Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Nefrologia (Engl Ed) ; 44(1): 90-99, 2024.
Article in English | MEDLINE | ID: mdl-37150673

ABSTRACT

INTRODUCTION: Activation of the focal adhesion kinase (FAK) in podocytes is involved in the pathogenesis of minimal change disease (MCD), but the pathway leading to its activation in this disease is unknown. Here, we tested whether podocyte ß1 integrin is the upstream modulator of FAK activation and podocyte injury in experimental models of MCD-like injury. METHODS: We used lipopolysaccharide (LPS) and MCD sera to induce MCD-like changes in vivo and in cultured human podocytes, respectively. We performed functional studies using specific ß1 integrin inhibitors in vivo and in vitro, and integrated histological analysis, western blotting, and immunofluorescence to assess for morphological and molecular changes in podocytes. By ELISA, we measured serum LPS levels in 35 children with MCD or presumed MCD (idiopathic nephrotic syndrome [INS]) and in 18 healthy controls. RESULTS: LPS-injected mice showed morphological (foot process effacement, and normal appearing glomeruli on light microscopy) and molecular features (synaptopodin loss, nephrin mislocalization, FAK phosphorylation) characteristic of human MCD. Administration of a ß1 integrin inhibitor to mice abrogated FAK phosphorylation, and ameliorated proteinuria and podocyte injury following LPS. Children with MCD/INS in relapse had higher serum LPS levels than controls. In cultured human podocytes, ß1 integrin blockade prevented cytoskeletal rearrangements following exposure to MCD sera in relapse. CONCLUSIONS: Podocyte ß1 integrin activation is an upstream mediator of FAK phosphorylation and podocyte injury in models of MCD-like injury.


Subject(s)
Nephrosis, Lipoid , Nephrotic Syndrome , Podocytes , Child , Mice , Humans , Animals , Nephrosis, Lipoid/chemically induced , Integrin beta1/metabolism , Lipopolysaccharides/metabolism , Models, Theoretical , Recurrence
2.
Kidney Int Rep ; 7(4): 797-809, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35497798

ABSTRACT

Introduction: Minimal change disease (MCD) is considered a podocyte disorder triggered by unknown circulating factors. Here, we hypothesized that the endothelial cell (EC) is also involved in MCD. Methods: We studied 45 children with idiopathic nephrotic syndrome (44 had steroid sensitive nephrotic syndrome [SSNS], and 12 had biopsy-proven MCD), 21 adults with MCD, and 38 healthy controls (30 children, 8 adults). In circulation, we measured products of endothelial glycocalyx (EG) degradation (syndecan-1, heparan sulfate [HS] fragments), HS proteoglycan cleaving enzymes (matrix metalloprotease-2 [MMP-2], heparanase activity), and markers of endothelial activation (von Willebrand factor [vWF], thrombomodulin) by enzyme-linked immunosorbent assay (ELISA) and mass spectrometry. In human kidney tissue, we assessed glomerular EC (GEnC) activation by immunofluorescence of caveolin-1 (n = 11 MCD, n = 5 controls). In vitro, we cultured immortalized human GEnC with sera from control subjects and patients with MCD/SSNS sera in relapse (n = 5 per group) and performed Western blotting of thrombomodulin of cell lysates as surrogate marker of endothelial activation. Results: In circulation, median concentrations of all endothelial markers were higher in patients with active disease compared with controls and remained high in some patients during remission. In the MCD glomerulus, caveolin-1 expression was higher, in an endothelial-specific pattern, compared with controls. In cultured human GEnC, sera from children with MCD/SSNS in relapse increased thrombomodulin expression compared with control sera. Conclusion: Our data show that alterations involving the systemic and glomerular endothelium are nearly universal in patients with MCD and SSNS, and that GEnC can be directly activated by circulating factors present in the MCD/SSNS sera during relapse.

3.
Calcif Tissue Int ; 108(4): 423-438, 2021 04.
Article in English | MEDLINE | ID: mdl-33452890

ABSTRACT

This manuscript discusses mineral and bone disorders of chronic kidney disease (MBD-CKD) in pediatric patients with special emphasis on the underlying pathophysiology, the causes and clinical profile of growth retardation, the alterations in the growth plate, the strategies to optimize growth and the medical recommendations for prevention and treatment.


