Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 39
1.
J Endocrinol Invest ; 45(1): 159-166, 2022 Jan.
Article En | MEDLINE | ID: mdl-34216372

PURPOSE: Denosumab is a fully human monoclonal anti-RANK-L antibody that is clinically used to counteract the bone loss induced by exacerbated osteoclast activity. Indeed, its binding to RANK-L prevents the interaction RANK-L/receptor RANK that is essential for osteoclastogenesis and bone resorbing activity. Although there are many medications available to treat bone loss diseases, including bisphosphonates, Denosumab is highly effective since it reduces the bone erosion. The use in pediatric patients is safe. However, some concerns are related to the interruption of the treatment. Indeed, in this study, we reported hypercalcemia in two pediatric patients and alterations of circulating osteoclast precursors. METHODS: Peripheral Blood Mononuclear Cells (PBMC) were isolated from two pediatric patients with hypercalcemia after Denosumab interruption and from 10 controls. Cytofluorimetric analysis and in vitro osteoclastogenesis experiments were performed. RESULTS: Increase of CD16-CD14+CD11b+ cells was revealed in PBMC from patients reflecting the enhanced in vitro osteoclastogenesis. CONCLUSION: Our data suggest that precautions must be taken when Denosumab therapy is interrupted and gradual decrease of dose and/or timing of treatment should be performed. To prevent the onset of hypercalcemia that could be in the discontinuation phase, cytofluorimetric analysis of PBMC should be performed to evaluate osteoclast precursors.


Denosumab/therapeutic use , Hypercalcemia/etiology , Withholding Treatment , Acute Disease , Adolescent , Belgium , Bone Cysts, Aneurysmal/blood , Bone Cysts, Aneurysmal/drug therapy , Case-Control Studies , Cells, Cultured , Child , Granuloma, Giant Cell/blood , Granuloma, Giant Cell/drug therapy , Humans , Hypercalcemia/blood , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Male , Mandibular Diseases/blood , Mandibular Diseases/drug therapy , Osteoclasts/drug effects , Osteoclasts/physiology , Primary Cell Culture , Signal Transduction/drug effects
2.
Mol Genet Metab ; 126(4): 413-415, 2019 04.
Article En | MEDLINE | ID: mdl-30685240

INTRODUCTION: Nephropathic cystinosis is a rare autosomal recessive lysosomal storage disorder caused by mutations in the CTNS gene. Patients with nephropathic cystinosis suffer not only from renal disease but have also other systemic complications like myopathy and swallowing dysfunction. Dysphagia for solid food is mentioned in patients with cystinosis, but in clinical practice swallowing investigations are only performed when the patient has complaints. The aim of this study was to explore the swallowing function in patients with cystinosis by use of the Test of Mastication and Swallowing Solids (TOMASS), and to compare their performance with patients with myotonic dystrophy type 1 - a neuromuscular disease in which dysphagia for solid food is a known problem. METHODS: Twenty adult patients with cystinosis (11 men and 9 women, range 19-51 years) and 10 patients with myotonic dystrophy type 1 (5 men and 5 women, range 20-60 years) were included. All cystinosis patients were treated with cysteamine. Data of the two groups were compared with normative data using independent-samples t-tests. In case the variables were not normally distributed, the non-parametric Mann-Whitney U test was used. RESULTS: There was a significant difference in the number of bites, masticatory cycles, swallows and total time between the normal values and cystinosis patients. The results of the cystinosis patients were comparable to those of the patients with myotonic dystrophy. DISCUSSION AND CONCLUSION: Adult patients with cystinosis have significant dysphagia for solid food. Clinicians treating these patients should be aware of this fact. The TOMASS can be performed easily in clinical practice to investigate whether patients with cystinosis have swallowing dysfunction. The swallowing dysfunction can now be diagnosed by use of a non-invasive, very simple, non-harmful test. It can be discussed whether this should be added to the regular care scheme of cystinosis patients in order to regularly follow-up swallowing function.


