary cysts, but without apparent clinical consequences. Cyst development in this cohort was not correlated with age at therapy onset, molecular diagnosis, growth parameters or renal function. CONCLUSION: In general, the prognosis of dRTA is good in children treated with alkali.


Subject(s)
Acidosis, Renal Tubular/epidemiology , Acidosis, Renal Tubular/genetics , Alkalies/therapeutic use , Growth Disorders/epidemiology , Hearing Loss, Sensorineural/epidemiology , Kidney Tubules, Collecting/metabolism , Acidosis, Renal Tubular/drug therapy , Anion Exchange Protein 1, Erythrocyte/genetics , Child, Preschool , Cohort Studies , Comorbidity , Cysts/epidemiology , Cysts/genetics , DNA Mutational Analysis , Female , Follow-Up Studies , Genetic Testing , Glomerular Filtration Rate , Growth Disorders/genetics , Hearing Loss, Sensorineural/genetics , Humans , Infant , Infant, Newborn , Kidney Medulla/pathology , Kidney Tubules, Collecting/cytology , Male , Mutation , Vacuolar Proton-Translocating ATPases/genetics
10.
BMC Nephrol ; 18(1): 136, 2017 Apr 18.
Article in English | MEDLINE | ID: mdl-28420322

ABSTRACT

BACKGROUND: Whilst still rare, the incidence of paediatric stone disease is increasing in developed countries and it is important to evaluate the aetiology. We set up a dedicated renal stone service for children combining medical and surgical expertise in 1993 and now have a large case series of children to investigate the epidemiology. METHODS: A retrospective hospital note review of children presenting with kidney stones during the last 22 years (1993-2015) was conducted. All patients had a comprehensive infective and metabolic screen and were classified as metabolic, infective or idiopathic stone disease. RESULTS: Five hundred eleven patients (322 male) were reviewed. The median age of presentation was 4.4y for males (1 m-16.6y) and 7.3y (1-18.5y) for females with a median height and weight on the 25th centile for male and on 10th and 25th for female, respectively. One hundred seventy five (34%) had an underlying metabolic abnormality, 112 (22%) had infective stones and 224 (44%) were classified as idiopathic. Of the 175 patients with a metabolic abnormality: 91 (52%) had hypercalciuria (76 persistent and 15 transient), 37 (21%) hyperoxaluria, 38 (22%) cystinuria, 3 (2%) abnormalities in the purine metabolism and the remainder other metabolic abnormalities. Bilateral stones occurred in 27% of the metabolic group compared to 16% in the non-metabolic group (OR 0.2, p < 0.05). Urinary tract infection was a common complication (27%) in the metabolic group. CONCLUSIONS: In this paper, we present the largest cohort of paediatric stone disease reported from a developed country giving details on both, clinical and laboratory data. We show that in the majority of the patients there is an identifiable underlying metabolic and/or infective aetiology emphasizing the importance of a full work up to provide adequate treatment and prevent recurrence. Moreover, we show that stone disease in children, in contrast to the adult population, does not seem to be associated with obesity, as children have a weight below average at presentation.


Subject(s)
Kidney Calculi/diagnosis , Kidney Calculi/epidemiology , Metabolic Diseases/diagnosis , Metabolic Diseases/epidemiology , Urinary Tract Infections/diagnosis , Urinary Tract Infections/epidemiology , Adolescent , Age Distribution , Causality , Child , Child, Preschool , Comorbidity , Female , Humans , Infant , Infant, Newborn , Longitudinal Studies , Male , Prevalence , Risk Factors , Sex Distribution
11.
BMC Nephrol ; 18(1): 230, 2017 Jul 11.
Article in English | MEDLINE | ID: mdl-28693455

ABSTRACT

BACKGROUND: Rare diseases may elude diagnosis due to unfamiliarity of the treating physicians with the specific disorder. Yet, advances in genetics have tremendously enhanced our ability to establish specific and sometimes surprising diagnoses. CASE PRESENTATION: We report a case of renal Fanconi syndrome associated with intermittent hypoglycemic episodes, the specific cause for which remained elusive for over 30 years, despite numerous investigations, including three kidney and one liver biopsy. The most recent kidney biopsy showed dysmorphic mitochondria, suggesting a mitochondrial disorder. When her son presented with hypoglycemia in the neonatal period, he underwent routine genetic testing for hyperinsulinemic hypoglycemia, which revealed a specific mutation in HNF4A. Subsequent testing of the mother confirmed the diagnosis also in her. CONCLUSION: Modern sequencing technologies that test multiple genes simultaneously enable specific diagnoses, even if the underlying disorder was not clinically suspected. The finding of mitochondrial dysmorphology provides a potential clue for the mechanism, by which the identified mutation causes renal Fanconi syndrome.