Subject(s)
Bone Diseases, Metabolic , Chronic Kidney Disease-Mineral and Bone Disorder , Renal Insufficiency, Chronic , Child , Humans , Minerals , Renal Insufficiency, Chronic/complications
4.
Front Med (Lausanne) ; 8: 761600, 2021.
Article in English | MEDLINE | ID: mdl-35004732

ABSTRACT

Minimal change disease (MCD) is the most common type of idiopathic nephrotic syndrome in childhood and represents about 15% cases in adults. It is characterized by massive proteinuria, edema, hypoalbuminemia, and podocyte foot process effacement on electron microscopy. Clinical and experimental studies have shown an association between MCD and immune dysregulation. Given the lack of inflammatory changes or immunocomplex deposits in the kidney tissue, MCD has been traditionally thought to be mediated by an unknown circulating factor(s), probably released by T cells that directly target podocytes leading to podocyte ultrastructural changes and proteinuria. Not surprisingly, research efforts have focused on the role of T cells and podocytes in the disease process. Nevertheless, the pathogenesis of the disease remains a mystery. More recently, B cells have been postulated as an important player in the disease either by activating T cells or by releasing circulating autoantibodies against podocyte targets. There are also few reports of endothelial injury in MCD, but whether glomerular endothelial cells play a role in the disease remains unexplored. Genome-wide association studies are providing insights into the genetic susceptibility to develop the disease and found a link between MCD and certain human haplotype antigen variants. Altogether, these findings emphasize the complex interplay between the immune system, glomerular cells, and the genome, raising the possibility of distinct underlying triggers and/or mechanisms of proteinuria among patients with MCD. The heterogeneity of the disease and the lack of good animal models of MCD remain major obstacles in the understanding of MCD. In this study, we will review the most relevant candidate mediators and mechanisms of proteinuria involved in MCD and the current models of MCD-like injury.

6.
Pediatr Res ; 82(1): 148-154, 2017 07.
Article in English | MEDLINE | ID: mdl-28376076

ABSTRACT

BackgroundIn a model of growth retardation secondary to chronic kidney disease (CKD) induced by adenine, this study explores the effects of growth hormone (GH) therapy on growth plate and mineral metabolism.MethodsWeaning female rats receiving a 0.5% adenine diet during 21 days, untreated (AD) or treated with GH (ADGH) for 1 week, were compared with control rats receiving normal diet, either ad libitum or pair-fed with AD animals. AD and ADGH rats had similarly elevated serum concentrations of urea nitrogen, parathyroid hormone (PTH), and fibroblast growth factor 23 (FGF23).ResultsUremia induced by adenine caused growth retardation and disturbed growth cartilage chondrocyte hypertrophy. We demonstrated marked expression of aquaporin 1 in the growth plate, but its immunohistochemical signal and the expression levels of other proteins potentially related with chondrocyte enlargement, such as Na-K-2Cl cotransporter, insulin-like growth factor 1 (IGF-1), and IGF-1 receptor, were not different among the four groups of rats. The distribution pattern of vascular endothelial growth factor was also similar. AD rats developed femur bone structure abnormalities analyzed by micro-computerized tomography.ConclusionGH treatment accelerated longitudinal growth velocity, stimulated the proliferation and enlargement of chondrocytes, and did not modify the elevated serum PTH or FGF23 concentrations or the abnormal bone structure.


Subject(s)
Growth Hormone/pharmacology , Growth Plate/drug effects , Minerals/metabolism , Uremia/metabolism , Adenine , Animals , Blood Urea Nitrogen , Chondrocytes/metabolism , Female , Fibroblast Growth Factors/blood , Inflammation , Parathyroid Hormone/blood , Rats , Rats, Sprague-Dawley , Uremia/chemically induced , Vascular Endothelial Growth Factor A/metabolism , X-Ray Microtomography
7.
Am J Physiol Renal Physiol ; 309(1): F57-62, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-25972508