Cystinosis/complications , Deglutition Disorders/etiology , Deglutition , Kidney Diseases/complications , Adult , Cysteamine/therapeutic use , Cystinosis/drug therapy , Female , Humans , Male , Mastication , Middle Aged , Myotonic Dystrophy/complications , Young Adult
3.
Sci Rep ; 6: 35395, 2016 10 13.
Article En | MEDLINE | ID: mdl-27734949

Cystinosis is a rare autosomal recessive disorder characterized by lysosomal cystine accumulation due to loss of function of the lysosomal cystine transporter (CTNS). The most common mutation in cystinosis patients of Northern Europe consists of a 57-kb deletion. This deletion not only inactivates the CTNS gene but also extends into the non-coding region upstream of the start codon of the TRPV1 gene, encoding the capsaicin- and heat-sensitive ion channel TRPV1. To evaluate the consequences of the 57-kb deletion on functional TRPV1 expression, we compared thermal, mechanical and chemical sensitivity of cystinosis patients with matched healthy controls. Whereas patients heterozygous for the 57-kb deletion showed normal sensory responses, homozygous subjects exhibited a 60% reduction in vasodilation and pain evoked by capsaicin, as well as an increase in heat detection threshold. Responses to cold, mechanical stimuli or cinnamaldehyde, an agonist of the related nociceptor channel TRPA1, were unaltered. We conclude that cystinosis patients homozygous for the 57-kb deletion exhibit a strong reduction of TRPV1 function, leading to sensory deficiencies akin to the phenotype of TRPV1-deficient mice. These deficits may account for the reported sensory alterations and thermoregulatory deficits in these patients, and provide a paradigm for life-long TRPV1 deficiency in humans.


Cystinosis/metabolism , Gene Deletion , Homozygote , TRPV Cation Channels/metabolism , Acrolein/analogs & derivatives , Acrolein/chemistry , Adolescent , Adult , Alleles , Capsaicin/chemistry , Codon , Cystinosis/genetics , Europe , Female , Hot Temperature , Humans , Lysosomes/metabolism , Male , Mutation , Sequence Deletion , TRPA1 Cation Channel/metabolism , TRPV Cation Channels/genetics , Young Adult
4.
J Thromb Haemost ; 13(5): 755-67, 2015 May.
Article En | MEDLINE | ID: mdl-25758343

BACKGROUND: Pituitary adenylate cyclase-activating polypeptide (PACAP) was recently identified as an inhibitor of megakaryopoiesis and platelet aggregability. OBJECTIVE: We studied PACAP levels in children with nephrotic syndrome (NS), which is associated with thrombocytosis, platelet hyperaggregability, and an increased risk of thrombosis. PATIENTS/METHODS: In four children with congenital NS (CNS) and 24 children with idiopathic NS (INS), plasma and urine levels of PACAP and ceruloplasmin were measured, as were platelet counts and platelet aggregation responses to collagen. In CNS patients, in vitro megakaryopoiesis and nuclear factor-κB expression in platelet lysates were also measured. All tests were performed during the nephrotic state and the non-nephrotic state. RESULTS: Urinary losses of PACAP and ceruloplasmin were observed during the nephrotic state, and disappeared during the non-nephrotic state. Plasma PACAP deficiency was more pronounced in CNS patients than in INS patients. Thrombocytosis was observed in all CNS patients and in 11 of 29 INS patients during the nephrotic state. During the PACAP-deficient state, in vitro megakaryopoiesis was increased for CNS patients, and this effect could be reversed by the addition of recombinant PACAP. Platelet hyperaggregability was observed during the nephrotic state in both CNS and INS patients. In INS patients, the addition of recombinant PACAP to patients' platelets was studied, and resulted in decreased aggregation during the nephrotic state. Platelet aggregation correlated inversely with plasma PACAP levels, but not with serum albumin levels. CONCLUSIONS: We demonstrate urinary losses of PACAP and plasma PACAP deficiency in children with NS, associated with thrombocytosis and platelet hyperaggregability.


Nephrotic Syndrome/blood , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Platelet Aggregation , Platelet Count , Adolescent , Child , Child, Preschool , Humans , Infant
5.
New Microbes New Infect ; 2(5): 138-43, 2014 Sep.
Article En | MEDLINE | ID: mdl-25356363

