990 resultados para Kidney development


Relevância:

70.00% 70.00%

Publicador:

Resumo:

The receptor tyrosine kinase RET functions during the development of the kidney and the enteric nervous system, yet no ligand has been identified to date. This report demonstrates that the glial cell line-derived neurotrophic factor (GDNF) activates RET, as measured by tyrosine phosphorylation of the intracellular catalytic domain. GDNF also binds RET with a dissociation constant of 8 nM, and 125I-labeled GDNF can be coimmunoprecipitated with anti-RET antibodies. In addition, exogenous GDNF stimulates both branching and proliferation of embryonic kidneys in organ culture, whereas neutralizing antibodies against GDNF inhibit branching morphogenesis. These data indicate that RET and GDNF are components of a common signaling pathway and point to a role for GDNF in kidney development.

Relevância:

60.00% 60.00%

Publicador:

Resumo:

Integrin-linked kinase (ILK) and p38MAPK are protein kinases that transduce extracellular signals regulating cell migration and actin cytoskeletal organization. ILK-dependent regulation of p38MAPK is critical for mammalian kidney development and in smooth muscle cell migration, however, specific p38 isoforms has not been previously examined in ILK-regulated responses. Signaling by ILK and p38MAPK is often dysregulated in bladder cancer, and here we report a strong positive correlation between protein levels of ILK and p38β, which is the predominant isoform found in bladder cancer cells, as well as in patient-matched normal bladder and tumor samples. Knockdown by RNA interference of either p38β or ILK disrupts serum-induced, Rac1-dependent migration and actin cytoskeletal organization in bladder cancer cells. Surprisingly, ILK knockdown causes the selective reduction in p38β cellular protein level, without inhibiting p38β messenger RNA (mRNA) expression. The loss of p38β protein in ILK-depleted cells is partially rescued by the 26S proteasomal inhibitor MG132. Using co-precipitation and bimolecular fluorescent complementation assays, we find that ILK selectively forms cytoplasmic complexes with p38β. In situ proximity ligation assays further demonstrate that serum-stimulated assembly of endogenous ILK–p38β complexes is sensitive to QLT-0267, a small molecule ILK kinase inhibitor. Finally, inhibition of ILK reduces the amplitude and period of serum-induced activation of heat shock protein 27 (Hsp27), a target of p38β implicated in actin cytoskeletal reorganization. Our work identifies Hsp27 as a novel target of ILK–p38β signaling complexes, playing a key role in bladder cancer cell migration.

Relevância:

60.00% 60.00%

Publicador:

Resumo:

Multicentric carpotarsal osteolysis (MCTO) is a rare skeletal dysplasia characterized by aggressive osteolysis, particularly affecting the carpal and tarsal bones, and is frequently associated with progressive renal failure. Using exome capture and next-generation sequencing in five unrelated simplex cases of MCTO, we identified previously unreported missense mutations clustering within a 51 base pair region of the single exon of MAFB, validated by Sanger sequencing. A further six unrelated simplex cases with MCTO were also heterozygous for previously unreported mutations within this same region, as were affected members of two families with autosomal-dominant MCTO. MAFB encodes a transcription factor that negatively regulates RANKL-induced osteoclastogenesis and is essential for normal renal development. Identification of this gene paves the way for development of novel therapeutic approaches for this crippling disease and provides insight into normal bone and kidney development.

Relevância:

60.00% 60.00%

Publicador:

Resumo:

