Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 142
Filter
1.
Eur J Hum Genet ; 28(12): 1681-1693, 2020 12.
Article in English | MEDLINE | ID: mdl-32737436

ABSTRACT

Although over 50 genes are known to cause renal malformation if mutated, the underlying genetic basis, most easily identified in syndromic cases, remains unsolved in most patients. In search of novel causative genes, whole-exome sequencing in a patient with renal, i.e., crossed fused renal ectopia, and extrarenal, i.e., skeletal, eye, and ear, malformations yielded a rare heterozygous variant in the GDF6 gene encoding growth differentiation factor 6, a member of the BMP family of ligands. Previously, GDF6 variants were reported to cause pleiotropic defects including skeletal, e.g., vertebral, carpal, tarsal fusions, and ocular, e.g., microphthalmia and coloboma, phenotypes. To assess the role of GDF6 in the pathogenesis of renal malformation, we performed targeted sequencing in 193 further patients identifying rare GDF6 variants in two cases with kidney hypodysplasia and extrarenal manifestations. During development, gdf6 was expressed in the pronephric tubule of Xenopus laevis, and Gdf6 expression was observed in the ureteric tree of the murine kidney by RNA in situ hybridization. CRISPR/Cas9-derived knockout of Gdf6 attenuated migration of murine IMCD3 cells, an effect rescued by expression of wild-type but not mutant GDF6, indicating affected variant function regarding a fundamental developmental process. Knockdown of gdf6 in Xenopus laevis resulted in impaired pronephros development. Altogether, we identified rare heterozygous GDF6 variants in 1.6% of all renal anomaly patients and 5.4% of renal anomaly patients additionally manifesting skeletal, ocular, or auricular abnormalities, adding renal hypodysplasia and fusion to the phenotype spectrum of GDF6 variant carriers and suggesting an involvement of GDF6 in nephrogenesis.


Subject(s)
Growth Differentiation Factor 6/genetics , Urogenital Abnormalities/genetics , Vesico-Ureteral Reflux/genetics , Adolescent , Adult , Animals , Cell Line , Child , Child, Preschool , Female , Growth Differentiation Factor 6/metabolism , Heterozygote , Humans , Infant , Kidney Tubules/abnormalities , Kidney Tubules/metabolism , Male , Mice , Mutation , Urogenital Abnormalities/pathology , Vesico-Ureteral Reflux/pathology , Xenopus
2.
Eur J Med Genet ; 62(4): 254-258, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30071301

ABSTRACT

Renal tubular dysgenesis (RTD) is a developmental abnormality of the nephron characterized by fetal anuria, oligohydramnios, and severe postnatal hypotension. Genetic forms have an autosomal recessive inheritance and are caused by mutations in genes encoding key components of the renin-angiotensin pathway. We report three patients from two unrelated families with RTD due to pathogenic variants of the angiotensin-converting enzyme (ACE) gene, in whom RTD was associated with microcolon. We also detail key variations of the renin-angiotensin system in one of these infants. The severe intestinal developmental abnormality culminating in microcolon and early terminal ileum perforation/necrotizing enterocolitis is a novel finding not previously associated with RTD, which points to a role of the renin-angiotensin system in gut development.


Subject(s)
Abnormalities, Multiple/genetics , Ileum/abnormalities , Kidney Tubules/abnormalities , Peptidyl-Dipeptidase A/genetics , Phenotype , Abnormalities, Multiple/pathology , Female , Humans , Infant, Newborn , Male
3.
Pediatr Nephrol ; 34(8): 1325-1335, 2019 08.
Article in English | MEDLINE | ID: mdl-29961928

ABSTRACT

Hepatocyte nuclear factor-1ß (HNF-1ß) is an essential transcription factor that regulates the development and function of epithelia in the kidney, liver, pancreas, and genitourinary tract. Humans who carry HNF1B mutations develop heterogeneous renal abnormalities, including multicystic dysplastic kidneys, glomerulocystic kidney disease, renal agenesis, renal hypoplasia, and renal interstitial fibrosis. In the embryonic kidney, HNF-1ß is required for ureteric bud branching, initiation of nephrogenesis, and nephron segmentation. Ablation of mouse Hnf1b in nephron progenitors causes defective tubulogenesis, whereas later inactivation in elongating tubules leads to cyst formation due to downregulation of cystic disease genes, including Umod, Pkhd1, and Pkd2. In the adult kidney, HNF-1ß controls the expression of genes required for intrarenal metabolism and solute transport by tubular epithelial cells. Tubular abnormalities observed in HNF-1ß nephropathy include hyperuricemia with or without gout, hypokalemia, hypomagnesemia, and polyuria. Recent studies have identified novel post-transcriptional and post-translational regulatory mechanisms that control HNF-1ß expression and activity, including the miRNA cluster miR17 ∼ 92 and the interacting proteins PCBD1 and zyxin. Further understanding of the molecular mechanisms upstream and downstream of HNF-1ß may lead to the development of new therapeutic approaches in cystic kidney disease and other HNF1B-related renal diseases.