In 2011, a large outbreak of infections caused by Shiga toxin-producing Escherichia coli (STEC) O104:H4 occurred in Germany. This exceptionally virulent strain combined virulence factors of enteroaggregative E. coli (EAggEC) and STEC. After the outbreak only a few sporadic cases of infection with this rare serotype were reported, most of which were related to travel to the Middle East or North Africa. Here we describe two cases of enteroaggregative STEC (Agg-STEC) O104:H4 infection that occurred in Belgium in 2012 and 2013 respectively. In both cases travel in a Mediterranean country preceded the infection. The first strain was isolated from the stool of a 42-year-old woman presenting bloody diarrhoea, who had travelled to Tunisia the week before. The second case involves a 14-year-old girl who, upon her return from Turkey to Belgium, suffered from an episode of bloody diarrhoea and haemolytic uraemic syndrome. Extended typing of the isolates with pulsed field gel electrophoresis revealed that the strains were closely related, though not exactly the same as the 2011 outbreak strain. This report supports the previously made hypothesis that Agg-STEC has a human reservoir and might be imported by travellers coming from an area where the pathogen is endemic. Furthermore, it emphasizes the concern that these bacteria may cause future outbreaks as evenly virulent O104:H4 isolates seem to be widespread.

6.
JIMD Rep ; 16: 25-30, 2014.
Article En | MEDLINE | ID: mdl-24798266

BACKGROUND: Cystinosis is an autosomal recessive disorder marked by intralysosomal cystine accumulation. Patients present with generalized proximal tubular dysfunction called renal Fanconi syndrome. Urinary carnitine loss results in plasma and muscle carnitine deficiency, but no clinical signs of carnitine deficiency have been described. Also, the optimal dose of carnitine supplementation is undefined. This study aimed to determine whether currently recommended carnitine doses result in adequate correction of plasma carnitine. METHODS: Five cystinosis patients with renal Fanconi syndrome, aged 2-18 years, were included. L-carnitine was prescribed 50 mg/kg/day since diagnosis: median 36 (range 18-207) months. Total and free plasma and urine carnitine and carnitine profiles were measured at study onset, after stopping L-carnitine for 3 months and 3 months after reintroducing L-carnitine 50 mg/kg/day. RESULTS: At study onset, plasma free carnitine was normal in all patients, total carnitine (1/5), acetylcarnitine (3/5), and several short- and medium-chain acylcarnitines ≤10 carbons (5/5) were increased indicating carnitine over-supplementation. Three months after cessation, carnitine profiles normalized and 3/5 patients showed plasma carnitine deficiency. Three months after reintroduction, plasma free carnitine normalized in all patients, however, carnitine profiles were disturbed in 4/5 patients. Urine free carnitine, acetylcarnitine, and acylcarnitines ≤10 carbons were increased in all patients independent of carnitine supplementation. CONCLUSION: Administration of recommended doses L-carnitine (50 mg/kg/day) resulted in over-supplementation. Although the drug is considered to be rather safe, long-term effects of over-supplementation remain unknown warranting cautious use of high doses. Plasma carnitine profile might be used as a monitor, to prevent overdosing.

7.
J Inherit Metab Dis ; 36(6): 1073-7, 2013 Nov.
Article En | MEDLINE | ID: mdl-23371451

BACKGROUND: Cystinosis is an autosomal recessive disease caused by intralysosomal cystine accumulation, treated with cysteamine. Recently, new adverse effects of cysteamine were reported. Skin biopsies showed microvascular proliferation (angioendotheliomatosis). To examine the mechanism of angioendotheliomatosis associated with cysteamine toxicity, we examined the effect of cysteamine on human dermal microvascular endothelial cells (HDMVEC). METHODS: After cysteamine exposure (range 0-3.0 mM) during 24 h, cell viability was measured using water soluble tetrazolium salt-1 (WST-1) in both control HDMVEC and fibroblasts. Cell proliferation and apoptosis rate were measured in HDMVEC by bromodeoxyuridine (BrdU) incorporation and caspase 3 and caspase 7 activity, respectively. Intracellular glutathione (GSH) was measured in HDMVEC after cysteamine exposure of 0, 0.1 or 1.0 mM. Medium and cysteamine were refreshed every 6 h to mimic the in vivo situation. Next, cell viability in HDMVEC was measured after 24 h of GSH exposure (range 0-10.0 mM). RESULTS: HDMVEC viability and proliferation increased after cysteamine exposure 0.03-3.0 mM (p < 0.01) and 0.03-1.0 mM (p = 0.01) respectively; cell viability in fibroblasts was not affected by incubation with cysteamine. Apoptosis remained unaffected by incubation with 0-1.0 mM cysteamine, 3.0 mM caused increased apoptosis. Intracellular GSH was significantly increased after incubation with cysteamine 0.1 mM (p = 0.02) and 1.0 mM (p < 0.01). HDMVEC viability increased after exposure to GSH 1.0-5.0 mM (p < 0.01). CONCLUSION: Cysteamine concentrations, similar to those described in plasma of cystinosis patients, stimulate HDMVEC viability and proliferation and increase intracellular GSH content. We postulate that this mechanism might underlie angioendotheliomatosis induced by cysteamine.


