Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 66
3.
J Clin Invest ; 130(1): 335-344, 2020 01 02.
Article En | MEDLINE | ID: mdl-31613795

BACKGROUNDProteinuria is considered an unfavorable clinical condition that accelerates renal and cardiovascular disease. However, it is not clear whether all forms of proteinuria are damaging. Mutations in CUBN cause Imerslund-Gräsbeck syndrome (IGS), which is characterized by intestinal malabsorption of vitamin B12 and in some cases proteinuria. CUBN encodes for cubilin, an intestinal and proximal tubular uptake receptor containing 27 CUB domains for ligand binding.METHODSWe used next-generation sequencing for renal disease genes to genotype cohorts of patients with suspected hereditary renal disease and chronic proteinuria. CUBN variants were analyzed using bioinformatics, structural modeling, and epidemiological methods.RESULTSWe identified 39 patients, in whom biallelic pathogenic variants in the CUBN gene were associated with chronic isolated proteinuria and early childhood onset. Since the proteinuria in these patients had a high proportion of albuminuria, glomerular diseases such as steroid-resistant nephrotic syndrome or Alport syndrome were often the primary clinical diagnosis, motivating renal biopsies and the use of proteinuria-lowering treatments. However, renal function was normal in all cases. By contrast, we did not found any biallelic CUBN variants in proteinuric patients with reduced renal function or focal segmental glomerulosclerosis. Unlike the more N-terminal IGS mutations, 37 of the 41 proteinuria-associated CUBN variants led to modifications or truncations after the vitamin B12-binding domain. Finally, we show that 4 C-terminal CUBN variants are associated with albuminuria and slightly increased GFR in meta-analyses of large population-based cohorts.CONCLUSIONCollectively, our data suggest an important role for the C-terminal half of cubilin in renal albumin reabsorption. Albuminuria due to reduced cubilin function could be an unexpectedly common benign condition in humans that may not require any proteinuria-lowering treatment or renal biopsy.FUNDINGATIP-Avenir program, Fondation Bettencourt-Schueller (Liliane Bettencourt Chair of Developmental Biology), Agence Nationale de la Recherche (ANR) Investissements d'avenir program (ANR-10-IAHU-01) and NEPHROFLY (ANR-14-ACHN-0013, to MS), Steno Collaborative Grant 2018 (NNF18OC0052457, to TSA and MS), Heisenberg Professorship of the German Research Foundation (KO 3598/5-1, to AK), Deutsche Forschungsgemeinschaft (DFG) Collaborative Research Centre (SFB) KIDGEM 1140 (project 246781735, to CB), and Federal Ministry of Education and Research (BMB) (01GM1515C, to CB).


Albuminuria , Anemia, Megaloblastic , Kidney Tubules, Proximal , Malabsorption Syndromes , Mutation , Proteinuria , Receptors, Cell Surface , Vitamin B 12 Deficiency , Albuminuria/epidemiology , Albuminuria/genetics , Albuminuria/metabolism , Albuminuria/pathology , Anemia, Megaloblastic/epidemiology , Anemia, Megaloblastic/genetics , Anemia, Megaloblastic/metabolism , Anemia, Megaloblastic/pathology , Female , Humans , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Malabsorption Syndromes/epidemiology , Malabsorption Syndromes/genetics , Malabsorption Syndromes/metabolism , Malabsorption Syndromes/pathology , Male , Proteinuria/epidemiology , Proteinuria/genetics , Proteinuria/metabolism , Proteinuria/pathology , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Vitamin B 12 Deficiency/epidemiology , Vitamin B 12 Deficiency/genetics , Vitamin B 12 Deficiency/metabolism , Vitamin B 12 Deficiency/pathology
4.
Front Pediatr ; 7: 517, 2019.
Article En | MEDLINE | ID: mdl-32039104

For the pediatric nephrologist, the over-the-counter status for non-steroidal anti-inflammatory drugs (NSAIDs) is surprising due to their possible harmful side effects. These can include acute renal failure due mainly to glomerular hypoperfusion which may lead to acute tubular necrosis; more rarely in children, medullary ischemic injury and cardiovascular diseases; acute or chronic interstitial nephritis which may cause chronic renal failure. All of them may create electrolyte abnormalities: hyponatremia, hyperkaliemia, renal tubular acidosis, fluid retention causing hypertension.