Glial cell line-derived neurotrophic factor (GDNF) family ligands: GDNF, neurturin, persephin and artemin, signal through a receptor tyrosine kinase Ret by binding first to a co-receptor (GFRα1-4) that is attached to the plasma membrane. The GDNF family factors can support the survival of various peripheral and central neuronal populations and have important functions also outside the nervous system, especially in kidney development. Activating mutations in the RET gene cause tumours in neuroendocrine cells, whereas inactivating mutations in RET are found in patients with Hirschsprung s disease (HSCR) characterized by loss of ganglionic cells along the intestine. The aim of this study was to examine the in vivo functions of neurturin receptor GFRα2 and persephin receptor GFRα4 using knockout (KO) mice. Mice lacking GFRα2 grow poorly after weaning and have deficits in parasympathetic and enteric innervation. This study shows that impaired secretion of the salivary glands and exocrine pancreas contribute to growth retardation in GFRα2-KO mice. These mice have a reduced number of intrapancreatic neurons and decreased cholinergic innervation of the exocrine pancreas as well as reduced excitatory fibres in the myenteric plexus of the small intestine. This study also demonstrates that GFRα2-mediated Ret signalling is required for target innervation and maintenance of soma size of sympathetic cholinergic neurons and sensory nociceptive IB4-binding neurons. Furthermore, lack of GFRα2 in mice results in deficient perception of temperatures above and below thermoneutrality and in attenuated inflammatory pain response. GFRα4 is co-expressed with Ret predominantly in calcitonin-producing thyroid C-cells in the mouse. In this study GFRα4-deficient mice were generated. The mice show no gross developmental deficits and have a normal number of C-cells. However, young but not adult mice lacking GFRα4 have a lower production of calcitonin in thyroid tissue and consequently, an increased bone formation rate. Thus, GFRα4/Ret signalling may regulate calcitonin production. In conclusion, this study reveals that GFRα2/Ret signalling is crucial for the development and function of specific components of the peripheral nervous system and that GFRα4-mediated Ret signalling is required for controlling transmitter synthesis in thyroid C-cells.

Relevância:

60.00% 60.00%

Publicador:

Resumo:

Neurotrophic factors play essential role in the development and functioning of the nervous system and other organs. Glial cell line-Derived Neurotrophic Factor (GDNF) family ligands (GFLs) are of particular interest because they promote the survival of dopaminergic neurons in vitro, in Parkinson s disease animal models and in patients. GDNF is also a potent survival factor for the central motoneurons and thus is considered as a potential lead for the treatment of amyotrophic lateral sclerosis. The survival promoting receptor complex for GFLs consists of a ligand-specific co-receptor, GFRα and a signal transducing module, receptor tyrosine kinase RET. At least GDNF and persephin, a GFL, have established functions outside central nervous system. GDNF is crucial for enteric nervous system and kidney development as well as for spermatogenesis. Persephin controls calcitonin secretion. Communication between cells often occurs in the extracellular matrix (ECM), a meshwork, which is secreted and deposited by the cells and is mainly composed of fibrillar proteins and polymerized sugars. We evaluated the relationship between GFLs and extracellular matrix components and demonstrated that three GFLs - GDNF, neurturin and artemin bind heparan sulfates with nanomolar affinities. The fourth member of the family - persephin binds these polysaccharides thousand times less tightly. GDNF, neurturin and artemin also bind with high affinity to heparan sulfate proteoglycan (HSPG) isolated from the nervous system, syndecan-3. GDNF signals through HSPGs, evoking Src family kinase activation. This signaling induces cell spreading, hippocampal neurite outgrowth in vitro and cellular migration. Specifically, GDNF signaling through syndecan-3 is important for embryonic cortical neuron migration. Syndecan-3-deficient mice, similarly to mice lacking GDNF, have less GABAergic neurons in their cortex, as compared to the wild-type mice. This fact provides indirect evidence that GDNF interaction with syndecan-3 is important for cortical brain development. Noteworthy, in non-neuronal tissues GFLs may signal via other syndecans. We also present the structural model for a GDNF co-receptor, GFRα1. The X-ray structure of the GFRα1 domain 3 was solved with 1.8 Å resolution, revealing a new protein fold. Later we also solved the structure of the truncated GFRα1 in the complex with GDNF and this model was confirmed by site-directed mutagenesis. In summary, our work contributed to the structural characterization of GFRα-based receptor complex and revealed a new receptor for GDNF, neurturin and artemin the HSPG syndecan-3. This information is critically important for the development of GFRα/RET agonists for the treatment of neurodegenerative diseases.