Cysteamine/pharmacology , Dermis/cytology , Endothelial Cells/drug effects , Endothelial Cells/physiology , Apoptosis/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Dermis/blood supply , Dermis/metabolism , Dose-Response Relationship, Drug , Endothelial Cells/cytology , Glutathione/analysis , Glutathione/metabolism , Glutathione/pharmacology , Humans
9.
Cell Calcium ; 51(6): 452-8, 2012 Jun.
Article En | MEDLINE | ID: mdl-22456092

Autosomal dominant polycystic kidney disease is caused by loss-of-function mutations in the PKD1 or PKD2 genes encoding respectively polycystin-1 and polycystin-2. Polycystin-2 stimulates the inositol trisphosphate (IP(3)) receptor (IP(3)R), a Ca(2+)-release channel in the endoplasmic reticulum (ER). The effect of ER-located polycystin-1 is less clear. Polycystin-1 has been reported both to stimulate and to inhibit the IP(3)R. We now studied the effect of polycystin-1 and of polycystin-2 on the IP(3)R activity under conditions where the cytosolic Ca(2+) concentration was kept constant and the reuptake of released Ca(2+) was prevented. We also studied the interdependence of the interaction of polycystin-1 and polycystin-2 with the IP(3)R. The experiments were done in conditionally immortalized human proximal-tubule epithelial cells in which one or both polycystins were knocked down using lentiviral vectors containing miRNA-based short hairpins. The Ca(2+) release was induced in plasma membrane-permeabilized cells by various IP(3) concentrations at a fixed Ca(2+) concentration under unidirectional (45)Ca(2+)-efflux conditions. We now report that knock down of polycystin-1 or of polycystin-2 inhibited the IP(3)-induced Ca(2+) release. The simultaneous presence of the two polycystins was required to fully amplify the IP(3)-induced Ca(2+) release, since the presence of polycystin-1 alone or of polycystin-2 alone did not result in an increased Ca(2+) release. These novel findings indicate that ER-located polycystin-1 and polycystin-2 operate as a functional complex. They are compatible with the view that loss-of-function mutations in PKD1 and in PKD2 both cause autosomal dominant polycystic kidney disease.


Calcium Signaling , Calcium/metabolism , Inositol 1,4,5-Trisphosphate/pharmacology , TRPP Cation Channels/metabolism , Animals , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Membrane Permeability , Cytosol/metabolism , Epithelium/metabolism , Epithelium/pathology , Feeder Cells , Gene Knockdown Techniques , Genetic Vectors/genetics , Humans , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Lentivirus/genetics , Mice , MicroRNAs/genetics , NIH 3T3 Cells , Polycystic Kidney, Autosomal Dominant/metabolism , Polycystic Kidney, Autosomal Dominant/pathology , Primary Cell Culture , Protein Interaction Mapping , TRPP Cation Channels/genetics
10.
Mol Genet Metab ; 102(3): 339-42, 2011 Mar.
Article En | MEDLINE | ID: mdl-21195649

Cystinosis is an autosomal recessive lysosomal storage disease caused by mutations in CTNS. The most prevalent CTNS mutation is a homozygous 57-kb deletion that also includes an adjacent gene named SHPK (CARKL), encoding sedoheptulokinase. Patients with this deletion have elevated urinary concentrations of sedoheptulose. Using derivatisation with pentafluorobenzyl hydroxylamine and liquid chromatography-tandem mass spectrometry (LC-MS/MS), we developed a new sensitive method for the quantification of sedoheptulose in dried blood spots. This method can be utilized as a quick screening test to detect cystinosis patients homozygous for the 57-kb deletion in CTNS; which is the most common mutation of cystinosis. Sedoheptulose concentrations in the deleted patients were 6 to 23 times above the upper limit for controls. The assessment of sedoheptulose in a bloodspot from a known cystinosis patient homozygous for the 57-kb deletion retrieved from the Dutch neonatal screening program showed that sedoheptulose was already elevated in the neonatal period. There was no overlap in sedoheptulose levels between cystinosis patients homozygous for the 57-kb deletion and cystinosis patients not homozygous for this deletion. Our presented method can be used prior to mutation analysis to detect cystinosis patients homozygous for the 57-kb deletion. We feel that the presented method enables fast (pre)-symptomatic detection of cystinosis patients homozygous for the 57-kb deletion, allowing early treatment.


