Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
FASEB J ; 38(1): e23329, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-38050412

RESUMEN

Flow-induced shear stress affects renal epithelial cells in the nephron tubule with potential implications for differential functionalities of the individual segments. Disruptions of cellular mechanosensation or flow conditions are associated with the development and progression of various renal diseases. This study investigates the effects of flow on the transcriptome of various renal tubular epithelial cell types. We analyzed the transcriptome of induced renal epithelial cells (iREC) cultured under physiological flow (0.57 ± 0.05 dyn/cm2 ) or in static conditions for 72 h. RNA sequencing showed 861 differentially expressed genes (DEGs), with 503 up- and 358 downregulated under flow. DEGs were linked to extracellular matrix (ECM) components (e.g. Col1a1, Col4a3, Col4a4, Fn1, Smoc2), junctions (Gja1, Tubb5), channel activities (Abcc4, Aqp1), and transcription factors (Foxq1, Lgr6). Next, we performed a meta-analysis comparing our data with three published datasets that subjected epithelial cell lines from distinct segments to flow, including proximal tubule and collecting duct cells. We found that TGF-ß, p53, MAPK, and PI3K are common flow-regulated pathways. Tfrc expression and thus the capability of iron uptake is commonly upregulated under flow. Many DEGs were related to kidney diseases, such as fibrosis (e.g. Tgfb1-3 and Serpine1). To obtain further mechanistic insights we investigated the role of the PI3K pathway in flow sensing. Applying flow and inhibition of PI3K showed significantly altered expression of transcripts related to ECM remodeling, angiogenesis, and ion transport. This suggests that the PI3K pathway is a critical mediator in flow-dependent cellular processes and gene expression, potentially influencing renal development and tissue remodeling. Finally, we derived a cross-cell-line summary of common as well as segment-specific transcriptomic effects, thus providing insights into the molecular mechanisms underlying flow sensing in the nephron tubule.


Asunto(s)
Riñón , Fosfatidilinositol 3-Quinasas , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Riñón/metabolismo , Túbulos Renales Proximales/fisiología , Perfilación de la Expresión Génica , Células Epiteliales/metabolismo
2.
Development ; 148(21)2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34739029

RESUMEN

Genome editing simplifies the generation of new animal models for congenital disorders. However, the detailed and unbiased phenotypic assessment of altered embryonic development remains a challenge. Here, we explore how deep learning (U-Net) can automate segmentation tasks in various imaging modalities, and we quantify phenotypes of altered renal, neural and craniofacial development in Xenopus embryos in comparison with normal variability. We demonstrate the utility of this approach in embryos with polycystic kidneys (pkd1 and pkd2) and craniofacial dysmorphia (six1). We highlight how in toto light-sheet microscopy facilitates accurate reconstruction of brain and craniofacial structures within X. tropicalis embryos upon dyrk1a and six1 loss of function or treatment with retinoic acid inhibitors. These tools increase the sensitivity and throughput of evaluating developmental malformations caused by chemical or genetic disruption. Furthermore, we provide a library of pre-trained networks and detailed instructions for applying deep learning to the reader's own datasets. We demonstrate the versatility, precision and scalability of deep neural network phenotyping on embryonic disease models. By combining light-sheet microscopy and deep learning, we provide a framework for higher-throughput characterization of embryonic model organisms. This article has an associated 'The people behind the papers' interview.


Asunto(s)
Aprendizaje Profundo , Desarrollo Embrionario/genética , Fenotipo , Animales , Anomalías Craneofaciales/embriología , Anomalías Craneofaciales/genética , Anomalías Craneofaciales/patología , Modelos Animales de Enfermedad , Procesamiento de Imagen Asistido por Computador , Ratones , Microscopía , Mutación , Redes Neurales de la Computación , Trastornos del Neurodesarrollo/genética , Trastornos del Neurodesarrollo/patología , Enfermedades Renales Poliquísticas/embriología , Enfermedades Renales Poliquísticas/genética , Enfermedades Renales Poliquísticas/patología , Proteínas de Xenopus/genética , Xenopus laevis
3.
Proc Natl Acad Sci U S A ; 118(39)2021 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-34548398