ABSTRACT

Growth retardation is a major manifestation of chronic kidney disease (CKD) in pediatric patients. The involvement of the various pathogenic factors is difficult to evaluate in clinical studies. Here, we present an experimental model of adenine-induced CKD for the study of growth failure. Three groups (n = 10) of weaning female rats were studied: normal diet (control), 0.5% adenine diet (AD), and normal diet pair fed with AD (PF). After 21 days, serum urea nitrogen, creatinine, parathyroid hormone (PTH), weight and length gains, femur osseous front advance as an index of longitudinal growth rate, growth plate histomorphometry, chondrocyte proliferative activity, bone structure, aorta calcifications, and kidney histology were analyzed. Results are means ± SE. AD rats developed renal failure (serum urea nitrogen: 70 ± 6 mg/dl and creatinine: 0.6 ± 0.1 mg/dl) and secondary hyperparathyroidism (PTH: 480 ± 31 pg/ml). Growth retardation of AD rats was demonstrated by lower weight (AD rats: 63.3 ± 4.8 g, control rats: 112.6 ± 4.7 g, and PF rats: 60.0 ± 3.8 g) and length (AD rats: 7.2 ± 0.2 cm, control rats: 11.1 ± 0.3 cm, and PF rats: 8.1 ± 0.3 cm) gains as well as lower osseous front advances (AD rats: 141 ± 13 µm/day, control rats: 293 ± 16 µm/day, and PF rats: 251 ± 10 µm/day). The processes of chondrocyte maturation and proliferation were impaired in AD rats, as shown by lower growth plate terminal chondrocyte height (21.7 ± 2.3 vs. 26.2 ± 1.9 and 23.9 ± 1.3 µm in control and PF rats) and proliferative activity index (AD rats: 30 ± 2%, control rats: 38 ± 2%, and PF rats: 42 ± 3%). The bone primary spongiosa structure of AD rats was markedly disorganized. In conclusion, adenine-induced CKD in young rats is associated with growth retardation and disturbed endochondral ossification. This animal protocol may be a useful new experimental model to study growth in CKD.


Subject(s)
Disease Models, Animal , Growth Disorders/etiology , Growth Plate/physiopathology , Kidney Failure, Chronic/complications , Uremia/complications , Adenine , Animals , Female , Growth , Growth Disorders/physiopathology , Kidney/pathology , Kidney Failure, Chronic/chemically induced , Kidney Failure, Chronic/physiopathology , Neovascularization, Physiologic , Nutritional Status , Rats, Sprague-Dawley , Uremia/chemically induced , Uremia/physiopathology
8.
Pediatr Nephrol ; 30(12): 2099-107, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25823989

ABSTRACT

In the absence of a gastrointestinal origin, a maintained hyperchloremic metabolic acidosis must raise the diagnostic suspicion of renal tubular acidosis (RTA). Unlike adults, in whom RTA is usually secondary to acquired causes, children most often have primary forms of RTA resulting from an inherited genetic defect in the tubular proteins involved in the renal regulation of acid-base homeostasis. According to their pathophysiological basis, four types of RTA are distinguished. Distal type 1 RTA, proximal type 2 RTA, mixed-type 3 RTA, and type 4 RTA can be differentiated based on the family history, the presenting manifestations, the biochemical profile, and the radiological findings. Functional tests to explore the proximal wasting of bicarbonate and the urinary acidification capacity are also useful diagnostic tools. Although currently the molecular basis of the disease can frequently be discovered by gene analysis, patients with RTA must undergo a detailed clinical study and laboratory work-up in order to understand the pathophysiology of the disease and to warrant a correct and accurate diagnosis.


Subject(s)
Acidosis, Renal Tubular/diagnosis , Kidney/physiopathology , Acidosis, Renal Tubular/genetics , Acidosis, Renal Tubular/physiopathology , Child , Child, Preschool , Humans
9.
Pediatr Nephrol ; 28(4): 595-603, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23179196

ABSTRACT

Over the last decade the discovery of fibroblast growth factor 23 (FGF23) and the progressive and ongoing clarification of its role in phosphate and mineral metabolism have led to expansion of the diagnostic spectrum of primary hypophosphatemic syndromes. This article focuses on the impairment of growth in these syndromes. Growth retardation is a common, but not constant, feature and it presents with large variability. As a result of the very low prevalence of other forms of primary hypophosphatemic syndromes, the description of longitudinal growth and the pathogenesis of its impairment have been mostly studied in X-linked hypophosphatemia (XLH) patients and in Hyp mice, the animal model of this disease. In general, children with XLH have short stature with greater shortness of lower limbs than trunk. Treatment with phosphate supplements and 1α vitamin D derivatives heals active lesions of rickets, but does not normalize growth of XLH patients. Patients might benefit from recombinant human growth hormone (rhGH) therapy, which may accelerate the growth rate without increasing body disproportion or correcting hypophosphatemia. These clinical data as well as research findings obtained in Hyp mice suggest that the pathogenesis of defective growth in XLH and other hypophosphatemic syndromes is not entirely dependent on the mineralization disorder and point to other effects of hypophosphatemia itself or FGF23 on the metabolism of bone and growth plate.