5.
Pediatr Nephrol ; 31(12): 2375-2378, 2016 12.
Article En | MEDLINE | ID: mdl-27744619

BACKGROUND: Rational options for the treatment of end-stage renal disease (ESRD) due to atypical hemolytic uremic syndrome (aHUS) in children are still open to discussion. In the case of human complement factor H (CFH) deficiency, the choice is either kidney transplantation in combination with eculizumab, a humanized anti-C5 monoclonal antibody, or a combined liver-kidney transplantation. CASE-DIAGNOSIS/TREATMENT: A child with a homozygous CFH deficiency underwent a successful liver-kidney transplantation. CFH levels normalized within days. After 6 years of follow-up, the graft function (Cockroft clearance 100 ml min-1 1.73 m-2) and the liver functions were normal. RESULTS AND CONCLUSIONS: The results of this long-term follow-up confirm that combined liver-kidney transplantation remains a reasonable option in patients with ESRD due to aHUS when an identified genetic abnormality of the C3 convertase regulator synthesized in the liver has been identified.


Atypical Hemolytic Uremic Syndrome/genetics , Atypical Hemolytic Uremic Syndrome/surgery , Complement Factor H/deficiency , Kidney Diseases/genetics , Kidney Transplantation/methods , Liver Transplantation/methods , Atypical Hemolytic Uremic Syndrome/etiology , Complement Factor H/genetics , Consanguinity , Hereditary Complement Deficiency Diseases , Humans , Infant , Kidney Diseases/complications , Kidney Failure, Chronic/etiology , Kidney Failure, Chronic/surgery , Male , Plasma Exchange , Treatment Outcome
6.
Rev Prat ; 66(7): 769-772, 2016 Sep.
Article Fr | MEDLINE | ID: mdl-30512300

Urinary tract infection in children. The care of a child suspected of urinary infection (UI) goes through 4 stages. The diagnosis of UI is based on the presence of bacteriuria > 105 / mL, the collection of urine through a bas is unreliable and source of false positives. Localization diagnosis (acute pyelonephritis or cystitis) is based on clinical and biological signs (leukocytosis and inflammation). Etiological diagnosis is based on renal ultrasonography looking for obstructive uropathy, stones and bladder dysfunction; the search for a vesico-ureteral reflux with retrograd cystography is not systematic at first infection. The treatment of acute pyelonephritis is initiated usually intravenously for a period of 3 or 4 days. Oral antibiotics is then possible.


Infections urinaires de l'enfant. La prise en charge d'un enfant suspect d'infection urinaire passe par quatre étapes. Le diagnostic positif d'infection urinaire est posé en présence d'une bactériurie supérieure à 105 germes/mL ; le recueil des urines sur poche est peu fiable et source de faux positifs. Le diagnostic de localisation (pyélonéphrite aiguë ou cystite) repose sur des signes cliniques et biologiques (hyperleucocytose et syndrome inflammatoire). Le diagnostic causal repose sur l'interrogatoire et l'échographie rénale à la recherche d'une uropathie obstructive, d'une lithiase et d'un dysfonctionnement vésical. La recherche d'un reflux vésico-urétéral par cystographie rétrograde dès la première pyélonéphrite aiguë n'est pas systématique. Le traitement de la pyélonéphrite aiguë est initié le plus souvent par voie intraveineuse pour une durée de 2 à 3 jours ; le relais est ensuite pris par un traitement oral.


Urinary Tract Infections , Vesico-Ureteral Reflux , Bacteriuria/etiology , Child , Cystitis , Humans , Infant , Pyelonephritis/diagnosis , Urinary Tract Infections/complications , Urinary Tract Infections/diagnosis , Urinary Tract Infections/drug therapy , Vesico-Ureteral Reflux/diagnosis
7.
Acta Paediatr ; 104(10): e460-5, 2015 Oct.
Article En | MEDLINE | ID: mdl-26099938