RESUMEN

Skeletal ciliopathies (e.g., Jeune syndrome, short rib polydactyly syndrome, and Sensenbrenner syndrome) are frequently associated with nephronophthisis-like cystic kidney disease and other organ manifestations. Despite recent progress in genetic mapping of causative loci, a common molecular mechanism of cartilage defects and cystic kidneys has remained elusive. Targeting two ciliary chondrodysplasia loci (ift80 and ift172) by CRISPR/Cas9 mutagenesis, we established models for skeletal ciliopathies in Xenopus tropicalis Froglets exhibited severe limb deformities, polydactyly, and cystic kidneys, closely matching the phenotype of affected patients. A data mining-based in silico screen found ttc30a to be related to known skeletal ciliopathy genes. CRISPR/Cas9 targeting replicated limb malformations and renal cysts identical to the models of established disease genes. Loss of Ttc30a impaired embryonic renal excretion and ciliogenesis because of altered posttranslational tubulin acetylation, glycylation, and defective axoneme compartmentalization. Ttc30a/b transcripts are enriched in chondrocytes and osteocytes of single-cell RNA-sequenced embryonic mouse limbs. We identify TTC30A/B as an essential node in the network of ciliary chondrodysplasia and nephronophthisis-like disease proteins and suggest that tubulin modifications and cilia segmentation contribute to skeletal and renal ciliopathy manifestations of ciliopathies in a cell type-specific manner. These findings have implications for potential therapeutic strategies.


Asunto(s)
Huesos/anomalías , Ciliopatías/patología , Craneosinostosis/patología , Proteínas del Citoesqueleto/metabolismo , Displasia Ectodérmica/patología , Embrión no Mamífero/patología , Anomalías Musculoesqueléticas/patología , Enfermedades Renales Poliquísticas/patología , Tubulina (Proteína)/química , Animales , Huesos/metabolismo , Huesos/patología , Ciliopatías/genética , Ciliopatías/metabolismo , Craneosinostosis/genética , Craneosinostosis/metabolismo , Proteínas del Citoesqueleto/genética , Modelos Animales de Enfermedad , Displasia Ectodérmica/genética , Displasia Ectodérmica/metabolismo , Embrión no Mamífero/metabolismo , Anomalías Musculoesqueléticas/genética , Anomalías Musculoesqueléticas/metabolismo , Fenotipo , Enfermedades Renales Poliquísticas/genética , Enfermedades Renales Poliquísticas/metabolismo , Tubulina (Proteína)/metabolismo , Xenopus laevis
4.
J Am Soc Nephrol ; 34(3): 412-432, 2023 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-36522156

RESUMEN

SIGNIFICANCE STATEMENT: Mutations in hepatocyte nuclear factor-1 ß ( HNF1B ) are the most common monogenic causes of congenital renal malformations. HNF1B is necessary to directly reprogram fibroblasts to induced renal tubule epithelial cells (iRECs) and, as we demonstrate, can induce ectopic pronephric tissue in Xenopus ectodermal organoids. Using these two systems, we analyzed the effect of HNF1B mutations found in patients with cystic dysplastic kidney disease. We found cross-species conserved targets of HNF1B, identified transcripts that are differentially regulated by the patient-specific mutant protein, and functionally validated novel HNF1B targets in vivo . These results highlight evolutionarily conserved transcriptional mechanisms and provide insights into the genetic circuitry of nephrogenesis. BACKGROUND: Hepatocyte nuclear factor-1 ß (HNF1B) is an essential transcription factor during embryogenesis. Mutations in HNF1B are the most common monogenic causes of congenital cystic dysplastic renal malformations. The direct functional consequences of mutations in HNF1B on its transcriptional activity are unknown. METHODS: Direct reprogramming of mouse fibroblasts to induced renal tubular epithelial cells was conducted both with wild-type HNF1B and with patient mutations. HNF1B was expressed in Xenopus ectodermal explants. Transcriptomic analysis by bulk RNA-Seq identified conserved targets with differentially regulated expression by the wild-type or R295C mutant. CRISPR/Cas9 genome editing in Xenopus embryos evaluated transcriptional targets in vivo . RESULTS: HNF1B is essential for reprogramming mouse fibroblasts to induced renal tubular epithelial cells and induces development of ectopic renal organoids from pluripotent Xenopus cells. The mutation R295C retains reprogramming and inductive capacity but alters the expression of specific sets of downstream target genes instead of diminishing overall transcriptional activity of HNF1B. Surprisingly, targets associated with polycystic kidney disease were less affected than genes affected in congenital renal anomalies. Cross-species-conserved transcriptional targets were dysregulated in hnf1b CRISPR-depleted Xenopus embryos, confirming their dependence on hnf1b . CONCLUSIONS: HNF1B activates an evolutionarily conserved program of target genes that disease-causing mutations selectively disrupt. These findings provide insights into the renal transcriptional network that controls nephrogenesis.