Subject(s)
Gene Expression Regulation, Developmental , Hepatocyte Nuclear Factor 1-beta/metabolism , Kidney Diseases, Cystic/genetics , Kidney Tubules/abnormalities , Urothelium/abnormalities , Down-Regulation , Hepatocyte Nuclear Factor 1-beta/genetics , Humans , Hydro-Lyases/metabolism , Kidney Diseases, Cystic/blood , Kidney Diseases, Cystic/pathology , Kidney Diseases, Cystic/urine , Kidney Tubules/pathology , MicroRNAs/metabolism , Mutation , RNA, Long Noncoding , Receptors, Cell Surface/genetics , TRPP Cation Channels/genetics , Uromodulin/genetics , Urothelium/pathology , Zyxin/metabolism
4.
J Obstet Gynaecol Res ; 44(12): 2181-2185, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30058238

ABSTRACT

We report a 32-week fetus conceived of consanguineous parentage which presented with severe early onset oligohydramnios and history of a similarly affected sibling in previous pregnancy. Ultrasonography and autopsy were inconclusive, prompting exome sequencing on fetal DNA. This resulted in identification of a homozygous novel 3' splice-site variation in intron 17 of the ACE gene (NM_000789.3:c.2642-1G>A), confirming diagnosis of autosomal recessive renal tubular dysgenesis, and facilitating prenatal diagnosis in subsequent pregnancy.


Subject(s)
Exome Sequencing , Kidney Tubules/abnormalities , Oligohydramnios , Peptidyl-Dipeptidase A/genetics , Urogenital Abnormalities/genetics , Consanguinity , Female , Humans , Infant, Newborn , Perinatal Death , Pregnancy
5.
Exp Cell Res ; 359(1): 138-144, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28778797

ABSTRACT

Renal ischemia reperfusion (IR) is a major cause of acute kidney injury (AKI) and no effective treatments have been established. Tisp40 is a transcription factor of the CREB/ATF family and involves in cell apoptosis, proliferation and differentiation, but its role in renal IR remains unknown. Here, we investigated the role of Tisp40 in renal IR injury. In vivo, Tisp40 knockout (KO) and wild-type (WT) mice were subjected to thirty minutes of bilateral renal ischemia and 48h reperfusion, the blood and kidneys were harvested for analysis. In vitro, Tisp40 overexpression and vector cells were subjected to hypoxia/reoxygenation (HR), the apoptosis rate and the expressions of related proteins were measured. Following IR, the expressions of Tisp40 protein, serum creatinine (sCr), blood urea nitrogen (BUN) and apoptosis of tubular cells were significantly increased in WT mice. However, Tisp40 deficiency significantly attenuated the increase of sCr, BUN and apoptosis of tubular cells. Following HR, apoptosis of tubular cells was increased in Tisp40 overexpression cells compared with vector cells. Mechanistically, Tisp40 promoted the expressions of C/EBP homologous protein (CHOP), Bax and Cleaved caspase3 and suppressed the expression of Bcl-2 in renal IR injury. In conclusion, Tisp40 aggravates tubular cells apoptosis in renal IR injury.