Relevância:

60.00% 60.00%

Publicador:

Resumo:

Congenital nephrotic syndrome of the Finnish type (NPHS1) is an autosomal recessive disease which is highly enriched in the Finnish population. It is caused by mutations in the NPHS1 gene encoding for nephrin, which is a major component of the glomerular filtration barrier in the kidney. Patients with NPHS1 have heavy proteinuria and nephrotic syndrome (NS) from birth and develop renal fibrosis in early childhood. Renal transplantation (TX) is the only curative treatment for NPHS1. These patients form the largest group of pediatric kidney transplant children in our country. The NPHS1 kidneys are removed in infancy and they serve as an excellent human material for studies of the pathophysiology of proteinuric kidney diseases. Sustained proteinuria is a major factor leading to end-stage renal failure and understanding this process is crucial for nephrology. In this study we investigated the glomerular and tubulointerstitial changes that occur in the NPHS1 kidneys during infancy as well as the expression of nephrin in non-renal tissues. We also studied the pathology and management of recurrent proteinuria in kidney grafts transplanted to NPHS1 children. Severe renal lesions evolved in patients with NPHS1 during the first months of life. Glomerular sclerosis developed through progressive mesangial sclerosis, and capillary obliteration was an early consequence of this process. Shrinkage of the glomerular tuft was common, whereas occlusion of tubular opening or protrusion of the glomerular tuft into subepithelial space or through the Bowman's capsule were not detected. Few inflammatory cells were detected in the mesangial area. The glomerular epithelial cells (podocytes) showed severe ultrastructural changes and hypertrophy. Podocyte proliferation and apoptosis were rare, but moderate amounts of podocytes were detached and ended up in the urine. The results showed that endocapillary lesions not extracapillary lesions, as generally believed were important for the sclerotic process in the NPHS1 glomeruli. In the tubulointerstitium, severe lesions developed in NPHS1 kidneys during infancy. Despite heavy proteinuria, tubular epithelial cells (TECs) did not show transition into myofibroblasts. The most abundant chemokines in NPHS1 tissue were neutrophil activating protein-2 (NAP-2), macrophage inhibiting factor (MIF), and monocyte chemoattractant protein-1 (MCP-1). Interstitial inflammation and fibrosis were first detected in the paraglomerular areas and the most abundant inflammatory cells were monocytes/macrophages. Arteries and arterioles showed intimal hypertrophy, but the pericapillary microvasculature remained quite normal. However, excessive oxidative stress was evident in NPHS1 kidneys. The results indicated that TECs were relatively resistant to the heavy tubular protein load. Nephrin was at first thought to be podocyte specific, but some studies especially in experimental animals have suggested that nephrin might also be expressed in non-renal tissues such as pancreas and central nervous system. The knowledge of nephrin biology is important for the evaluation of nephrin related diseases. In our study, no significant amounts of nephrin protein or mRNA were detected in non-renal tissues of man and pig as studied by immunohistochemistry and in situ hybridization. The phenotype analysis of NPHS1 children, who totally lack nephrin, revealed no marked impairment in the neurological, testicular, or pancreatic function speaking against the idea that nephrin would play an important functional role outside the kidney. The NPHS1 kidneys do not express nephrin and antibodies against this major glomerular filter protein have been observed in NPHS1 children after renal TX most likely as an immune reaction against a novel antigen. These antibodies have been associated with the development of recurrent NS in the kidney graft of NPHS1 patients. In our study, a third of the NPHS1 patients homozygous for Fin-Major mutation developed recurrent NS in the transplanted graft. Re-transplantations were performed to patients who lost their graft due to recurrent NS and heavy proteinuria immediately developed in all cases. While 73% of the patients had detectable serum anti-nephrin antibodies, the kidney biopsy findings were minimal. Introduction of plasma exchange (PE) to the treatment of recurrent nephroses increased the remission rate from 54% to 89%. If remission was achieved, recurrent NS did not significantly deteriorate the long term graft function. In conclusion, the results show that the lack of nephrin in podocyte slit diaphragm in NPHS1 kidneys induces progressive mesangial expansion and glomerular capillary obliteration and inflicts interstitial fibrosis, inflammation, and oxidative stress with surprisingly little involvement of the TECs in this process. Nephrin appears to have no clinical significance outside the kidney. Development of antibodies against nephrin seems to be a major cause of recurrent NS in kidney grafts of NPHS1 patients and combined use of PE and cyclophosphamide markedly improved remission rates.