Asunto(s)
Factor Nuclear 1-beta del Hepatocito , Enfermedades Renales Quísticas , Animales , Ratones , Factor Nuclear 1-beta del Hepatocito/genética , Riñón/metabolismo , Enfermedades Renales Quísticas/genética , Mutación , Xenopus laevis
5.
J Am Soc Nephrol ; 34(2): 273-290, 2023 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-36414417

RESUMEN

BACKGROUND: About 40 disease genes have been described to date for isolated CAKUT, the most common cause of childhood CKD. However, these genes account for only 20% of cases. ARHGEF6, a guanine nucleotide exchange factor that is implicated in biologic processes such as cell migration and focal adhesion, acts downstream of integrin-linked kinase (ILK) and parvin proteins. A genetic variant of ILK that causes murine renal agenesis abrogates the interaction of ILK with a murine focal adhesion protein encoded by Parva , leading to CAKUT in mice with this variant. METHODS: To identify novel genes that, when mutated, result in CAKUT, we performed exome sequencing in an international cohort of 1265 families with CAKUT. We also assessed the effects in vitro of wild-type and mutant ARHGEF6 proteins, and the effects of Arhgef6 deficiency in mouse and frog models. RESULTS: We detected six different hemizygous variants in the gene ARHGEF6 (which is located on the X chromosome in humans) in eight individuals from six families with CAKUT. In kidney cells, overexpression of wild-type ARHGEF6 -but not proband-derived mutant ARHGEF6 -increased active levels of CDC42/RAC1, induced lamellipodia formation, and stimulated PARVA-dependent cell spreading. ARHGEF6-mutant proteins showed loss of interaction with PARVA. Three-dimensional Madin-Darby canine kidney cell cultures expressing ARHGEF6-mutant proteins exhibited reduced lumen formation and polarity defects. Arhgef6 deficiency in mouse and frog models recapitulated features of human CAKUT. CONCLUSIONS: Deleterious variants in ARHGEF6 may cause dysregulation of integrin-parvin-RAC1/CDC42 signaling, thereby leading to X-linked CAKUT.


Asunto(s)
Sistema Urinario , Anomalías Urogenitales , Humanos , Ratones , Animales , Perros , Anomalías Urogenitales/genética , Riñón/anomalías , Sistema Urinario/anomalías , Integrinas/metabolismo , Proteínas Mutantes/metabolismo , Factores de Intercambio de Guanina Nucleótido Rho/genética
6.
Physiology (Bethesda) ; 37(4): 0, 2022 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-35253460

RESUMEN

The kidney is a complex organ, and how it forms is a fascinating process. New technologies, such as single-cell transcriptomics, and enhanced imaging modalities are offering new approaches to understand the complex and intertwined processes during embryonic kidney development.