Subject(s)
Body Height , Bone Development , Bone and Bones/physiopathology , Familial Hypophosphatemic Rickets/complications , Genetic Diseases, X-Linked , Growth Disorders/etiology , Phosphates/metabolism , Animals , Body Height/genetics , Bone Development/genetics , Bone and Bones/metabolism , Dietary Supplements , Disease Models, Animal , Familial Hypophosphatemic Rickets/genetics , Familial Hypophosphatemic Rickets/metabolism , Familial Hypophosphatemic Rickets/physiopathology , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/metabolism , Growth Disorders/metabolism , Growth Disorders/physiopathology , Human Growth Hormone/therapeutic use , Humans , Mice , Mice, Transgenic , PHEX Phosphate Regulating Neutral Endopeptidase/genetics , PHEX Phosphate Regulating Neutral Endopeptidase/metabolism , Phosphates/therapeutic use , Recombinant Proteins/therapeutic use , Vitamins/therapeutic use
10.
PLoS One ; 7(4): e34788, 2012.
Article in English | MEDLINE | ID: mdl-22493717

ABSTRACT

Rapamycin, an immunosuppressant agent used in renal transplantation with antitumoral properties, has been reported to impair longitudinal growth in young individuals. As growth hormone (GH) can be used to treat growth retardation in transplanted children, we aimed this study to find out the effect of GH therapy in a model of young rat with growth retardation induced by rapamycin administration. Three groups of 4-week-old rats treated with vehicle (C), daily injections of rapamycin alone (RAPA) or in combination with GH (RGH) at pharmacological doses for 1 week were compared. GH treatment caused a 20% increase in both growth velocity and body length in RGH animals when compared with RAPA group. GH treatment did not increase circulating levels of insulin-like growth factor I, a systemic mediator of GH actions. Instead, GH promoted the maturation and hypertrophy of growth plate chondrocytes, an effect likely related to AKT and ERK1/2 mediated inactivation of GSK3ß, increase of glycogen deposits and stabilization of ß-catenin. Interestingly, GH did not interfere with the antiproliferative and antiangiogenic activities of rapamycin in the growth plate and did not cause changes in chondrocyte autophagy markers. In summary, these findings indicate that GH administration improves longitudinal growth in rapamycin-treated rats by specifically acting on the process of growth plate chondrocyte hypertrophy but not by counteracting the effects of rapamycin on proliferation and angiogenesis.


Subject(s)
Chondrocytes/drug effects , Chondrogenesis/drug effects , Growth Hormone/therapeutic use , Growth Plate/drug effects , Neovascularization, Physiologic/drug effects , Animals , Antibiotics, Antineoplastic/administration & dosage , Autophagy/drug effects , Cell Differentiation/drug effects , Child , Chondrocytes/metabolism , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Gene Expression/drug effects , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Growth Hormone/administration & dosage , Growth Plate/blood supply , Growth Plate/growth & development , Humans , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Rats , Rats, Sprague-Dawley , Sirolimus/administration & dosage , beta Catenin/genetics , beta Catenin/metabolism
11.
Kidney Int ; 78(6): 561-8, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20555322