AIM: Ultrasound and biological tools are used to predict high-grade vesicoureteral reflux, but other markers are needed to better select patients who need voiding cystography. Our aim was to determine whether studying Escherichia coli virulence factors would help to predict vesicoureteral reflux in patients with their first acute pyelonephritis. METHODS: We included children presenting with E. coli-related acute pyelonephritis or cystitis. Vesicoureteral reflux was assessed by voiding cystography. Virulence factors were identified by multiplex polymerase chain reaction. Statistical analysis was performed using logistic regression and the mean c-statistic test. RESULTS: We included 198 patients: 30 with cystitis and 168 with acute pyelonephritis, including 46 with vesicoureteral reflux. High-grade reflux was associated with acute pyelonephritis caused by the E. coli lacking virulence factors papGII (82% versus 47%, p < 0.001) or papC (85% versus 53%, p < 0.001) or belonging to phylogenetic group A or B1. When we added genetic data (lack of papGII, fyuA and phylogenetic groups) to classical predictors of vesicoureteral reflux (ultrasound examination, gender, age), the ability to predict high-grade reflux increased, with the c-statistic rising from 0.88 to 0.93. CONCLUSION: Bacterial virulence factors and clinical factors helped to predict high-grade reflux and may help to avoid unnecessary voiding cystographies.


Bacteriuria/complications , Escherichia coli/pathogenicity , Vesico-Ureteral Reflux/microbiology , Virulence Factors/genetics , Adhesins, Bacterial/genetics , Bacterial Toxins/genetics , Bacteriuria/microbiology , Escherichia coli/genetics , Female , Humans , Infant , Male , Retrospective Studies
8.
Liver Transpl ; 20(12): 1475-85, 2014 Dec.
Article En | MEDLINE | ID: mdl-25267365

Primary hyperoxaluria type 1 (PH1) is a hepatic metabolic defect leading to end-stage renal failure. The posttransplant recurrence of kidney disease can suggest a need for combined liver-kidney transplantation (LKT). However, the risk of LKT is theoretically far higher than the risk of kidney-alone transplantation (KAT). An unselected consecutive series of 54 patients with PH1 was analyzed according to the type of transplantation initially performed between May 1979 and June 2010 at 10 French centers. The duration of dialysis, extrarenal lesions, age, and follow-up were similar between the groups. Postoperative morbidity and mortality did not differ between the groups, and 10-year patient survival rates were similar for the LKT (n = 33) and KAT groups (n = 21; 78% versus 70%). Kidney graft survival at 10 years was better after LKT (87% versus 13%, P < .001) . Four patients (12.1%) lost their first kidney graft in the LKT group, whereas 19 (90%) did in the KAT group (P < .001). The recurrence of oxalosis occurred in 11 renal grafts (52%) in the KAT group but in none in the LKT group (P < .001). End-stage renal failure resulting from rejection was also higher in the KAT group (19% versus 9%, P < 0.0001). A second kidney transplant was performed for 15 patients (71%) in the KAT group versus 4 patients (12%) in the LKT group (P < 0.001). In conclusion, LKT for PH1 provides better kidney graft survival, less rejection, and similar long-term patient survival and is not associated with an increased short-term mortality risk. LKT must be the first-line treatment for PH1 patients with end-stage renal disease.


Hyperoxaluria, Primary/surgery , Kidney Transplantation , Liver Transplantation , Adolescent , Adult , Child , Child, Preschool , Female , Follow-Up Studies , France , Graft Survival , Humans , Hyperoxaluria/complications , Hyperoxaluria/surgery , Hyperoxaluria, Primary/mortality , Immunosuppressive Agents/therapeutic use , Infant , Kidney Failure, Chronic/surgery , Male , Middle Aged , Reoperation , Treatment Outcome , Young Adult
9.
Nephrol Dial Transplant ; 29 Suppl 4: iv113-6, 2014 Sep.
Article En | MEDLINE | ID: mdl-25165176

Fanconi-Bickel syndrome is a rare autosomal-recessive disorder caused by mutations in the SLC2A2 gene coding for the glucose transporter protein 2 (GLUT2). Major manifestations include hepatomegaly, glucose intolerance, post-prandial hypoglycaemia and renal disease that usually presents as proximal tubular acidosis associated with proximal tubule dysfunction (renal Fanconi syndrome). We report a patient harbouring a homozygous mutation of SLC2A2 who presented a dramatic exacerbation of metabolic acidosis in the context of a viral infection, owing to both ketosis and major urinary bicarbonate loss. The kidney biopsy revealed nuclear and cytoplasmic accumulation of glycogen in proximal tubule cells, a lack of expression of GLUT2, and major defects of key proteins of the proximal tubule such as megalin, cubilin and the B2 subunit of H(+)-ATPase. These profound alterations of the transport systems most likely contributed to proximal tubule alterations and profound bicarbonate loss.