Subject(s)
Apoptosis , Cyclic AMP Response Element-Binding Protein/deficiency , Epithelial Cells/metabolism , Kidney Tubules/pathology , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Animals , Cyclic AMP Response Element-Binding Protein/metabolism , Epithelial Cells/pathology , Hypoxia/complications , Hypoxia/pathology , Kidney Tubules/abnormalities , Kidney Tubules/physiopathology , Mice, Inbred C57BL , Mice, Knockout , Oxygen , Reperfusion Injury/physiopathology , Transcription Factor CHOP/metabolism
6.
Am J Physiol Renal Physiol ; 312(6): F1141-F1157, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28100499

ABSTRACT

Gremlin1 (Grem1), an antagonist of bone morphogenetic proteins, plays a key role in embryogenesis. A highly specific temporospatial gradient of Grem1 and bone morphogenetic protein signaling is critical to normal lung, kidney, and limb development. Grem1 levels are increased in renal fibrotic conditions, including acute kidney injury, diabetic nephropathy, chronic allograft nephropathy, and immune glomerulonephritis. We demonstrate that a small number of grem1-/- whole body knockout mice on a mixed genetic background (8%) are viable, with a single, enlarged left kidney and grossly normal histology. The grem1-/- mice displayed mild renal dysfunction at 4 wk, which recovered by 16 wk. Tubular epithelial cell-specific targeted deletion of Grem1 (TEC-grem1-cKO) mice displayed a milder response in the acute injury and recovery phases of the folic acid model. Increases in indexes of kidney damage were smaller in TEC-grem1-cKO than wild-type mice. In the recovery phase of the folic acid model, associated with renal fibrosis, TEC-grem1-cKO mice displayed reduced histological damage and an attenuated fibrotic gene response compared with wild-type controls. Together, these data demonstrate that Grem1 expression in the tubular epithelial compartment plays a significant role in the fibrotic response to renal injury in vivo.


Subject(s)
Acute Kidney Injury/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Kidney Tubules/metabolism , Urogenital Abnormalities/metabolism , Acute Kidney Injury/chemically induced , Acute Kidney Injury/pathology , Acute Kidney Injury/prevention & control , Animals , Disease Models, Animal , Female , Fibrosis , Folic Acid , Gene Expression Regulation , Genetic Predisposition to Disease , Intercellular Signaling Peptides and Proteins/deficiency , Intercellular Signaling Peptides and Proteins/genetics , Kidney Tubules/abnormalities , Kidney Tubules/physiopathology , Mice, Inbred C57BL , Mice, Knockout , Organogenesis , Phenotype , Signal Transduction , Time Factors , Urogenital Abnormalities/genetics , Urogenital Abnormalities/pathology
7.
BMC Dev Biol ; 16(1): 30, 2016 08 31.
Article in English | MEDLINE | ID: mdl-27582005

ABSTRACT

BACKGROUND: Wnt11 is a member of the Wnt family of secreted signals controlling the early steps in ureteric bud (UB) branching. Due to the reported lethality of Wnt11 knockout embryos in utero, its role in later mammalian kidney organogenesis remains open. The presence of Wnt11 in the emerging tubular system suggests that it may have certain roles later in the development of the epithelial ductal system. RESULTS: The Wnt11 knockout allele was backcrossed with the C57Bl6 strain for several generations to address possible differences in penetrance of the kidney phenotypes. Strikingly, around one third of the null mice with this inbred background survived to the postnatal stages. Many of them also reached adulthood, but urine and plasma analyses pointed out to compromised kidney function. Consistent with these data the tubules of the C57Bl6 Wnt11 (-/-) mice appeared to be enlarged, and the optical projection tomography indicated changes in tubular convolution. Moreover, the C57Bl6 Wnt11 (-/-) mice developed secondary glomerular cysts not observed in the controls. The failure of Wnt11 signaling reduced the expression of several genes implicated in kidney development, such as Wnt9b, Six2, Foxd1 and Hox10. Also Dvl2, an important PCP pathway component, was downregulated by more than 90 % due to Wnt11 deficiency in both the E16.5 and NB kidneys. Since all these genes take part in the control of UB, nephron and stromal progenitor cell differentiation, their disrupted expression may contribute to the observed anomalies in the kidney tubular system caused by Wnt11 deficiency. CONCLUSIONS: The Wnt11 signal has roles at the later stages of kidney development, namely in coordinating the development of the tubular system. The C57Bl6 Wnt11 (-/-) mouse generated here provides a model for studying the mechanisms behind tubular anomalies and glomerular cyst formation.