Asunto(s)
Desarrollo Embrionario , Riñón , Humanos
7.
Nephrol Dial Transplant ; 36(2): 237-246, 2021 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-33097957

RESUMEN

BACKGROUND: An underlying monogenic cause of early-onset chronic kidney disease (CKD) can be detected in ∼20% of individuals. For many etiologies of CKD manifesting before 25 years of age, >200 monogenic causative genes have been identified to date, leading to the elucidation of mechanisms of renal pathogenesis. METHODS: In 51 families with echogenic kidneys and CKD, we performed whole-exome sequencing to identify novel monogenic causes of CKD. RESULTS: We discovered a homozygous truncating mutation in the transcription factor gene transcription factor CP2-like 1 (TFCP2L1) in an Arabic patient of consanguineous descent. The patient developed CKD by the age of 2 months and had episodes of severe hypochloremic, hyponatremic and hypokalemic alkalosis, seizures, developmental delay and hypotonia together with cataracts. We found that TFCP2L1 was localized throughout kidney development particularly in the distal nephron. Interestingly, TFCP2L1 induced the growth and development of renal tubules from rat mesenchymal cells. Conversely, the deletion of TFCP2L1 in mice was previously shown to lead to reduced expression of renal cell markers including ion transporters and cell identity proteins expressed in different segments of the distal nephron. TFCP2L1 localized to the nucleus in HEK293T cells only upon coexpression with its paralog upstream-binding protein 1 (UBP1). A TFCP2L1 mutant complementary DNA (cDNA) clone that represented the patient's mutation failed to form homo- and heterodimers with UBP1, an essential step for its transcriptional activity. CONCLUSION: Here, we identified a loss-of-function TFCP2L1 mutation as a potential novel cause of CKD in childhood accompanied by a salt-losing tubulopathy.


Asunto(s)
Transición Epitelial-Mesenquimal , Enfermedades Renales/etiología , Mutación , Proteínas Represoras/genética , Animales , Niño , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Femenino , Células HEK293 , Humanos , Enfermedades Renales/metabolismo , Enfermedades Renales/patología , Ratones , Ratones Noqueados , Ratas , Proteínas Represoras/metabolismo , Análisis de la Célula Individual , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Secuenciación del Exoma
8.
Int J Mol Sci ; 22(18)2021 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-34576273

RESUMEN

Vancomycin is a glycopeptide antibiotic used against multi-drug resistant gram-positive bacteria such as Staphylococcus aureus (MRSA). Although invaluable against resistant bacteria, vancomycin harbors adverse drug reactions including cytopenia, ototoxicity, as well as nephrotoxicity. Since nephrotoxicity is a rarely occurring side effect, its mechanism is incompletely understood. Only recently, the actual clinically relevant concentration the in kidneys of patients receiving vancomycin was investigated and were found to exceed plasma concentrations by far. We applied these clinically relevant vancomycin concentrations to murine and canine renal epithelial cell lines and assessed metabolic and lipidomic alterations by untargeted and targeted gas chromatography-mass spectrometry and liquid chromatography-mass spectrometry analyses. Despite marked differences in the lipidome, both cell lines increased anabolic glucose reactions, resulting in higher sorbitol and lactate levels. To the best of our knowledge, this is the first endometabolic profiling of kidney cells exposed to clinically relevant vancomycin concentrations. The presented study will provide a valuable dataset to nephrotoxicity researchers and might add to unveiling the nephrotoxic mechanism of vancomycin.


Asunto(s)
Riñón/efectos de los fármacos , Lipidómica , Vancomicina/farmacología , Animales , Antibacterianos/farmacología , Cromatografía Liquida , Perros , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Cromatografía de Gases y Espectrometría de Masas , Glutatión/metabolismo , Túbulos Renales Colectores/metabolismo , Lípidos/química , Células de Riñón Canino Madin Darby , Espectrometría de Masas , Metabolómica , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Ratones , Infecciones Estafilocócicas/tratamiento farmacológico
9.
Hum Mutat ; 41(12): 2179-2194, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33131181

RESUMEN

Ciliopathies are clinically and genetically heterogeneous diseases. We studied three patients from two independent families presenting with features of Joubert syndrome: abnormal breathing pattern during infancy, developmental delay/intellectual disability, cerebellar ataxia, molar tooth sign on magnetic resonance imaging scans, and polydactyly. We identified biallelic loss-of-function (LOF) variants in CBY1, segregating with the clinical features of Joubert syndrome in the families. CBY1 localizes to the distal end of the mother centriole, contributing to the formation and function of cilia. In accordance with the clinical and mutational findings in the affected individuals, we demonstrated that depletion of Cby1 in zebrafish causes ciliopathy-related phenotypes. Levels of CBY1 transcript were found reduced in the patients compared with controls, suggesting degradation of the mutated transcript through nonsense-mediated messenger RNA decay. Accordingly, we could detect CBY1 protein in fibroblasts from controls, but not from patients by immunofluorescence. Furthermore, we observed reduced ability to ciliate, increased ciliary length, and reduced levels of the ciliary proteins AHI1 and ARL13B in patient fibroblasts. Our data show that CBY1 LOF-variants cause a ciliopathy with features of Joubert syndrome.