Acidosis/physiopathology , Fanconi Syndrome/physiopathology , Glucose Transporter Type 2/genetics , Kidney Tubules, Proximal/physiopathology , Mutation/genetics , Acidosis/genetics , Acute Disease , Fanconi Syndrome/genetics , Glucose Transporter Type 2/deficiency , Homozygote , Humans , Infant , Male
10.
Article En | MEDLINE | ID: mdl-24940675

Adenine phosphoribosyltransferase (APRT) deficiency is a rare inherited metabolic disorder that leads to the formation and hyperexcretion of 2,8-dihydroxyadenine (DHA) into urine. The low solubility of DHA results in precipitation and formation of urinary crystals and kidney stones. The disease can be present as recurrent urolithiasis or nephropathy secondary to crystal precipitation into renal parenchyma (DHA nephropathy). The diagnostic tools available, including stone analysis, crystalluria, and APRT activity in red blood cells, make the diagnosis easy to confirm when APRT deficiency is suspected. However, the lack of recognition of this metabolic disorder frequently resulted in a delay in diagnosis and treatment with grave consequences. The early recognition and treatment of APRT deficiency are of crucial importance to prevent irreversible loss of renal function. This review summarizes the genetic and metabolic mechanisms underlying DHA stones formation and chronic kidney disease, along with the issues of diagnosis and management of APRT deficiency. Moreover, we report the mutations in the APRT gene responsible for APRT deficiency in 51 French patients (43 families) including 22 pediatric cases (18 families) among the 64 patients identified in the biochemistry laboratories of Necker Hospital, Paris (1978-2013).


Adenine Phosphoribosyltransferase/deficiency , Metabolism, Inborn Errors , Urolithiasis , Adenine Phosphoribosyltransferase/metabolism , Humans , Metabolism, Inborn Errors/diagnosis , Metabolism, Inborn Errors/epidemiology , Metabolism, Inborn Errors/metabolism , Metabolism, Inborn Errors/therapy , Prevalence , Urolithiasis/diagnosis , Urolithiasis/epidemiology , Urolithiasis/metabolism , Urolithiasis/therapy
12.
Clin J Am Soc Nephrol ; 7(9): 1521-7, 2012 Sep.
Article En | MEDLINE | ID: mdl-22700886

Complete adenine phosphoribosyltransferase (APRT) deficiency is a rare inherited metabolic disorder that leads to the formation and hyperexcretion of 2,8-dihydroxyadenine (DHA) into urine. The low solubility of DHA results in precipitation of this compound and the formation of urinary crystals and stones. The disease can present as recurrent urolithiasis or nephropathy secondary to crystal precipitation into renal parenchyma (DHA nephropathy). The diagnostic tools available-including stone analysis, crystalluria, and APRT activity measurement-make the diagnosis easy to confirm when APRT deficiency is suspected. However, the disease can present at any age, and the variability of symptoms can present a diagnostic challenge to many physicians. The early recognition and treatment of APRT deficiency are of crucial importance for preventing irreversible loss of renal function, which still occurs in a non-negligible proportion of cases. This review summarizes the genetic and metabolic mechanisms underlying stone formation and renal disease, along with the diagnosis and management of APRT deficiency.