Subject(s)
Kidney Glomerulus/abnormalities , Kidney Tubules/abnormalities , Wnt Proteins/genetics , Wnt Proteins/metabolism , Animals , Cell Differentiation , Embryo, Mammalian/abnormalities , Embryo, Mammalian/metabolism , Embryonic Development , Gene Expression Regulation, Developmental , Kidney Glomerulus/embryology , Kidney Tubules/embryology , Mice , Mice, Knockout , Signal Transduction
8.
PLoS Genet ; 12(2): e1005785, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26859289

ABSTRACT

Rho family GTPases act as molecular switches regulating actin cytoskeleton dynamics. Attenuation of their signaling capacity is provided by GTPase-activating proteins (GAPs), including p190A, that promote the intrinsic GTPase activity of Rho proteins. In the current study we have performed a small-scale ENU mutagenesis screen and identified a novel loss of function allele of the p190A gene Arhgap35, which introduces a Leu1396 to Gln substitution in the GAP domain. This results in decreased GAP activity for the prototypical Rho-family members, RhoA and Rac1, likely due to disrupted ordering of the Rho binding surface. Consequently, Arhgap35-deficient animals exhibit hypoplastic and glomerulocystic kidneys. Investigation into the cystic phenotype shows that p190A is required for appropriate primary cilium formation in renal nephrons. P190A specifically localizes to the base of the cilia to permit axoneme elongation, which requires a functional GAP domain. Pharmacological manipulations further reveal that inhibition of either Rho kinase (ROCK) or F-actin polymerization is able to rescue the ciliogenesis defects observed upon loss of p190A activity. We propose a model in which p190A acts as a modulator of Rho GTPases in a localized area around the cilia to permit the dynamic actin rearrangement required for cilia elongation. Together, our results establish an unexpected link between Rho GTPase regulation, ciliogenesis and glomerulocystic kidney disease.


Subject(s)
Cilia/metabolism , GTPase-Activating Proteins/genetics , Kidney Diseases, Cystic/genetics , Kidney Glomerulus/pathology , Organogenesis , Point Mutation/genetics , Repressor Proteins/genetics , Actins/metabolism , Alleles , Amino Acid Sequence , Amino Acid Substitution , Animals , Cytoskeleton/metabolism , Embryo, Mammalian/cytology , Ethylnitrosourea , Female , Fibroblasts/metabolism , GTPase-Activating Proteins/chemistry , GTPase-Activating Proteins/metabolism , Kidney Diseases, Cystic/pathology , Kidney Glomerulus/metabolism , Kidney Tubules/abnormalities , Kidney Tubules/pathology , Male , Mice, Inbred C3H , Mice, Inbred C57BL , Molecular Sequence Data , Neural Tube Defects/pathology , Phenotype , Protein Structure, Tertiary , Repressor Proteins/chemistry , Repressor Proteins/metabolism , Reproducibility of Results
9.
Pediatr Nephrol ; 29(7): 1221-30, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24477978

ABSTRACT

BACKGROUND: Fetuses exposed to angiotensin-converting enzyme inhibitors or angiotensin receptor antagonists during the second and/or third trimesters of gestation are at high risk of developing severe complications. They consist in fetal hypotension, and anuria/oligohydramnios leading to Potter sequence, frequently associated with hypocalvaria. Most fetuses die during the pre- or postnatal period, whereas others recover normal or subnormal renal function. However, the secondary occurrence of renal failure or hypertension has been reported in children after apparent complete recovery. METHODS: In this context, we analyzed renal lesions in 14 fetus/neonates who died soon after exposure to renin-angiotensin-system (RAS) blockers. Our objective was to determine the causes for the persistence or the secondary occurrence of renal complications reported in some of the survivors. RESULTS: As previously described, renal tubular dysgenesis is usually observed. Additional lesions, such as thickening of the muscular wall of arterioles and interlobular arteries, glomerular cysts, and interstitial fibrosis, develop early during fetal life. CONCLUSION: We suggest that renal lesions that develop before birth may persist after withdrawal of the causative drugs and normalization of blood and renal perfusion pressure. Their persistence could explain the severe long-term outcome of some of these patients. Long-term study of children exposed to RAS blockers during fetal life is strongly recommended.