Relevância:

60.00% 60.00%

Publicador:

Resumo:

The Wilms tumor suppressor WT1 encodes a zinc finger transcription factor that is expressed in glomerular podocytes during a narrow window in kidney development. By immunoprecipitation and protein microsequencing analysis, we have identified a major cellular protein associated with endogenous WT1 to be the inducible chaperone Hsp70. WT1 and Hsp70 are physically associated in embryonic rat kidney cells, in primary Wilms tumor specimens and in cultured cells with inducible expression of WT1. Colocalization of WT1 and Hsp70 is evident within podocytes of the developing kidney, and Hsp70 is recruited to the characteristic subnuclear clusters that contain WT1. The amino-terminal transactivation domain of WT1 is required for binding to Hsp70, and expression of that domain itself is sufficient to induce expression of Hsp70 through the heat shock element (HSE). Substitution of a heterologous Hsp70-binding domain derived from human DNAJ is sufficient to restore the functional properties of a WT1 protein with an amino-terminal deletion, an effect that is abrogated by a point mutation in DNAJ that reduces binding to Hsp70. These observations indicate that Hsp70 is an important cofactor for the function of WT1, and suggest a potential role for this chaperone during kidney differentiation.

Relevância:

60.00% 60.00%

Publicador:

Resumo:

Introduction : La Vitamine A (rétinol, ROL) et son métabolite l’acide rétinoïque (AR) sont essentielles pour l’embryogénèse. L’excès comme l’insuffisance d’AR sont nocives. L’AR est régularisé dans l’embryon par des gènes spécifiques (ALDH, CRABP, CYP). Hypothèse : Les grandes variations d’AR dans le plasma des adultes normaux, nous ont orienté à mesurer les rétinoïdes (ROL et RA) dans le sang de cordon ombilical, pour évaluer des corrélations avec des polymorphismes des gènes impliquées dans le métabolisme de l’AR et le développement rénal-(RALDH2, CRABP2, CYP26A1; B1). Vérifier pour des corrélations entre ces rétinoïdes et/ou avec la taille de reins à la naissance. Méthodes : Extraction du ROL et RA du sang de cordon ombilical de 145 enfants et analyse par HPLC. Le volume des reins a été mesuré par ultrasonographie et l’ADN génomique leucocytaire extrait (FlexiGene DNA-Kit). 10 échantillons d’ADN ont été exclus (qualité). Les htSNP : ALDH1A2, CRABP2, CYP26A1;B1 du génome humain (HapMap) ont été séquencés et génotypés (Sequenom iPlex PCR).Des testes bio-statistiques des fréquences génotypiques et alléliques ont été effectués (Single-Locus, χ2, Kruskal-Wallis, Allelic-Exact).Des corrélations (ROL, RA, SNPs, V-reins) ont été analysés (Kendall-tau /Oakes). Résultats : La Δ RA (0.07-550.27 nmol/l) non corrélé avec la Δ ROL (51.39-3892.70 nmol/l). Il n’y a pas d’association ROL ou RA avec les volumes des reins ou avec les SNPs/ CYP21A1;B1. Corrélations trouvées : 1. (p=0.035), polymorphisme génétique ALDH1A2-SNP (rs12591551:A/C) hétérozygote/CA, (25enfants, 19%) avec moyennes d’AR (62.21nmol/l). 2. (p=0.013), polymorphisme CRABP2-SNP (rs12724719:A/G) homozygote/AA (4 enfants, 3%) avec hautes valeurs moyennes d’AR (141,3 nmol/l). Discussion-Conclusion : Les grandes ΔRA suggèrent une variabilité génique individuelle du métabolisme de ROL. Les génotypes (CA)-ALDH1A2/ SNP (rs12591551:A/C) et (AA) -CRABP2/SNP (rs12724719:A/G) sont associés à des valeurs moyennes hautes d’AR, pouvant protéger l’embryogénèse lors d’une hypovitaminose A maternelle.