Subject(s)
Fanconi Syndrome/diagnosis , Fanconi Syndrome/genetics , Hepatocyte Nuclear Factor 4/genetics , Proxy , Syncope/diagnosis , Syncope/genetics , Adult , Fanconi Syndrome/complications , Female , Follow-Up Studies , Genetic Testing/trends , Humans , Infant, Newborn , Male , Syncope/complications
12.
Pediatr Transplant ; 20(4): 523-9, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27061278

ABSTRACT

Primary hyperoxaluria type 1 (PH1) is an inherited metabolic disease that culminates in ESRF. Pre-emptive liver transplantation (pLTx) treats the metabolic defect and avoids the need for kidney transplantation (KTx). An institutional experience of pediatric PH1 LTx is reported and compared to the literature. Between 2004 and 2015, eight children underwent pLTx for PH1. Three underwent pLTx with a median GFR of 40 (30-46) mL/min/1.73 m(2) and five underwent sequential combined liver-kidney transplantation (cLKTx); all were on RRT at the time of cLKTx. In one case of pLTx, KTx was required eight and a half yr later. pLTx was performed in older (median 8 vs. 2 yr) and larger children (median 27 vs. 7.75 kg) that had a milder PH1 phenotype. In pediatric PH1, pLTx, ideally, should be performed before renal and extrarenal systemic oxalosis complications have occurred, and pLTx can be used "early" or "late." Early is when renal function is preserved with the aim to avoid renal replacement. However, in late (GFR < 30 mL/min/1.73 m(2) ), the aim is to stabilize renal function and delay the need for KTx. Ultimately, transplant strategy depends on PH1 phenotype, disease stage, child size, and organ availability.


Subject(s)
Hyperoxaluria, Primary/surgery , Kidney Failure, Chronic/prevention & control , Liver Transplantation , Adolescent , Child , Child, Preschool , Female , Follow-Up Studies , Humans , Hyperoxaluria, Primary/complications , Infant , Kidney Failure, Chronic/etiology , Kidney Transplantation , Male , Retrospective Studies , Time Factors , Treatment Outcome
13.
Acta Paediatr ; 105(7): e334-7, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27028950

ABSTRACT

AIM: Using fluid restriction to treat the syndrome of inappropriate antidiuretic hormone secretion (SIADH) in infants is potentially hazardous, as fluid intake and caloric intake are connected. Antagonists for the type 2 vasopressin receptor have demonstrated efficacy in adult patients with SIADH, but evidence in children is lacking. We reviewed our experience from two cases in the UK. METHODS: This was a retrospective review of the clinical data on two patients diagnosed with SIADH in infancy and treated with tolvaptan, an oral vasopressin receptor antagonist. RESULTS: Persistent hyponatraemia was noted in both patients in the first month of life and eventually led to SIADH diagnoses. Initial salt supplementation in one patient resulted in severe hypertension, treated with four antihypertensive drugs. Tolvaptan was commenced at two and four months of age, respectively, and was associated with normalisation of plasma sodium values and blood pressure without the need for antihypertensive treatment. There was transient hypernatraemia in one patient, which was normalised with a dose reduction. Tolvaptan was administered by crushing the tablet and mixing it with water. CONCLUSION: Tolvaptan provided effective treatment for SIADH in both infants and could be administered orally.


Subject(s)
Antidiuretic Hormone Receptor Antagonists/therapeutic use , Benzazepines/therapeutic use , Inappropriate ADH Syndrome/drug therapy , Female , Humans , Infant, Newborn , Male , Retrospective Studies , Tolvaptan
14.
Pharm Res ; 32(2): 590-603, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25190007

ABSTRACT

PURPOSE: To explore the potential of non-invasive reverse iontophoresis transdermal extraction of iohexol as a marker of glomerular filtration rate. METHODS: A series of in vitro experiments were undertaken to establish the feasibility of iohexol reverse iontophoresis and to determine the optimal conditions for the approach. Subsequently, a pilot study in paediatric patients was performed to provide proof-of-concept. RESULTS: The iontophoretic extraction fluxes of iohexol in vitro were proportional to the marker subdermal concentration and the reverse iontophoretic technique was able to track changes dynamically in simulated pharmacokinetic profiles. Reverse iontophoresis sampling was well tolerated by the four paediatric participants. The deduced values of the iohexol terminal elimination rate constant from transdermal reverse iontophoresis sampling agreed with those estimated by conventional blood sampling. CONCLUSIONS: Reverse iontophoretic transdermal extraction fluxes mirrored the subdermal concentration profiles of iohexol, a relatively large neutral marker of glomerular filtration both in vitro and in vivo. The efficiency of extraction in vivo was well predicted by the in vitro model used.