Cystinosis/diagnosis , Cystinosis/enzymology , Gene Deletion , Heptoses/blood , Neonatal Screening/methods , Amino Acid Transport Systems, Neutral/genetics , Cystinosis/blood , Cystinosis/genetics , Humans , Infant, Newborn , Phosphotransferases (Alcohol Group Acceptor)/genetics , Reproducibility of Results , Sensitivity and Specificity , Tandem Mass Spectrometry , Transcription Factors/genetics
11.
J Inherit Metab Dis ; 33(6): 787-93, 2010 Dec.
Article En | MEDLINE | ID: mdl-20814825

INTRODUCTION: Cystinosis is an autosomal recessive disorder leading to intralysosomal cystine accumulation in various tissues. It causes renal Fanconi syndrome and end stage renal failure around the age of 10 years if not treated with cysteamine. Children with cystinosis seem to have a normal intelligence but frequently show learning difficulties. These problems may be due to specific neurocognitive deficits rather than impaired renal function. Whether cysteamine treatment can improve cognitive functioning of cystinosis patients is thus far unknown. We aim to analyze neurocognitive functioning of school-aged cystinosis patients treated with cysteamine in order to identify specific deficits that can lead to learning difficulties. PATIENTS AND METHODS: Fourteen Dutch and Belgian school-aged cystinosis patients were included. Glomerular filtration rate was estimated using the Schwartz formula. Children were tested for general intelligence, visual-motor integration, inhibition, interference, sustained attention, accuracy, planning, visual memory, processing speed, motor planning, fluency and speed, and behavioural and emotional functioning using standardized methods. RESULTS: Glomerular filtration rate ranged from 22 to 120 ml min(-1) 1.73 m(-2). Median full-scale intelligence was below the average of a normal population (87, range 60-132), with a discrepancy between verbal (median 95, range 60-125) and performance (median 87, range 65-130) intelligence. Over 50% of the patients scored poorly on visual-motor integration, sustained attention, visual memory, planning, or motor speed. The other tested areas showed no differences between patients' and normal values. CONCLUSION: Neurocognitive diagnostics are indicated in cystinosis patients. Early recognition of specific deficits and supervision from special education services might reduce learning difficulties and improve school careers.


Cognition/physiology , Cystinosis/physiopathology , Cystinosis/psychology , Adolescent , Belgium , Child , Child Behavior/physiology , Cystinosis/epidemiology , Emotions/physiology , Female , Humans , Intelligence Tests , Male , Memory, Short-Term/physiology , Mental Recall/physiology , Nervous System Physiological Phenomena , Netherlands , Population
12.
Minerva Pediatr ; 62(3): 307-14, 2010 Jun.
Article En | MEDLINE | ID: mdl-20467383

Cystinosis is a rare autosomal recessive disorder characterized by the intralysosomal accumulation of cystine in all tissues due to mutations in the CTNS gene (17p13.3). Infantile nephropatic cystinosis is the most severe and the most frequent form of the disease. It causes renal Fanconi syndrome, leading to end stage renal failure around the age of 10 years if left untreated. Cystine accumulation also affects the eyes, muscles, central nervous system and various endocrine organs. Children with cystinosis often suffer from growth retardation, which is multifactorial in origin. It is not only caused by a decreased renal function, but is aggravated by a poor metabolic status due to renal Fanconi syndrome, pronounced feeding difficulties, often requiring tube feeding and possibly cystine accumulation in the bone. Longitudinal growth can be improved by the correction of metabolic and nutritional deficits and by the treatment of hypothyroidism. The cystine depleting drug cysteamine slows down the progression of renal disease, protects extra-renal organs, accelerates growth and therefore should be administered as early as possible. Despite these treatment strategies, growth retardation remains a severe complication in cystinosis patients and frequently requires the administration of recombinant human growth hormone.