Subject(s)
Angiotensin Receptor Antagonists/adverse effects , Angiotensin-Converting Enzyme Inhibitors/adverse effects , Fetal Diseases/chemically induced , Kidney Tubules/abnormalities , Kidney/drug effects , Abnormalities, Drug-Induced , Humans , Immunohistochemistry , Infant, Newborn , Kidney/chemistry , Kidney/pathology , Renin/analysis
10.
Anat Rec (Hoboken) ; 296(12): 1936-42, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24142560

ABSTRACT

Congenital anomalies of the kidney and urinary tract occur at a frequency of 1 in 500 live births in humans. Mutant mice null for Dlg1 (Dlg1(-/-) mice), a membrane-associated guanylate kinase containing PDZ domains, exhibit various urogenital malformations, including hypoplasia of the kidney and ureter, megaureter, hydronephrosis, and aplasia of the seminal vesicle and the vagina. The common nephric duct (CND) is a distal part of the Wolffian duct between the ureteric branch and the opening to the urogenital sinus, and normally disappears by embryonic day (E) 12.5 by a downward shift of the ureteric branch. Although retardation of the disappearance of the CND is apparent during urogenital development in Dlg1(-/-) mice, its pathogenesis and prognosis are unclear. In the present study, we found a decrease in apoptotic cells in the CND epithelium in Dlg1(-/-) mice at E11.5. Cell proliferation did not change. Additionally, histological observation of the development of the ureteral orifice indicated that the CND remained at E15.5 and was widely open to the vesical lumen in Dlg1(-/-) mice, in contrast to the complete disappearance of the CND and a narrow ureteric orifice in control mice. The dilatation of the vesicoureteral junction remained at E18.5. Opening of the vesicoureteral junction is known to cause vesicoureteral reflux and subsequent megaureter and hydronephrosis. Therefore, our present observation demonstrates that lack of the Dlg1 gene induces a decrease in apoptotic epithelial cell death and the persistence of the CND, which result in a dysfunctional vesicoureteral junction and cause megaureter or hydronephrosis through vesicoureteral reflux.


Subject(s)
Apoptosis , Embryo, Mammalian/pathology , Kidney Tubules/embryology , Nerve Tissue Proteins/physiology , Ureter/embryology , Vesico-Ureteral Reflux/pathology , Wolffian Ducts/pathology , Animals , Bromodeoxyuridine , Discs Large Homolog 1 Protein , Female , Kidney Tubules/abnormalities , Mice , Mice, Knockout , SAP90-PSD95 Associated Proteins , Ureter/abnormalities , Wolffian Ducts/abnormalities , Wolffian Ducts/embryology
11.
PLoS One ; 8(6): e66164, 2013.
Article in English | MEDLINE | ID: mdl-23799078

ABSTRACT

Genetic evidence links mutations in the LRRK2 gene with an increased risk of Parkinson's disease, for which no neuroprotective or neurorestorative therapies currently exist. While the role of LRRK2 in normal cellular function has yet to be fully described, evidence suggests involvement with immune and kidney functions. A comparative study of LRRK2-deficient and wild type rats investigated the influence that this gene has on the phenotype of these rats. Significant weight gain in the LRRK2 null rats was observed and was accompanied by significant increases in insulin and insulin-like growth factors. Additionally, LRRK2-deficient rats displayed kidney morphological and histopathological alterations in the renal tubule epithelial cells of all animals assessed. These perturbations in renal morphology were accompanied by significant decreases of lipocalin-2, in both the urine and plasma of knockout animals. Significant alterations in the cellular composition of the spleen between LRRK2 knockout and wild type animals were identified by immunophenotyping and were associated with subtle differences in response to dual infection with rat-adapted influenza virus (RAIV) and Streptococcus pneumoniae. Ontological pathway analysis of LRRK2 across metabolic and kidney processes and pathological categories suggested that the thioredoxin network may play a role in perturbing these organ systems. The phenotype of the LRRK2 null rat is suggestive of a complex biology influencing metabolism, immune function and kidney homeostasis. These data need to be extended to better understand the role of the kinase domain or other biological functions of the gene to better inform the development of pharmacological inhibitors.


Subject(s)
Homeostasis , Immunity, Humoral , Kidney Tubules/pathology , Protein Serine-Threonine Kinases/deficiency , Animals , Antibodies, Viral/blood , Blood Proteins/metabolism , Epithelial Cells/pathology , Gene Knockout Techniques , Gene Regulatory Networks , Immunophenotyping , Kidney Tubules/abnormalities , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Male , Orthomyxoviridae/immunology , Orthomyxoviridae Infections/immunology , Pneumonia, Pneumococcal/immunology , Protein Serine-Threonine Kinases/genetics , Rats , Streptococcus pneumoniae/immunology , Weight Gain/genetics
12.
Biochem Biophys Res Commun ; 425(2): 212-8, 2012 Aug 24.
Article in English | MEDLINE | ID: mdl-22835934