Relevância:

60.00% 60.00%

Publicador:

Resumo:

Over the past 100 years, advances in pharmaceutical and medical technology have reduced the burden of communicable disease, and our appreciation of the mechanisms underlying the development of noncommunicable disease has broadened. During this time, a number of studies, both in humans and animal models, have highlighted the importance of maintaining an optimal diet during pregnancy. In particular, a number of studies support the hypothesis that suboptimal maternal protein and fat intake during pregnancy can have long-term effects on the growing fetus, and increase the likelihood of these offspring developing cardiovascular, renal, or metabolic diseases in adulthood. More recently, it has been shown that dietary intake of a number of micronutrients may offset or reverse the deleterious effects of macronutrient imbalance. Furthermore, maternal fat intake has also been identified as a major contributor to a healthy fetal environment, with a beneficial role for unsaturated fats during development as well as a beneficial impact on cell membrane physiology. Together these studies indicate that attempts to optimise maternal nutrition may prove to be an efficient and cost-effective strategy for preventing the development of cardiovascular, renal, or metabolic diseases.

Relevância:

60.00% 60.00%

Publicador:

Resumo:

Multicentric carpotarsal osteolysis (MCTO) is a rare skeletal dysplasia characterized by aggressive osteolysis, particularly affecting the carpal and tarsal bones, and is frequently associated with progressive renal failure. Using exome capture and next-generation sequencing in five unrelated simplex cases of MCTO, we identified previously unreported missense mutations clustering within a 51 base pair region of the single exon of MAFB, validated by Sanger sequencing. A further six unrelated simplex cases with MCTO were also heterozygous for previously unreported mutations within this same region, as were affected members of two families with autosomal-dominant MCTO. MAFB encodes a transcription factor that negatively regulates RANKL-induced osteoclastogenesis and is essential for normal renal development. Identification of this gene paves the way for development of novel therapeutic approaches for this crippling disease and provides insight into normal bone and kidney development.

Relevância:

60.00% 60.00%

Publicador:

Resumo:

Ansatz zur Generierung einer konditionalen, reversiblen Wt1 k.o.-Maus Der Wilms-Tumor (WT, Nephroblastom) ist ein embryonaler Nierentumor, der durch die maligne Transformation von undifferenziertem Nierengewebe, sog. nephrogenen Resten, entsteht. WT treten mit einer Inzidenz von 1 in 10.000 Lebendgeburten auf. Das Hauptmanifestationsalter, der normalerweise einseitig und sporadisch auftretenden Tumore, liegt zwischen dem 3. und 4. Lebensjahr. Etwa 10 % der Patienten entwickeln jedoch bilaterale Tumore. In diesen Fällen ist eine Assoziation mit komplexen genetischen Krankheitsbildern (u. a. WAGR-, Denys-Drash-, Frasier- und Beckwith-Wiedemann-Syndrom) festzustellen. In 15 % der sporadischen WT sind Mutationen im WT1 (Wilms-Tumor 1)-Gen beschrieben. WT1 besteht aus zehn Exons und weist typische Merkmale von Transkriptionsfaktoren (z. B. vier Zinkfinger) auf. Zwei alternative Spleißereignisse betreffen Exon 5 (+/−Exon 5) und Exon 9 (Transkripte mit bzw. ohne die codierenden Sequenzen für die AS Lysin-Threonin-Serin; +/−KTS). Die Lage der drei alternativ vorhandenen AS zwischen den Zinkfingern 3 und 4 bestimmt die verschiedenen Funktionen der WT1-Proteine (4 Isoformen) als Transkriptionsfaktor (−KTS) bzw. als RNA-bindendes Protein (+KTS). Das zunächst im Zusammenhang mit WT als Tumorsuppressorgen identifizierte WT1 ist ein Entwicklungsgen mit einem sehr komplexen Expressionsmuster in der Embryonalentwicklung. Dabei ist v. a. die Bedeutung in der Urogenitalentwicklung entscheidend. Konstitutive, homozygote Wt1−/− k.o.-Mäuse sind embryonal (~ E12,5 dpc) letal und bilden u. a. keine Gonaden und keine Nieren. Aus diesem Grund existiert bisher kein Wilms-Tumormodell. Die Herstellung eines konditionalen murinen Tiermodells auf Basis des Tet on/off-Systems zur Untersuchung der Nierenentwicklung bzw. zur Analyse der Wilms-Tumorpathogenese war Ziel dieser Arbeit. Hierfür wurden drei Mauslinien generiert: Zwei transgene sog. Responder-Linien, die eine chimäre spleißbare Wt1-cDNA der Variante musWt1+Exon 5;+/−KTS unter der Kontrolle eines Tet-responsiven Promotors im Genom tragen. Dieses tTA/Dox-abhängig regulierbare Wt1-Transgen (tgWt1) sollte (exogen regulierbar) die Expression des endogenen Wt1-Lokus ausreichend nachahmen, um die kritischen Phasen der Embryogenese zu überwinden und lebensfähige Tiere zu erhalten. Parallel dazu wurde die Wt1-Effektor-Mauslinie (WE2) generiert. Diese trägt einen tetrazyklinabhängigen Transaktivator (tTA) zur Steuerung Tet-regulierbarer Transgene unter der Kontrolle des endogenen Wt1-Promotors. Die durch homologe Rekombination in ES-Zellen erreichte Integration des tTA direkt am Translationsstartpunkt des Wt1-Lokus hat in den Tieren einen heterozygoten Wt1 knock out/tTA knock in zur Folge. Die bisher vorgenommenen Verpaarungen doppelt transgener Wt1-tTA+/−/Resp-Mäuse ergaben keinen Rescue des letalen Wt1 k.o. und es konnten bislang keine Wilms-Tumore induziert werden. Alle im Verlauf der Arbeit generierten Mauslinien wurden umfassend charakterisiert. So konnte für die Tiere der Responder-Linien Wt1-Resp1 (mit zusätzlichen Isolator-Sequenzen zum Schutz des Transgens vor Positionseffekten) und Wt1-Resp2 (ohne Isolatoren) konnte die Tet-induzierbare Expression und die Spleißbarkeit des tgWt1 in MEF-Assays und mittels Effektor-Mäusen auf RNA-Ebene nachgewiesen werden. Die genomische Charakterisierung der WE2-Linie ergab eine ungeklärte etwa 120 kb große Inversion am Wt1-Lokus, die alle 5'-regulatorischen Sequenzen mitsamt des tTA vom Rest von Wt1 trennt. Tiere dieser Linie weisen aber dennoch einen funktionalen Wt1 k.o. auf: Unter den Nachkommen aus Intercross-Verpaarungen von Wt1-tTA+/−-Mäusen lassen sich auf Grund der Letalität keine Wt1−/−-Genotypen nachweisen. Die Charakterisierung der Effektor-Linie auf RNA-Ebene und mittels Reporter-Mäusen liefert ein Wt1-analoges tTA-Expressionsmuster: So findet man eine deutliche tTA-Expression u. a. in Niere (Glomeruli), Uterus, Ovar und Testis. Die hier vorgestellten Experimente ergeben darüber hinaus eindeutige Hinweise einer Beteiligung von Wt1 in der Entstehung der glatten Muskulatur bzw. in der Vaskulogenese.

Relevância:

60.00% 60.00%

Publicador:

Resumo:

FGFRL1 (fibroblast growth factor receptor like 1) is the most recently discovered member of the FGFR family. It contains three extracellular Ig-like domains similar to the classical FGFRs, but it lacks the protein tyrosine kinase domain and instead contains a short intracellular tail with a peculiar histidine-rich motif. The gene for FGFRL1 is found in all metazoans from sea anemone to mammals. FGFRL1 binds to FGF ligands and heparin with high affinity. It exerts a negative effect on cell proliferation, but a positive effect on cell differentiation. Mice with a targeted deletion of the Fgfrl1 gene die perinatally due to alterations in their diaphragm. These mice also show bilateral kidney agenesis, suggesting an essential role for Fgfrl1 in kidney development. A human patient with a frameshift mutation exhibits craniosynostosis, arguing for an additional role of FGFRL1 during bone formation. FGFRL1 contributes to the complexity of the FGF signaling system.