ABSTRACT

Rapamycin, a potent immunosuppressant used in renal transplantation, has been reported to impair longitudinal growth in experimental studies. Rapamycin is both antiproliferative and antiangiogenic; therefore, it has the potential to disrupt vascular endothelial growth factor (VEGF) action in the growth plate and to interfere with insulin-like growth factor I (IGF-I) signaling. To further investigate the mechanisms of rapamycin action on longitudinal growth, we gave the 4-week-old rats rapamycin daily for two weeks. Compared with a vehicle-treated group, rapamycin-treated animals were severely growth retarded and had marked alterations in the growth plate. Vascular invasion was disturbed in the rapamycin group, there was a significant reduction in osteoclast cells near the chondro-osseus junction, and there was lower VEGF protein and mRNA expression in the terminal chondrocytes of the growth cartilage. Compared with the control group, the rapamycin group had higher levels of circulating IGF-I as well as the mRNAs for IGF-I and of the receptors of IGF-I and growth hormone in the liver but not in the growth cartilage. Thus our findings explain the adverse effect of rapamycin on growth plate dynamics. This should be taken into account when the drug is administered to children.


Subject(s)
Growth Plate/drug effects , Neovascularization, Physiologic/drug effects , Sirolimus/adverse effects , Animals , Animals, Newborn , Chondrocytes/pathology , Growth/drug effects , Growth Plate/growth & development , Immunosuppressive Agents/adverse effects , Insulin-Like Growth Factor I/analysis , Osteoclasts/pathology , RNA, Messenger/analysis , Rats , Receptor, IGF Type 1/genetics , Vascular Endothelial Growth Factor A/analysis , Vascular Endothelial Growth Factor A/genetics
12.
Am J Physiol Renal Physiol ; 297(3): F639-45, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19587145

ABSTRACT

Hypokalemic tubular disorders may lead to growth retardation which is resistant to growth hormone (GH) treatment. The mechanism of these alterations is unknown. Weaning female rats were grouped (n = 10) in control, potassium-depleted (KD), KD treated with intraperitoneal GH at 3.3 mg x kg(-1) x day(-1) during the last week (KDGH), and control pair-fed with KD (CPF). After 2 wk, KD rats were growth retarded compared with CPF rats, the osseous front advance (+/-SD) being 67.07 +/- 10.44 and 81.56 +/- 12.70 microm/day, respectively. GH treatment did not accelerate growth rate. The tibial growth plate of KD rats had marked morphological alterations: lower heights of growth cartilage (228.26 +/- 23.58 microm), hypertrophic zone (123.68 +/- 13.49 microm), and terminal chondrocytes (20.8 +/- 2.39 microm) than normokalemic CPF (264.21 +/- 21.77, 153.18 +/- 15.80, and 24.21 +/- 5.86 microm). GH administration normalized these changes except for the distal chondrocyte height. Quantitative PCR of insulin-like growth factor I (IGF-I), IGF-I receptor, and GH receptor genes in KD growth plates showed downregulation of IGF-I and upregulation of IGF-I receptor mRNAs, without changes in their distribution as analyzed by immunohistochemistry and in situ hybridization. GH did not further modify IGF-I mRNA expression. KD rats had normal hepatic IGF-I mRNA levels and low serum IGF-I values. GH increased liver IGF-I mRNA, but circulating IGF-I levels remained reduced. This study discloses the structural and molecular alterations induced by potassium depletion on the growth plate and shows that the lack of response to GH administration is associated with persistence of the disturbed process of chondrocyte hypertrophy and depressed mRNA expression of local IGF-I in the growth plate.


Subject(s)
Growth Disorders/drug therapy , Growth Plate/drug effects , Human Growth Hormone/pharmacology , Hyperkalemia/complications , Insulin-Like Growth Factor I/metabolism , Animal Nutritional Physiological Phenomena , Animals , Body Size , Cell Proliferation/drug effects , Chondrocytes/drug effects , Chondrocytes/metabolism , Chronic Disease , Disease Models, Animal , Female , Growth Disorders/etiology , Growth Disorders/metabolism , Growth Disorders/physiopathology , Growth Plate/growth & development , Growth Plate/metabolism , Growth Plate/pathology , Human Growth Hormone/administration & dosage , Humans , Hyperkalemia/metabolism , Hyperkalemia/physiopathology , Injections, Intraperitoneal , Insulin-Like Growth Factor I/genetics , Liver/drug effects , Liver/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptor, IGF Type 1/metabolism , Receptors, Somatotropin/metabolism , Recombinant Proteins/pharmacology , Weaning , Weight Gain
13.
Pediatr Nephrol ; 22(7): 954-61, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17370095