Adenine Phosphoribosyltransferase/deficiency , Metabolism, Inborn Errors/enzymology , Urolithiasis/enzymology , Adenine/analogs & derivatives , Adenine/urine , Adenine Phosphoribosyltransferase/genetics , Adenine Phosphoribosyltransferase/urine , Allopurinol/therapeutic use , Animals , Biomarkers/urine , Disease Progression , Enzyme Inhibitors/therapeutic use , Genetic Predisposition to Disease , Humans , Kidney Diseases/enzymology , Kidney Diseases/etiology , Metabolism, Inborn Errors/complications , Metabolism, Inborn Errors/diagnosis , Metabolism, Inborn Errors/drug therapy , Metabolism, Inborn Errors/genetics , Metabolism, Inborn Errors/urine , Phenotype , Predictive Value of Tests , Prognosis , Recurrence , Urolithiasis/complications , Urolithiasis/diagnosis , Urolithiasis/drug therapy , Urolithiasis/etiology , Urolithiasis/genetics , Urolithiasis/urine , Xanthine Dehydrogenase/antagonists & inhibitors , Xanthine Dehydrogenase/metabolism
13.
Pediatr Nephrol ; 27(4): 571-9, 2012 Apr.
Article En | MEDLINE | ID: mdl-22212387

Adenine phosphoribosyltransferase (APRT) deficiency is a rare autosomal recessive disorder characterized by 2,8-dihydroxyadenine (2,8-DHA) crystalluria that can cause nephrolithiasis and chronic kidney disease. The aim of our study was to assess the clinical presentation, diagnosis, and outcome of APRT deficiency in a large pediatric cohort. All pediatric cases of APRT deficiency confirmed at the same French reference laboratories between 1978 and 2010 were retrospectively reviewed. Twenty-one patients from 18 families were identified. The median age at diagnosis was 3 years. Diagnosis was made after one or more episodes of nephrolithiasis (17 patients), after urinary tract infection (1 patient), and by family screening (3 patients). The diagnosis was based on stone analysis and microscopic examination of urine and/or enzymatic determination of APRT on red blood cells. All children had null APRT enzyme activity in erythrocytes. APRT gene sequencing was performed on 18 patients, revealing six homozygous and 12 compound heterozygous mutations. At diagnosis, half of the patients had decreased kidney function, and two children presented with acute renal failure. Allopurinol treatment was given to all patients at a median dose of 9 mg/kg/day. After a median follow-up of 5 years, all patients showed stabilization or improvement of kidney function, normal growth and development, and six patients had recurrence of nephrolithiasis. Based on these results, we conclude that an excellent outcome can be achieved in children with APRT deficiency who receive the proper treatment.


Metabolism, Inborn Errors/drug therapy , Urolithiasis/drug therapy , Adenine Phosphoribosyltransferase/deficiency , Adenine Phosphoribosyltransferase/genetics , Adolescent , Allopurinol/therapeutic use , Antimetabolites/therapeutic use , Child , Child, Preschool , Female , Humans , Infant , Male , Metabolism, Inborn Errors/diagnosis , Metabolism, Inborn Errors/genetics , Retrospective Studies , Urolithiasis/diagnosis , Urolithiasis/genetics , Young Adult
14.
Pediatr Diabetes ; 13(6): e35-9, 2012 Sep.
Article En | MEDLINE | ID: mdl-22260488

Heterozygous mutations of TCF2 (transcription factor 2) have been associated with maturity onset diabetes of the young, renal malformations, hyperuricemia, and occasionally internal genital malformations in female. We report a female patient with bilateral renal hypodysplasia and de novo heterozygous TCF2 gene mutation. At the age of 9 yr, she developed transient ketoacidosis immediately posttransplant, temporarily requiring insulin. During glucocorticoid tapering, impaired glucose tolerance persisted and overt insulin-dependent diabetes mellitus developed 1 yr later. Pathogenic factors which might have played a role in the acceleration of diabetes were (i) switch from cyclosporine to tacrolimus, (ii) weight excess, and (iii) cytomegalovirus infection. TCF2 analysis might, therefore, be of interest in patients with congenital abnormalities of the kidney and the urinary tract in order to improve posttransplant management in terms of steroid and tacrolimus exposure.