ABSTRACT

Autosomal dominant polycystic kidney disease (ADPKD), the most common hereditary disease affecting the kidneys, is caused in 85% of cases by mutations in the PKD1 gene. The protein encoded by this gene, polycystin-1, is a renal epithelial cell membrane mechanoreceptor, sensing morphogenetic cues in the extracellular environment, which regulate the tissue architecture and differentiation. However, how such mutations result in the formation of cysts is still unclear. We performed a precise characterization of mesenchymal differentiation using PAX2, WNT4 and WT1 as a marker, which revealed that impairment of the differentiation process preceded the development of cysts in Pkd1(-/-) mice. We performed an in vitro organ culture and found that progesterone and a derivative thereof facilitated mesenchymal differentiation, and partially prevented the formation of cysts in Pkd1(-/-) kidneys. An injection of progesterone or this derivative into the intraperitoneal space of pregnant females also improved the survival of Pkd1(-/-) embryos. Our findings suggest that compounds which enhance mesenchymal differentiation in the nephrogenesis might be useful for the therapeutic approach to prevent the formation of cysts in ADPKD patients.


Subject(s)
Cell Differentiation/drug effects , Cysts/prevention & control , Kidney Tubules/abnormalities , Mesoderm/drug effects , Polycystic Kidney, Autosomal Dominant/prevention & control , Progesterone/administration & dosage , Animals , Cysts/embryology , Cysts/genetics , Dilatation, Pathologic/embryology , Dilatation, Pathologic/prevention & control , Female , Mesoderm/cytology , Mice , Mice, Mutant Strains , Polycystic Kidney, Autosomal Dominant/embryology , Polycystic Kidney, Autosomal Dominant/genetics , Pregnancy , TRPP Cation Channels/genetics
13.
Autophagy ; 8(5): 826-37, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22617445

ABSTRACT

Autophagy is responsible for the degradation of protein aggregates and damaged organelles. Several studies have reported increased autophagic activity in tubular cells after kidney injury. Here, we examine the role of tubular cell autophagy in vivo under both physiological conditions and stress using two different tubular-specific Atg5-knockout mouse models. While Atg5 deletion in distal tubule cells does not cause a significant alteration in kidney function, deleting Atg5 in both distal and proximal tubule cells results in impaired kidney function. Already under physiological conditions, Atg5-null tubule cells display a significant accumulation of p62 and oxidative stress markers. Strikingly, tubular cell Atg5-deficiency dramatically sensitizes the kidneys to ischemic injury, resulting in impaired kidney function, accumulation of damaged mitochondria as well as increased tubular cell apoptosis and proliferation, highlighting the critical role that autophagy plays in maintaining tubular cell integrity during stress conditions.


Subject(s)
Aging/pathology , Autophagy , Kidney Tubules/pathology , Reperfusion Injury/pathology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Autophagy-Related Protein 5 , Biomarkers/metabolism , Creatinine/blood , Embryo, Mammalian/abnormalities , Embryo, Mammalian/pathology , Gene Deletion , Gene Knockout Techniques , Heat-Shock Proteins/metabolism , Kidney Tubules/abnormalities , Kidney Tubules/metabolism , Kidney Tubules/ultrastructure , Mice , Microtubule-Associated Proteins/deficiency , Microtubule-Associated Proteins/metabolism , Organ Specificity , Oxidative Stress , Reperfusion Injury/blood , Sequestosome-1 Protein , Time Factors
14.
Nephrology (Carlton) ; 16(5): 495-501, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21265929

ABSTRACT

AIM: The relationship between abnormalities of tubular architecture and tubulointerstitial nephritis antigen (TIN-ag) in juvenile nephronophthisis (J-NPH) was evaluated. METHODS: Sixteen J-NPH patients were examined. Nephrocystin-1, TIN-ag, type IV collagen, Fas antigen and the C5b-9 complement complex were stained by immunohistochemical methods. RESULTS: Renal tubules of patients with J-NPH showed morphological abnormalities of tubular basement membranes (TBM) and frequent apoptosis of tubular epithelial cells. Additionally, the C5b-9 complement complex was deposited within the TBM in the absence of immunoglobulin deposition, suggesting complement-dependent TBM injury. Localization of TIN-ag in the TBM of J-NPH patients disclosed a partial defect or discontinuity in 14 of the 16 patients, while type IV collagen immunoreactivity was relatively preserved. These findings suggest that tubulogenesis is disturbed during nephronogenesis in J-NPH patients because of a defect in nephrocystin, an NPHP gene product. TBM defects induce further morphological abnormalities such as cystic dilation of tubules; as tubular function impairment advances, the incomplete tubules may be injured by C5b-9 complement complexes, followed by apoptotic cell death. CONCLUSION: TIN-ag, which is important in early nephrogenesis, lacks normal activity, and vulnerable and incomplete tubules with deficient TIN-ag expression are formed. Removal of these defective tubules by apoptosis combined with the C5b-9 complement complex could be the primary reason for progression to end-stage renal disease in J-NPH patients.