ABSTRACT

Rapamycin is a potent immunosuppressant with antitumoral properties widely used in the field of renal transplantation. To test the hypothesis that the antiproliferative and antiangiogenic activity of rapamycin interferes with the normal structure and function of growth plate and impairs longitudinal growth, 4-week-old male rats (n = 10/group) receiving 2 mg/kg per day of intraperitoneal rapamycin (RAPA) or vehicle (C) for 14 days were compared. Rapamycin markedly decreased bone longitudinal growth rate (94 +/- 3 vs. 182 +/- 3 microm/day), body weight gain (60.2 +/- 1.4 vs. 113.6 +/- 1.9 g), food intake (227.8 +/- 2.6 vs. 287.5 +/- 3.4 g), and food efficiency (0.26 +/- 0.00 vs. 0.40 +/- 0.01 g/g). Signs of altered cartilage formation such as reduced chondrocyte proliferation (bromodeoxiuridine-labeled cells 32.9 +/- 1.4 vs. 45.2 +/- 1.1%), disturbed maturation and hypertrophy (height of terminal chondrocytes 26 +/- 0 vs. 29 +/- 0 microm), and decreased cartilage resorption (18.7 +/- 0.5 vs. 31.0 +/- 0.8 tartrate-resistant phosphatase alkaline reactive cells per 100 terminal chondrocytes), together with morphological evidence of altered vascular invasion, were seen in the growth plate of RAPA animals. This study indicates that rapamycin can severely impair body growth in fast-growing rats and distort growth-plate structure and dynamics. These undesirable effects must be kept in mind when rapamycin is administered to children.


Subject(s)
Growth Plate/drug effects , Growth Plate/physiology , Growth/drug effects , Immunosuppressive Agents/pharmacology , Sirolimus/pharmacology , Acid Phosphatase/metabolism , Administration, Oral , Animals , Body Weight/drug effects , Cell Proliferation/drug effects , Chondrocytes/metabolism , Chondrocytes/physiology , Eating/drug effects , Growth Plate/cytology , Immunohistochemistry , Immunosuppressive Agents/blood , Injections, Intraperitoneal , Isoenzymes/metabolism , Male , Rats , Rats, Sprague-Dawley , Sirolimus/blood , Tartrate-Resistant Acid Phosphatase , Vascular Endothelial Growth Factor A/metabolism
14.
Pediatr Nephrol ; 22(6): 825-8, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17216496

ABSTRACT

Distal renal tubular acidosis (RTA) with nerve deafness is caused by mutations in the ATP6V1B1 gene causing defective function of the H+ -ATPase proton pump. We report five acidotic children (four males) from four unrelated families: blood pH 7.21-7.33, serum bicarbonate 10.8-14.7 mEq/l, minimum urinary pH 6.5-7.1 and fractional excretion of bicarbonate in the presence of normal bicarbonatemia 1.1-5.7%. Growth retardation and nephrocalcinosis, but not hypercalciuria, were common presenting manifestations. Hearing was normally preserved in one of the patients whose sister was severely deaf. One child was homozygous for a known mutation in exon 1: C>T (R31X). Three children were homozygous for a splicing mutation, intron 6 + 1G>A. The other patient was a compound heterozygote, having this mutation and a previously unreported mutation in exon 10: G>A (E330K). Our report shows that hearing loss is not always present in the syndrome of distal renal tubular acidosis with nerve deafness and the absence of hypercalciuria at diagnosis and describes a new mutation responsible for the disease in the ATP6V1B1 gene.


Subject(s)
Acidosis, Renal Tubular/genetics , Genetic Predisposition to Disease , Hearing Loss, Sensorineural/genetics , Mutation , Vacuolar Proton-Translocating ATPases/genetics , Acidosis, Renal Tubular/pathology , Acidosis, Renal Tubular/physiopathology , Child, Preschool , DNA Mutational Analysis , Female , Growth Disorders/genetics , Growth Disorders/pathology , Growth Disorders/physiopathology , Hearing Loss, Sensorineural/pathology , Hearing Loss, Sensorineural/physiopathology , Humans , Infant , Male , Nephrocalcinosis/genetics , Nephrocalcinosis/pathology , Nephrocalcinosis/physiopathology , Polymerase Chain Reaction , Syndrome
SELECTION OF CITATIONS
SEARCH DETAIL
...