Abnormalities, Multiple/genetics , Diabetes Mellitus, Type 2/etiology , Hepatocyte Nuclear Factor 1-beta/genetics , Kidney Transplantation/adverse effects , Mutation , Abnormalities, Multiple/surgery , Child , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/diagnosis , Diabetes Mellitus, Type 2/genetics , Female , Humans , Kidney/abnormalities , Kidney/surgery , Mutation/physiology , Transplantation Conditioning/adverse effects , Urinary Tract/abnormalities , Urinary Tract/surgery , Urogenital Abnormalities/complications , Urogenital Abnormalities/genetics , Urogenital Abnormalities/surgery
15.
Pediatr Nephrol ; 27(3): 389-96, 2012 Mar.
Article En | MEDLINE | ID: mdl-21947272

Mycophenolate mofetil (MMF) has emerged as a new therapeutic option in steroid-dependent nephrotic syndrome (SDNS). We conducted a phase II Bayesian trial of MMF in children with SDNS. Phase II trials, usually single-arm studies, investigate the effect of new treatments. Standard Fleming's procedure relies on observed results (relapse rate during the trial), whereas Bayesian approach combines observed results with prior information (expected relapse rate according to prior studies and clinical experience). All patients were required to have received prior alkylating-agent treatment. Sixty-seven percent of them had also received levamisole. Patients received MMF (1,200 mg/m(2)/day) and prednisone according to a defined schedule [reduction of alternate-day (e.o.d) dose to 50% of pre-MMF dose at 3 months, 25% at 6 months]. Twenty-four children (median age 6.0 years, 2.8-14.4) entered the study and 23 completed it. Bayesian analysis showed that adding four patients would not change significance of results, allowing stopping inclusions. Four patients relapsed during the first 6 months (estimated probability 17.6%, 95% credibility interval: 5.4-35.0%) and two at months 8 and 11.5. In the 19 patients free of relapse during the first 6 months, median (Q1-Q3) prednisone maintenance dose decreased from 25 (10-44) to 9 (7.5-11.2) mg/m(2) e.o.d (p < 0.001) and cumulative dose from 459 (382-689) to 264 (196-306) mg/m(2)/month (p < 0.001) before and on MMF respectively. Pre-MMF patient characteristics and MMF pharmacokinetics did not differ between patients with or without relapse. MMF reduces relapse rate and steroid dose in children with SDNS and should be proposed before cyclosporine and cyclophosphamide.


Immunosuppressive Agents/therapeutic use , Mycophenolic Acid/analogs & derivatives , Nephrotic Syndrome/drug therapy , Adolescent , Area Under Curve , Bayes Theorem , Child , Child, Preschool , Female , Humans , Male , Mycophenolic Acid/adverse effects , Mycophenolic Acid/pharmacokinetics , Mycophenolic Acid/therapeutic use , Prednisone/therapeutic use , Prospective Studies
16.
Pediatr Nephrol ; 27(7): 1051-7, 2012 Jul.
Article En | MEDLINE | ID: mdl-21638156

Acute tubulointerstitial nephritis (TIN) is a frequent cause of acute renal failure, characterised by the presence of inflammatory cell infiltrate in the interstitium of the kidney. Immuno-allergic reaction to certain medications, mainly non-steroidal anti-inflammatory drugs and antibiotics are by far the most important etiology for TIN today, but other situations such as infections, toxins, and vasculitis are known to induce TIN. Incidence of TIN is increasing, probably due to prescription habits and NSAID overuse, representing 3-7% of acute kidney injury in biopsies in children. Avoidance of the causal substance and rapid steroid therapy are hallmarks for patient care, but spontaneous initial recovery is very frequent and the general prognosis seems satisfactory. However, development of chronic TIN, without response to steroid or other immunosuppressive treatment, is possible. As the largest part of TIN is secondary to certain drugs, clear indications in particular for NSAID or antibiotics should be respected to reduce the number of TIN cases.


Nephritis, Interstitial/diagnosis , Nephritis, Interstitial/therapy , Acute Disease , Humans , Nephritis, Interstitial/etiology
17.
Hum Pathol ; 42(8): 1142-8, 2011 Aug.
Article En | MEDLINE | ID: mdl-21315411