Subject(s)
Cell Adhesion Molecules/physiology , Kidney Diseases, Cystic/congenital , Kidney Failure, Chronic/etiology , Kidney Tubules/abnormalities , Basement Membrane/pathology , Cell Adhesion Molecules/analysis , Collagen Type IV/analysis , Complement Membrane Attack Complex/analysis , Humans , Kidney Diseases, Cystic/complications , fas Receptor/analysis
15.
Am J Med Genet A ; 152A(9): 2318-21, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20683989

ABSTRACT

Peroxisomal biogenesis disorders (PBD) are groups of inherited neurometabolic disorders caused by defects in PEX genes. We report on a female infant, born to a consanguineous parents (first degree cousins), who presented with inactivity, poor sucking, and hypotonia early in the neonatal period. She had subtle dysmorphic features. Liver function tests were impaired with raised liver enzymes, conjugated and unconjugated hyperbilirubinemia. CT of the brain showed diffuse bilateral changes. She developed seizures with an abnormal EEG. Plasma very long chain fatty acid analysis showed high C26:0 levels and increasedC26:0/C22:0 and C24:0/C22:0 ratios, which is consistent with a PBD. Studies in fibroblasts including plasmalogen biosynthesis, peroxisomal fatty acid alfa and beta oxidation confirmed the diagnosis of PBD. Immunofluoresence microscopy revealed the absence of peroxisomes in fibroblasts. The patient was assigned to the PEX19 complementation group. Subsequent mutation analysis of the PEX19 gene revealed homozygosity for a c.320delA frameshift mutation. The patient had a stormy course with multiple admissions to the pediatric intensive care unit with pneumonia, liver impairment, sepsis, and epilepsy. At 1 year of age she developed metabolic acidosis with normal anion gap, proteinuria, aminoaciduria, and glucosuria consistent with a renal tubular defect. Abdominal ultrasound showed multiple gallstones. Other causes of gallstones like haemoglobinopathy were excluded. So far, only two siblings had been reported with mutations in the PEX19 gene. Our patient showed a previously unrecognized association of gallstones and a renal tubular defect with a PBD.


Subject(s)
Frameshift Mutation , Membrane Proteins/genetics , Peroxisomal Disorders/genetics , Female , Gallstones , Humans , Infant , Kidney Tubules/abnormalities , Peroxisomal Disorders/diagnosis , Peroxisomal Disorders/pathology , Phenotype
17.
Pediatr Nephrol ; 25(4): 687-97, 2010 Apr.
Article in English | MEDLINE | ID: mdl-19844747

ABSTRACT

Congenital obstructive nephropathy accounts for the greatest fraction of chronic kidney disease in children. Genetic and nongenetic factors responsible for the lesions are largely unidentified, and attention has been focused on minimizing obstructive renal injury and optimizing long-term outcomes. The cellular and molecular events responsible for obstructive injury to the developing kidney have been elucidated from animal models. These have revealed nephron loss through cellular phenotypic transition and cell death, leading to the formation of atubular glomeruli and tubular atrophy. Altered renal expression of growth factors and cytokines, including angiotensin, transforming growth factor-beta, and adhesion molecules, modulate cell death by apoptosis or phenotypic transition of glomerular, tubular, and vascular cells. Mediators of cellular injury include hypoxia, ischemia, and reactive oxygen species, while fibroblasts undergo myofibroblast transformation with increased deposition of extracellular matrix. Progression of the lesions involves interstitial inflammation and interstitial fibrosis, both of which impair growth of the obstructed kidney and result in compensatory growth of the contralateral kidney. The long-term outcome depends on timing and severity of the obstruction and its relief, minimizing ongoing injury, and enhancing remodeling. Advances will depend on new biomarkers to evaluate the severity of obstruction, to determine therapy, and to follow the evolution of lesions.