Asunto(s)
Anomalías Múltiples/genética , Proteínas Portadoras/genética , Cerebelo/anomalías , Ciliopatías/genética , Anomalías del Ojo/genética , Enfermedades Renales Quísticas/genética , Mutación/genética , Proteínas Nucleares/genética , Retina/anomalías , Anomalías Múltiples/diagnóstico por imagen , Anomalías Múltiples/patología , Adolescente , Animales , Cerebelo/diagnóstico por imagen , Cerebelo/patología , Niño , Preescolar , Cilios/metabolismo , Cilios/patología , Ciliopatías/diagnóstico por imagen , Ciliopatías/patología , Anomalías del Ojo/diagnóstico por imagen , Anomalías del Ojo/patología , Femenino , Fibroblastos/metabolismo , Fibroblastos/patología , Homocigoto , Humanos , Lactante , Recién Nacido , Enfermedades Renales Quísticas/diagnóstico por imagen , Enfermedades Renales Quísticas/patología , Imagen por Resonancia Magnética , Masculino , Linaje , Fenotipo , Retina/diagnóstico por imagen , Retina/patología , Receptor Smoothened/metabolismo , Adulto Joven , Pez Cebra/genética
10.
J Biol Chem ; 293(39): 15243-15255, 2018 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-30111592

RESUMEN

Nephronophthisis (NPH) is an autosomal recessive renal disease leading to kidney failure in children and young adults. The protein products of the corresponding genes (NPHPs) are localized in primary cilia or their appendages. Only about 70% of affected individuals have a mutation in one of 100 renal ciliopathy genes, and no unifying pathogenic mechanism has been identified. Recently, some NPHPs, including NIMA-related kinase 8 (NEK8) and centrosomal protein 164 (CEP164), have been found to act in the DNA-damage response pathway and to contribute to genome stability. Here, we show that NME/NM23 nucleoside-diphosphate kinase 3 (NME3) that has recently been found to facilitate DNA-repair mechanisms binds to several NPHPs, including NEK8, CEP164, and ankyrin repeat and sterile α motif domain-containing 6 (ANKS6). Depletion of nme3 in zebrafish and Xenopus resulted in typical ciliopathy-associated phenotypes, such as renal malformations and left-right asymmetry defects. We further found that endogenous NME3 localizes to the basal body and that it associates also with centrosomal proteins, such as NEK6, which regulates cell cycle arrest after DNA damage. The ciliopathy-typical manifestations of NME3 depletion in two vertebrate in vivo models, the biochemical association of NME3 with validated NPHPs, and its localization to the basal body reveal a role for NME3 in ciliary function. We conclude that mutations in the NME3 gene may aggravate the ciliopathy phenotypes observed in humans.


Asunto(s)
Ciliopatías/genética , Enfermedades Renales Quísticas/congénito , Nucleósido Difosfato Quinasas NM23/genética , Insuficiencia Renal/genética , Animales , Puntos de Control del Ciclo Celular/genética , Cilios/genética , Cilios/patología , Ciliopatías/fisiopatología , Daño del ADN/genética , Reparación del ADN/genética , Modelos Animales de Enfermedad , Humanos , Riñón/patología , Enfermedades Renales Quísticas/genética , Enfermedades Renales Quísticas/patología , Proteínas de Microtúbulos/genética , Quinasas Relacionadas con NIMA/genética , Proteínas Nucleares/genética , Insuficiencia Renal/patología , Xenopus/genética , Pez Cebra/genética
11.
EMBO J ; 34(8): 1078-89, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25712475