The transcription factor Snail is an important repressor of E-cadherin gene expression. It plays a key role in the induction of epithelial-mesenchymal transition, an essential process important not only in embryonic development and tumor progression but also in organ fibrogenesis. We studied the expression of Snail by immunohistochemistry, along with several epithelial phenotypic changes suggestive of epithelial-mesenchymal transition, in 14 patients with multiple myeloma cast nephropathy. This nephropathy is characterized by a rapid progression toward fibrosis. As controls, we used normal kidneys and kidneys from patients displaying an idiopathic nephrotic syndrome, a syndrome unassociated with renal fibrosis. We discovered that, in all patients with multiple myeloma nephropathy, a drastic accumulation of Snail is seen in the nuclei from tubular epithelial cells showing epithelial phenotypic changes. In contrast, normal and idiopathic nephrotic syndrome kidneys did not exhibit either of these markers. Snail, a major player in the process of epithelial-to-mesenchymal transition, is highly expressed by tubular epithelial cells during multiple myeloma nephropathy. It is, therefore, a potential target to prevent multiple myeloma kidneys from fibrosing. Intranuclear accumulation of Snail is a characteristic in phenotypically altered tubular cells from multiple myeloma kidneys. The epithelial-mesenchymal transition pathway could, therefore, be involved in the rapid renal fibrogenesis observed in this setting.


Acute Kidney Injury/pathology , Cell Nucleus/metabolism , Epithelial Cells/pathology , Kidney Tubules/metabolism , Multiple Myeloma/pathology , Transcription Factors/metabolism , Acute Kidney Injury/metabolism , Biomarkers/metabolism , Cell Nucleus/pathology , Epithelial Cells/metabolism , Humans , Kidney/pathology , Kidney Tubules/pathology , Middle Aged , Multiple Myeloma/metabolism , Phenotype , Snail Family Transcription Factors
18.
PLoS One ; 6(12): e29556, 2011.
Article En | MEDLINE | ID: mdl-22216314

BACKGROUND: Predicting vesico-ureteral reflux (VUR) ≥3 at the time of the first urinary tract infection (UTI) would make it possible to restrict cystography to high-risk children. We previously derived the following clinical decision rule for that purpose: cystography should be performed in cases with ureteral dilation and a serum procalcitonin level ≥0.17 ng/mL, or without ureteral dilatation when the serum procalcitonin level ≥0.63 ng/mL. The rule yielded a 86% sensitivity with a 46% specificity. We aimed to test its reproducibility. STUDY DESIGN: A secondary analysis of prospective series of children with a first UTI. The rule was applied, and predictive ability was calculated. RESULTS: The study included 413 patients (157 boys, VUR ≥3 in 11%) from eight centers in five countries. The rule offered a 46% specificity (95% CI, 41-52), not different from the one in the derivation study. However, the sensitivity significantly decreased to 64% (95%CI, 50-76), leading to a difference of 20% (95%CI, 17-36). In all, 16 (34%) patients among the 47 with VUR ≥3 were misdiagnosed by the rule. This lack of reproducibility might result primarily from a difference between derivation and validation populations regarding inflammatory parameters (CRP, PCT); the validation set samples may have been collected earlier than for the derivation one. CONCLUSIONS: The rule built to predict VUR ≥3 had a stable specificity (ie. 46%), but a decreased sensitivity (ie. 64%) because of the time variability of PCT measurement. Some refinement may be warranted.


Calcitonin/blood , Protein Precursors/blood , Urinary Tract Infections/complications , Vesico-Ureteral Reflux/complications , Calcitonin Gene-Related Peptide , Child , Decision Making , Female , Humans , Male , Prospective Studies , Sensitivity and Specificity , Urinary Tract Infections/blood , Vesico-Ureteral Reflux/blood , Vesico-Ureteral Reflux/diagnosis
19.
Radiology ; 255(3): 890-8, 2010 Jun.
Article En | MEDLINE | ID: mdl-20501726