Amino Acid Metabolism, Inborn Errors/complications , Cystine/metabolism , Growth Disorders/etiology , Lysosomes/metabolism , Child , Cysteamine/therapeutic use , Forecasting , Growth Disorders/therapy , Human Growth Hormone/therapeutic use , Humans , Recombinant Proteins/therapeutic use
13.
Eur J Pediatr ; 168(11): 1291-304, 2009 Nov.
Article En | MEDLINE | ID: mdl-19562370

This review deals with podocyte proteins that play a significant role in the structure and function of the glomerular filter. Genetic linkage studies has identified several genes involved in the development of nephrotic syndrome and contributed to the understanding of the pathophysiology of glomerular proteinuria and/or focal segmental glomerulosclerosis. Here, we describe already well-characterized genetic diseases due to mutations in nephrin, podocin, CD2AP, alpha-actinin-4, WT1, and laminin beta2 chain, as well as more recently identified genetic abnormalities in TRPC6, phospholipase C epsilon, and the proteins encoded by the mitochondrial genome. In addition, the role of the proteins which have shown to be important for the structure and functions by gene knockout studies in mice, are also discussed. Furthermore, some rare syndromes with glomerular involvement, in which molecular defects have been recently identified, are briefly described. In summary, this review updates the current knowledge of genetic causes of congenital and childhood nephrotic syndrome and provides new insights into mechanisms of glomerular dysfunction.


Genetic Markers/genetics , Glomerulosclerosis, Focal Segmental/genetics , Kidney Glomerulus/physiopathology , Podocytes , Actinin/genetics , Adaptor Proteins, Signal Transducing/genetics , Cytoskeletal Proteins/genetics , Genome , Glomerulosclerosis, Focal Segmental/physiopathology , Humans , Intracellular Signaling Peptides and Proteins/genetics , Laminin/genetics , Membrane Proteins/genetics , Mutation , Phosphoinositide Phospholipase C/genetics , Proteinuria/genetics , TRPC Cation Channels/genetics , TRPC6 Cation Channel , WT1 Proteins/genetics
14.
Eur J Pediatr ; 167(7): 807-10, 2008 Jul.
Article En | MEDLINE | ID: mdl-17874131

Wolf-Hirschhorn syndrome is characterized by severe growth and mental retardation, microcephaly, seizures and 'Greek helmet' facies, caused by partial deletion of the short arm of chromosome 4. Growth charts are given from 0-4 years of age, based on the study of 101 individuals. Use of these specific growth charts is recommended, because standard growth charts are inapplicable for patients with WHS.


Growth , Wolf-Hirschhorn Syndrome , Birth Weight , Body Height , Body Weight , Child, Preschool , Female , Humans , Infant , Infant, Newborn , Male , Medical Records , Multicenter Studies as Topic , Sex Factors
15.
Ned Tijdschr Geneeskd ; 151(43): 2377-80, 2007 Oct 27.
Article Nl | MEDLINE | ID: mdl-18019214

Dent's disease is an X-linked disorder, characterized by generalized proximal tubular dysfunction, nephrolithiasis, nephrocalcinosis and the development ofend-stage renal disease, generally occurring after the age of thirty. In the majority of cases, the disease is caused by mutations in the CLCN5-gene. The pathogenesis of the disease has not yet been clarified. Defective recycling of multi-ligand proximal tubular receptors megalin and cubilin is considered responsible for the defective reabsorption of low molecular weight proteins, albumin, hormones and vitamins. Treatment with thiazide diuretics to diminish the hypercalciuria in combination with citrate supplements might prevent renal stone formation and deterioration of renal function. In the laboratory ofDNA diagnostics in the Radboud University Nijmegen Medical Centre, the molecular analysis of the CLCN5-gene in patients suspected with this disease is performed.


Chloride Channels/genetics , Chromosomes, Human, X , Genetic Diseases, X-Linked/genetics , Kidney Diseases/genetics , Phosphoric Monoester Hydrolases/genetics , DNA Mutational Analysis , Diagnosis, Differential , Female , Genetic Diseases, X-Linked/diagnosis , Humans , Kidney Diseases/diagnosis , Male , Nephrons/pathology , Nephrons/physiology , Oculocerebrorenal Syndrome/diagnosis , Oculocerebrorenal Syndrome/genetics , Point Mutation , Syndrome
16.
Kidney Int ; 72(10): 1233-41, 2007 Nov.
Article En | MEDLINE | ID: mdl-17851469