Relevância:

60.00% 60.00%

Publicador:

Resumo:

BACKGROUND: Renal hypodysplasia, characterized by a decrease in nephron number, small overall kidney size, and maldeveloped renal tissue, is a leading cause of chronic renal failure in young children. Familial clustering and renal hypodysplasia phenotypes observed in transgenic animal models suggest a genetic contribution. Uroplakin IIIa (encoded by UPIIIA) is an integral membrane protein present in urothelial plaques, and the murine UPIIIa knockout is associated with urothelial anomalies and vesicoureteral reflux. De novo UPIIIA mutations recently were identified in 4 of 17 patients with severe bilateral renal adysplasia. METHODS: To evaluate the overall role of UPIIIA in human renal hypodysplasia pathogenesis, we performed UPIIIA mutation analysis in a cohort of 170 pediatric patients affected by severe unilateral or bilateral renal hypodysplasia. Eighty-one patients were affected by bilateral nonobstructive renal hypodysplasia; of these, 61 were without vesicoureteral reflux. Eighty-four patients presented with unilateral nonobstructive renal hypodysplasia, including 24 patients with unilateral multicystic dysplastic kidneys. Family history was positive in 11%. RESULTS: Mutation analysis showed 2 heterozygous mutations not observed in 200 race-matched control chromosomes. In only 1 family was distribution of the UPIIIA mutation consistent with a disease-causing effect. This de novo missense mutation (Gly202Asp) was identified in a patient with unilateral multicystic dysplastic kidneys. The second (intronically located) mutation appeared unlikely to be disease causing because it did not segregate with an obvious disease phenotype in the affected family. CONCLUSION: Our results indicate that de novo mutations in UPIIIA can be involved in defective early kidney development, but probably constitute only a rare cause of human renal hypodysplasia in a minor subset of individuals.

Relevância:

60.00% 60.00%

Publicador:

Resumo:

Morphogenesis is the process by which the 3-dimensional structure of the developing embryo takes shape. We are studying xlcaax-1, a gene whose product can be used as a molecular marker for several morphogenetic events. We report here the cellular and subcellular localization of the xlcaax-1 protein during development of Xenopus laevis. Whole mount immunocytochemistry and immunoperoxidase staining of tissue sections showed that during development the xlcaax-1 protein accumulation was coincident with the differentiation of the epidermis, pronephros and mesonephros. In the pronephros and mesonephros the xlcaax-1 protein was localized to the basolateral membrane of differentiated tubule epithelial cells. Thus, the xlcaax-1 protein served as a marker for tubule formation and polarization during Xenopus kidney development. Xlcaax-1 may also be used as a marker for the functional differentiation of the epidermis and the epidermally derived portions of the lens and some cranial nerves. The xlcaax-1 protein was most abundant in kidney and immunogold EM analysis showed that the xlcaax-1 protein was highly enriched in the basal infoldings of the basolateral membrane of the epithelial cells in adult kidney distal tubules. The xlcaax-1 protein was also localized in other ion transporting epithelia. The localization pattern and preliminary functional assays of xlcaax-1 suggest that the protein may function in association with an ion transport channel or pump.^ Cell migration and cell-cell interactions play important roles in numerous processes during morphogenesis. One of these is the formation of the pronephric (wolffian) duct (PD), which connects the pronephros to the cloaca. It is currently accepted that in most amphibians the pronephric duct is formed by active migration of the pronephric duct rudiment (PDR) cells along a pre-determined pathway. However, there is evidence that in Xenopus, the PD may be formed entirely by in situ segregation of cells out of the lateral mesoderm. In this study, we showed, using PDR ablation and X. laevis - X. borealis chimeras, that PD elongation in Xenopus required both active cell migration and an induced recruitment of cells from the posterior lateral plate mesoderm. We also showed that PDR cell migration was limited to only a few stages during development and that this temporal control is due, at least in part, to changes in the competence of the PD pathway to support cell migration. In addition, our data suggested that an alkaline phosphatase (APase) adhesion gradient may be involved in determining this competence. ^