Subject(s)
Hydronephrosis/pathology , Kidney/pathology , Ureteral Obstruction/pathology , Animals , Animals, Newborn , Apoptosis , Biomarkers/metabolism , Disease Models, Animal , Disease Progression , Humans , Hydronephrosis/congenital , Hydronephrosis/physiopathology , Kidney/metabolism , Kidney/physiopathology , Kidney Function Tests , Kidney Tubules/abnormalities , Kidney Tubules/pathology , Nephrons/abnormalities , Nephrons/pathology , Recovery of Function , Ureteral Obstruction/congenital , Ureteral Obstruction/physiopathology
18.
Kidney Int ; 77(5): 400-6, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19924102

ABSTRACT

Autosomal recessive renal tubular dysgenesis (RTD) is a severe disorder of renal tubular development characterized by early onset and persistent fetal anuria leading to oligohydramnios and the Potter sequence. At birth, blood pressure is dramatically low and perinatal death occurs in most cases. Skull ossification defects are frequently associated with RTD. The disease is genetically heterogeneous and linked to mutations in the genes encoding any of the components of the renin-angiotensin system (RAS). An intense stimulation of renin production is noted in the kidneys of patients with mutations in the genes encoding angiotensinogen, angiotensin-converting enzyme, or AT1 receptor, whereas absence or increased renin production is associated with REN defects depending on the type of mutation. The severity of the disease underlines the importance of a functional RAS in the maintenance of blood pressure and renal blood flow during fetal life. The absence or poor development of proximal tubules, as well as renal vascular changes, may be attributable to renal hypoperfusion rather than to a morphogenic property of the RAS. The less severe phenotype in mice devoid of RAS may be linked to differences between mice and humans in the time of nephrogenesis and maturation of the RAS. The identification of the disease on the basis of precise clinical and histological analyses and the characterization of the genetic defects allow genetic counseling and early prenatal diagnosis.


Subject(s)
Kidney Tubules/abnormalities , Renin-Angiotensin System/physiology , Animals , Anuria/etiology , Embryonic Development/physiology , Female , Fetal Death/etiology , Genes, Recessive , Genetic Heterogeneity , Humans , Infant, Newborn , Kidney Tubules/embryology , Kidney Tubules/pathology , Mice , Mutation , Oligohydramnios/etiology , Pregnancy , Receptor, Angiotensin, Type 1/genetics , Renin/genetics , Renin-Angiotensin System/genetics , Skull/pathology
20.
Isr Med Assoc J ; 11(1): 6-10, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19344005

ABSTRACT

BACKGROUND: Renal tubular dysgenesis is a rare lethal kidney abnormality clinically manifested by olighydramnios, anuria and respiratory distress. Most of the information on this entity is provided by case reports and short series. OBJECTIVES: To evaluate the incidence and comparative frequency of clinical manifestations in different etiologic-pathogenic variants of RTD in Israel and in summarized published data. METHODS: Stillborn and neonatal autopsy material from nine medical centers in northern and central Israel was studied. Information concerning pregnancy, labor and postnatal status and autopsy findings of cases with histologically, histochemically and immunohistochemically confirmed RTD were obtained from corresponding reports and from published material. RESULTS: From the 1538 autopsies of fetuses (2 20 weeks gestation) and neonates that were performed between 1976 and 2007 we identified 12 cases of RTD (0.78%). Abnormality occurred more often (1.4%) in the Upper and Western Galilee than in Israel as a whole. CONCLUSIONS: Our study and a review of the literature showed that the autosomal recessive variant of RTD was more frequent than twin-twin transfusion-induced. Most symptoms were similar in all variants of RTD, but their frequency was different in each of them.


Subject(s)
Anuria/etiology , Congenital Abnormalities/diagnosis , Congenital Abnormalities/physiopathology , Kidney Tubules/abnormalities , Oligohydramnios/etiology , Respiratory Distress Syndrome, Newborn/etiology , Anuria/physiopathology , Autopsy , Congenital Abnormalities/epidemiology , Female , Humans , Incidence , Infant, Newborn , Israel/epidemiology , Male , Oligohydramnios/physiopathology , Pregnancy , Respiratory Distress Syndrome, Newborn/physiopathology , Risk Factors
SELECTION OF CITATIONS
SEARCH DETAIL