PURPOSE: To directly compare various renal ultrasonography (US) criteria for vesicoureteral reflux (VUR) with voiding cystography, the reference method, for diagnostic accuracy in helping to determine an intermediate strategy of screening children who require cystography. MATERIALS AND METHODS: Institutional review board approval and parental consent were obtained for this prospective hospital-based cohort study involving children with urinary tract infections (UTIs). Renal length, ureteral dilatation, pelvic dilatation, and corticomedullary differentiation were analyzed and compared. One hundred seventeen patients (median age, 0.8 year; age range, 0.0-13.9 years) were included: 46 (39%) boys (median age, 0.3 year; age range, 0.5-13.9 years) and 71 girls (median age, 1.2 years; age range, 0.0-11.5 years). A two-level logistic regression model was used to analyze data, and diagnostic accuracy calculations were performed. RESULTS: Thirty-two (27%) children had all-grade VUR, and eight (7%) had VUR of grade 3 or higher. Only ureteral dilatation was significantly related to all-grade VUR (odds ratio [OR], 7.5; 95% confidence interval [CI]: 1.0, 58.2; P = .05), with 25% sensitivity (95% CI: 15%, 39%) and 88% specificity (95% CI: 83%, 92%). Ureteral, pelvic, and urinary tract dilatations were significantly associated with VUR of grade 3 or higher, with ORs of 20.2 (95% CI: 3.5, 118.2; P = .001), 13.7 (95% CI: 4.1, 46.0; P < .001), and 20.0 (95% CI: 4.4, 90.1; P < .001), respectively. The best compromise between sensitivity and specificity was achieved by using the ureteral dilatation criterion, which had 73% sensitivity (95% CI: 43%, 90%) and 88% specificity (95% CI: 84%, 92%) for high-grade VUR. CONCLUSION: Ureteral dilatation may yield the best accuracy for the US-based diagnosis of both all-grade and high-grade VUR. This US criterion, perhaps in combination with other predictors, might find a place in an evidence-based selective strategy for limiting cystography in children with UTIs.


Urinary Tract Infections/diagnostic imaging , Vesico-Ureteral Reflux/diagnostic imaging , Adolescent , Bayes Theorem , Chi-Square Distribution , Child , Child, Preschool , Female , Humans , Infant , Infant, Newborn , Logistic Models , Male , Prospective Studies , Sensitivity and Specificity , Ultrasonography , Urinary Tract Infections/etiology , Vesico-Ureteral Reflux/complications
20.
Br J Clin Pharmacol ; 69(4): 358-66, 2010 Apr.
Article En | MEDLINE | ID: mdl-20406220

AIMS: To develop a population pharmacokinetic model for mycophenolic acid (MPA) in children with idiopathic nephrotic syndrome (INS) treated with mycophenolate mofetil (MMF), identify covariates that explain variability and determine the Bayesian estimator of the area under the concentration-time curve over 12 h (AUC(0-12)). METHODS: The pharmacokinetic model of MMF was described from 23 patients aged 7.4 +/- 3.9 years (range 2.9-14.9) using nonlinear mixed-effects modelling (NONMEM) software. A two-compartment model with lag-time and first-order absorption and elimination was developed. The final model was validated using visual predictive check. Bayesian estimator was validated using circular permutation method. RESULTS: The population pharmacokinetic parameters were apparent oral clearance 9.7 l h(-1), apparent central volume of distribution 22.3 l, apparent peripheral volume of distribution 250 l, inter-compartment clearance 18.8 l h(-1), absorption rate constant 5.16 h(-1), lag time 0.215 h. The covariate analysis identified body weight and serum albumin as individual factors influencing the apparent oral clearance. Accurate Bayesian estimation of AUC(0-12) was obtained using the combination of three MPA concentrations measured just before (T(0)), 1 and 4 h (T(1) and T(4)) after drug intake with a small error of 0.298 microg h(-1) ml(-1) between estimated and reference AUC(0-12). CONCLUSIONS: The population pharmacokinetic model of MPA was developed in children with INS. A three-point (T(0), T(1) and T(4)h) Bayesian estimator of AUC(0-12) was developed and might be used to investigate the relation between MPA pharmacokinetic and pharmacodynamics in children with INS and determine if there is any indication to monitor MPA exposure in order to improve patient outcome based on individual AUC-controlled MMF dosing.


Enzyme Inhibitors/pharmacokinetics , Immunosuppressive Agents/pharmacokinetics , Mycophenolic Acid/analogs & derivatives , Mycophenolic Acid/pharmacokinetics , Nephrotic Syndrome/drug therapy , Administration, Oral , Adolescent , Area Under Curve , Bayes Theorem , Child , Child, Preschool , Chromatography, High Pressure Liquid , Enzyme Inhibitors/blood , Female , Humans , Immunosuppressive Agents/therapeutic use , Male , Models, Biological , Mycophenolic Acid/blood , Mycophenolic Acid/therapeutic use , Prospective Studies
...