The multidrug resistance gene 1 product, P-glycoprotein (P-gp), is expressed in several excretory organs, including the apical membrane of proximal tubules. After inducing acute renal failure, P-gp expression is upregulated and this might be a protective function by pumping out toxicants and harmful products of oxidative stress. We characterized renal function of P-gp knockout mice and studied its consequences in renal ischemic damage. Compared with wild-type mice, knockout mice have a lower glomerular filtration rate and renal plasma flow. An augmented urinary excretion of sodium, numerous amino acids, calcium, glucose, and low molecular weight proteins was observed along with an increased diuresis. A higher lithium plasma clearance in the knockout mice suggested proximal tubular dysfunction. Electron microscopy showed mitochondrial abnormalities in proximal tubular cells that could account for decreased adenosine triphosphate levels in the cortex. After inducing ischemia, wild-type mice showed a decrease in creatinine clearance and severe proximal tubular necrosis. In contrast, knockout mice had no signs of tubular damage. Our data indicate that P-gp knockout mice have impaired renal function but are protected against ischemic renal injury.


ATP Binding Cassette Transporter, Subfamily B, Member 1/deficiency , Ischemia/prevention & control , Kidney Tubules, Proximal/physiopathology , Acute Kidney Injury/physiopathology , Amino Acids/urine , Animals , Calcium/urine , Diuresis , Fluorescent Antibody Technique , Glomerular Filtration Rate/genetics , Glycosuria , Immunohistochemistry , Ischemia/etiology , Kidney Tubules, Proximal/ultrastructure , Male , Mice , Mice, Inbred Strains , Mice, Knockout , Mitochondria/pathology , Mitochondria/ultrastructure , Proteinuria , Renal Circulation/genetics , Sodium/urine
17.
Kidney Int ; 72(10): 1198-203, 2007 Nov.
Article En | MEDLINE | ID: mdl-17713465

Focal segmental glomerulosclerosis (FSGS) is a histologic diagnosis in several kidney diseases characterized by proteinuria and a severe decrease in kidney function. Mutations in several genes were found in patients with primary FSGS, one of which is a CD2-associated protein CD2AP (originally referred to as CMS). This gene encodes an adaptor protein that plays a role in endocytosis, cell motility, and cell survival. Mice deficient in Cd2ap (the mouse homolog) die due to kidney failure, while heterozygous mice develop lesions similar to those of FSGS patients. In the kidney, CD2AP regulates the actin cytoskeleton. The only previously described patient with CD2AP mutation had a severely truncated protein. In this study, we describe a patient with a novel mutation resulting in a premature stop codon yielding a protein truncated by only 4%. This shortened CD2AP protein displays a significantly decreased F-actin binding efficiency in vitro with no expression of the mutated allele in the patient's lymphocytes. Heterozygous expression of the CD2AP mutation in both parents did not lead to any kidney pathology, as both have normal glomerular filtration rates and no proteinuria.


Adaptor Proteins, Signal Transducing/genetics , Cytoskeletal Proteins/genetics , Glomerulosclerosis, Focal Segmental/genetics , Glomerulosclerosis, Focal Segmental/pathology , Homozygote , Mutation , Actins/metabolism , Amino Acid Sequence , Binding Sites , Biopsy , Cadaver , Child, Preschool , Codon, Terminator/genetics , Consanguinity , Glomerular Filtration Rate , Glomerulosclerosis, Focal Segmental/surgery , Humans , Kidney Glomerulus/metabolism , Kidney Glomerulus/physiology , Kidney Glomerulus/ultrastructure , Kidney Transplantation , Male , Molecular Sequence Data , Nucleic Acid Amplification Techniques , Protein Binding , Protein Structure, Tertiary , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Treatment Outcome
18.
Ned Tijdschr Geneeskd ; 151(12): 692-4, 2007 Mar 24.
Article Nl | MEDLINE | ID: mdl-17447595