Subject(s)
Glomerular Filtration Rate/drug effects , Glomerular Filtration Rate/physiology , Iohexol/metabolism , Iontophoresis/methods , Skin Absorption/drug effects , Skin Absorption/physiology , Administration, Cutaneous , Adolescent , Animals , Child , Female , Humans , Iohexol/administration & dosage , Male , Organ Culture Techniques , Pilot Projects , Swine
15.
Pediatr Nephrol ; 30(2): 317-25, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25119682

ABSTRACT

BACKGROUND: Vitamin A accumulates in renal failure, but the prevalence of hypervitaminosis A in children with predialysis chronic kidney disease (CKD) is not known. Hypervitaminosis A has been associated with hypercalcemia. In this study we compared dietary vitamin A intake with serum retinoid levels and their associations with hypercalcemia. METHODS: We studied the relationship between vitamin A intake, serum retinoid levels, and serum calcium in 105 children with CKD stages 2-5 on dialysis and posttransplant. Serum retinoid measures included retinol (ROH), its active retinoic acid (RA) metabolites [all-trans RA (at-RA) and 13-cis RA] and carrier proteins [retinol-binding protein-4 (RBP4) and transthyretin (TTR)]. Dietary vitamin A intake was assessed using a food diary. RESULTS: Twenty-five children were in CKD 2-3, 35 in CKD 4-5, 23 on dialysis and 22 posttransplant; 53 % had vitamin A intake above the Reference Nutrient Intake (RNI) value. Children receiving supplemental feeds compared with diet alone had higher vitamin A intake (p = 0.02) and higher serum ROH (p < 0.001). Notably, increased ROH was seen as early as CKD stage 2. For every 10 ml/min/1.73 m(2) fall in estimated glomerular filtration rate (eGFR), there was a 13 % increase in ROH. RBP4 levels were increased in CKD 3-5 and dialysis patients. The lowest ratios of ROH:RBP4 were seen in dialysis compared with CKD 2-3 (p = 0.03), suggesting a relative increase in circulating RBP4. Serum ROH, RBP4 and at-RA were associated with serum calcium. On multivariable analysis RBP4 levels and alfacalcidol dose were significant predictors of serum calcium (model R (2) 32 %) in dialysis patients. CONCLUSIONS: Hypervitaminosis A is seen in early CKD, with highest levels in children on supplemental feeds compared with diet alone. Serum retinoid levels significantly predict hypercalcemia.


Subject(s)
Hypercalcemia/etiology , Hypervitaminosis A/complications , Renal Insufficiency, Chronic/blood , Renal Insufficiency, Chronic/complications , Adolescent , Child , Child, Preschool , Chromatography, High Pressure Liquid , Diet , Female , Humans , Infant , Male
16.
Clin Sci (Lond) ; 126(10): 721-6, 2014 May.
Article in English | MEDLINE | ID: mdl-24266877

ABSTRACT

The study of families with rare inherited forms of hypo- and hyper-tension has been one of the most successful strategies to probe the molecular pathophysiology of blood pressure control and has revealed dysregulation of distal nephron Na+ reabsorption to be a common mechanism. FHHt (familial hyperkalaemic hypertension; also known as Gordon's syndrome) is a salt-dependent form of hypertension caused by mutations in the regulators of the thiazide-sensitive Na+-Cl- co-transporter NCC [also known as SLC12A3 (solute carrier family 12 member 3)] and is effectively treated by thiazide diuretics and/or dietary salt restriction. Variation in at least four genes can cause FHHt, including WNK1 [With No lysine (=K) 1] and WNK4, KLHL3 (kelch-like family member 3), and CUL3 (cullin 3). In the present study we have identified novel disease-causing variants in CUL3 and KLHL3 segregating in 63% of the pedigrees with previously unexplained FHHt, confirming the importance of these recently described FHHt genes. We have demonstrated conclusively, in two unrelated affected individuals, that rare intronic variants in CUL3 cause the skipping of exon 9 as has been proposed previously. KLHL3 variants all occur in kelch-repeat domains and so probably disrupt WNK complex binding. We have found no evidence of any plausible disease-causing variants within SLC4A8 (an alternative thiazide-sensitive sodium transporter) in this population. The results of the present study support the existing evidence that the CUL3 and KLHL3 gene products are physiologically important regulators of thiazide-sensitive distal nephron NaCl reabsorption, and hence potentially interesting novel anti-hypertensive drug targets. As a third of our non-WNK FHHt families do not have plausible CUL3 or KLHL3 variants, there are probably additional, as yet undiscovered, regulators of the thiazide-sensitive pathways.