Apparent mineralocorticoid excess (AME) is an autosomal recessive disease caused by deficiency of the enzyme 11beta-hydroxysteroid dehydrogenase type 2 (11beta-HSD2). 11beta-HSD2 converts cortisol into inactive cortisone and prevents the stimulation of the mineralocorticoid receptor by cortisol. In patients with AME, an enhanced stimulation of mineralocorticoid receptors by cortisol in the distal nephron causes an elevated sodium reabsorption and increased potassium excretion. Sodium retention leads to severe low renin hypertension. The diagnosis of AME is based on the detection of an increased concentration of cortisol metabolites and a low or undetectable concentration of cortisone metabolites in urine. Molecular analysis of the HSD11B2 gene confirms the diagnosis. AME is successfully treated by potassium-sparing diuretics, sometimes in combination with loop diuretics (furosemide). Mild forms of AME might occur more frequently than is currently known and should be suspected in patients with hypertension, hypokalemia and decreased plasma renin concentration. Since liquorice can induce the clinical symptoms of AME due to reversible inhibition of the 11beta-HSD2 enzyme by glycyrrhetinic acid, the active ingredient of liquorice, patients suspected of having AME should not consume liquorice.


11-beta-Hydroxysteroid Dehydrogenase Type 2/genetics , Hydrocortisone/metabolism , Mineralocorticoid Excess Syndrome, Apparent/genetics , Sodium Channel Blockers/therapeutic use , Diagnosis, Differential , Glycyrrhiza/adverse effects , Humans , Hypertension/etiology , Hypokalemia/etiology , Mineralocorticoid Excess Syndrome, Apparent/diagnosis , Mineralocorticoid Excess Syndrome, Apparent/drug therapy
19.
Kidney Int ; 71(1): 44-52, 2007 Jan.
Article En | MEDLINE | ID: mdl-17035937

Global glomerulosclerosis can be divided in the vascular (obsolescent) type and the glomerulopathic (solidified) type. In biopsies from children with recurrent nephrotic syndrome owing to minimal change nephropathy (MCN), we noticed small, globally sclerosed glomeruli that appeared to be distinct from global glomerulosclerosis. These small sclerosed glomeruli are best described as involuted glomeruli. We have characterized these involuted glomeruli in detail. We studied biopsies of 18 children (11 male, 7 female) with frequently relapsing MCN and evaluated possible explanatory variables. The involuted glomeruli can be differentiated from the other types of global glomerulosclerosis. Most notable is the presence of vital podocytes and parietal epithelial cells, which have retained their staining characteristics, in between the matrix, and the absence of periglomerular and tubulo-interstitial fibrosis. We observed involuted glomeruli in 12 out of 18 biopsies; the median percentage of involuted glomeruli was 6% (range 0-33%). The percentage of involuted glomeruli correlated with age at renal biopsy and the interval between onset of disease and time of renal biopsy, but not with gender, age at onset of disease, or prednisone dose. Multivariate analysis revealed that the interval between onset of disease and time of renal biopsy was the only independent predictor. In conclusion, glomerular involution is a special form of global glomerulosclerosis. The absence of periglomerular and tubulo-interstitial fibrosis suggests a different pathogenesis. Glomerular involution is a slow process. The clinical data suggest that involution is related to the duration of the disease process.


Kidney Glomerulus/pathology , Nephrosis, Lipoid/diagnosis , Nephrosis, Lipoid/pathology , Child , Child, Preschool , Female , Humans , Immunohistochemistry , Infant , Kidney Glomerulus/metabolism , Male , Microscopy, Electron , Nephrosis, Lipoid/metabolism , Recurrence
20.
Clin Nephrol ; 66(4): 306-9, 2006 Oct.
Article En | MEDLINE | ID: mdl-17064000

Cystinosis is a rare metabolic disorder characterized by lysosomal cystine accumulation leading to multi-organ damage, with kidneys being clinically first affected. Longer survival of cystinosis patients due to successful renal replacement therapy, revealed previously unknown extra-renal symptoms of cystinosis, generally appearing after the first decade. Respiratory insufficiency caused by overall respiratory muscle myopathy is a severely invalidating and sometimes a life-threatening complication of cystinosis. We report a successful treatment of hypoventilation, due to diaphragm myopathy in a cystinosis patient, by nocturnal non-invasive positive pressure ventilation (NIPPV). After initiation of NIPPV the clinical condition of the patient improved and blood-gasses normalized, indicating that this treatment modality should be considered in cystinosis patients with severe respiratory insufficiency.


Cystinosis/complications , Positive-Pressure Respiration/methods , Respiration Disorders/complications , Respiration Disorders/therapy , Adult , Blood Gas Analysis , Humans , Male , Respiratory Physiological Phenomena , Supine Position
...