Subject(s)
Carrier Proteins/genetics , Cullin Proteins/genetics , Genetic Predisposition to Disease , Mutation/genetics , Pseudohypoaldosteronism/genetics , Adaptor Proteins, Signal Transducing , Alternative Splicing/genetics , Chromosome Segregation/genetics , DNA Mutational Analysis , Exons/genetics , Family , Female , Humans , Male , Microfilament Proteins , Pedigree , Phenotype
17.
Nephrol Dial Transplant ; 29 Suppl 4: iv87-94, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25165189

ABSTRACT

Cystinosis is caused by mutations in the CTNS gene (17p13.2), which encodes for a lysosomal cystine/proton symporter termed cystinosin. It is the most common cause of inherited renal Fanconi syndrome in young children. Because of its rarity, the diagnosis and specific treatment of cystinosis are frequently delayed, which has a significant impact on the overall prognosis. In this document, we have summarized expert opinions on several aspects of the disease to improve knowledge and provide guidance for diagnosis and treatment.


Subject(s)
Cystinosis/diagnosis , Cystinosis/therapy , Child , Cystinosis/genetics , Fanconi Syndrome/diagnosis , Fanconi Syndrome/therapy , Humans , Practice Guidelines as Topic , Societies, Medical
18.
Pediatr Nephrol ; 29(11): 2173-80, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24902943

ABSTRACT

BACKGROUND: Nephrotic syndrome (NS) presenting early in life is caused by heterogeneous glomerular diseases. We retrospectively evaluated whether histological diagnosis in children presenting with NS in the first year of life predicts remission or progression to end-stage kidney disease (ESKD). METHODS: This is a single centre retrospective review of all children diagnosed with NS before one year of age between 1990 and 2009. All subjects had a renal biopsy, which was independently blindly reviewed by a single renal pathologist for the purpose of this study. RESULTS: Forty-nine children (25 female) who presented at 0.1-11.6 (median 1.6) months were included with 31 presenting within the first three months of life. Histopathological review diagnostic categories were; 13 Mesangial proliferative glomerulopathy (MesGN), 12 Focal and segmental glomerulosclerosis (FSGS), 11 Finnish type changes, eight Diffuse Mesangial Sclerosis (DMS), three Minimal change disease (MCD) and one each of Dense Deposit Disease (DDD) and Membranous nephropathy. Two children died from haemorrhagic complications of the biopsy. Eight children achieved remission (four MesGN, one Finnish type changes, one FSGS, one MCD and one membranous) with patient and renal survival of 73 % and 43 %, respectively, at follow-up duration of 5-222 (median 73) months (with five lost to follow-up). All children with Finnish-type histopathological changes presented within five months of age. Due to the historical nature of the cohort, genetic testing was only available for 14 children, nine of whom had an identifiable genetic basis (seven NPHS1, one PLCE1 and one ITGA3) with none of these nine children achieving remission. All of them had presented within four months of age and required renal replacement therapy, and two died. CONCLUSIONS: Histopathological findings are varied in children presenting with NS early in life. Whilst groups of histological patterns of disease are associated with differing outcomes, accurate prediction of disease course in a specific case is difficult and more widespread genetic testing may improve the understanding of this group of diseases and their optimal management.


Subject(s)
Nephrotic Syndrome/congenital , Nephrotic Syndrome/physiopathology , Adolescent , Age of Onset , Biopsy/adverse effects , Child , Child, Preschool , Disease Progression , Female , Follow-Up Studies , Humans , Immunosuppressive Agents/therapeutic use , Infant , Infant, Newborn , Kidney/pathology , Kidney Failure, Chronic/etiology , Kidney Failure, Chronic/pathology , Male , Mutation/genetics , Nephrectomy , Nephrotic Syndrome/genetics , Retrospective Studies , Treatment Outcome
19.
Skeletal Radiol ; 2013 Sep 28.
Article in English | MEDLINE | ID: mdl-24077756

ABSTRACT

Nephropathic cystinosis is an autosomal recessive lysosomal storage disorder in which intracellular cystine accumulates. It is caused by mutations in the CTNS gene. Clinical manifestations include renal tubular Fanconi syndrome in the first year of life, rickets, hypokalaemia, polyuria, dehydration and acidosis, growth retardation, hypothyroidism, photophobia and renal glomerular deterioration. Late complications include myopathy, pancreatic insufficiency and retinal blindness. Skeletal manifestations described in these patients include failure to thrive, osteomalacia, rickets and short stature. This paper describes progressive bony abnormalities in three unrelated patients with nephropathic cystinosis that have not been reported previously.

SELECTION OF CITATIONS
SEARCH DETAIL