RESUMEN

Mucociliary clearance and fluid transport along epithelial surfaces are carried out by multiciliated cells (MCCs). Recently, human mutations in Cyclin O (CCNO) were linked to severe airway disease. Here, we show that Ccno expression is restricted to MCCs and the genetic deletion of Ccno in mouse leads to reduced numbers of multiple motile cilia and characteristic phenotypes of MCC dysfunction including severe hydrocephalus and mucociliary clearance deficits. Reduced cilia numbers are caused by compromised generation of centrioles at deuterosomes, which serve as major amplification platform for centrioles in MCCs. Ccno-deficient MCCs fail to sufficiently generate deuterosomes, and only reduced numbers of fully functional centrioles that undergo maturation to ciliary basal bodies are formed. Collectively, this study implicates CCNO as first known regulator of deuterosome formation and function for the amplification of centrioles in MCCs.


Asunto(s)
Centriolos/fisiología , Ciclinas/fisiología , Animales , Diferenciación Celular/genética , Células Cultivadas , Centriolos/ultraestructura , Cilios/fisiología , Cilios/ultraestructura , Embrión de Mamíferos , Regulación del Desarrollo de la Expresión Génica , Hidrocefalia/embriología , Hidrocefalia/genética , Ratones , Ratones Transgénicos , Depuración Mucociliar/genética , Organogénesis/genética , Tráquea/citología , Tráquea/embriología , Tráquea/metabolismo
12.
Semin Cell Dev Biol ; 51: 117-24, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26851624

RESUMEN

Modern sequencing technology is revolutionizing our knowledge of inherited kidney disease. However, the molecular role of genes affected by the rapidly rising number of identified mutations is lagging behind. Xenopus is a highly useful, but underutilized model organism with unique properties excellently suited to decipher the molecular mechanisms of kidney development and disease. The embryonic kidney (pronephros) can be manipulated on only one side of the animal and its formation observed directly through the translucent skin. The moderate evolutionary distance between Xenopus and humans is a huge advantage for studying basic principles of kidney development, but still allows us to analyze the function of disease related genes. Optogenetic manipulations and genome editing by CRISPR/Cas are exciting additions to the toolbox for disease modelling and will facilitate the use of Xenopus in translational research. Therefore, the future of Xenopus in kidney research is bright.


Asunto(s)
Modelos Animales de Enfermedad , Enfermedades Renales/genética , Riñón/embriología , Xenopus/genética , Animales , Evaluación Preclínica de Medicamentos , Humanos , Riñón/patología , Riñón/fisiopatología , Enfermedades Renales/tratamiento farmacológico , Mutación , Regeneración , Proteínas de Xenopus/genética
13.
Am J Hum Genet ; 97(2): 291-301, 2015 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-26235987

RESUMEN

Congenital anomalies of the kidneys and urinary tract (CAKUT) are the most common cause of chronic kidney disease in the first three decades of life. Identification of single-gene mutations that cause CAKUT permits the first insights into related disease mechanisms. However, for most cases the underlying defect remains elusive. We identified a kindred with an autosomal-dominant form of CAKUT with predominant ureteropelvic junction obstruction. By whole exome sequencing, we identified a heterozygous truncating mutation (c.1010delG) of T-Box transcription factor 18 (TBX18) in seven affected members of the large kindred. A screen of additional families with CAKUT identified three families harboring two heterozygous TBX18 mutations (c.1570C>T and c.487A>G). TBX18 is essential for developmental specification of the ureteric mesenchyme and ureteric smooth muscle cells. We found that all three TBX18 altered proteins still dimerized with the wild-type protein but had prolonged protein half life and exhibited reduced transcriptional repression activity compared to wild-type TBX18. The p.Lys163Glu substitution altered an amino acid residue critical for TBX18-DNA interaction, resulting in impaired TBX18-DNA binding. These data indicate that dominant-negative TBX18 mutations cause human CAKUT by interference with TBX18 transcriptional repression, thus implicating ureter smooth muscle cell development in the pathogenesis of human CAKUT.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/genética , Genes Dominantes/genética , Músculo Liso/embriología , Mutación/genética , Proteínas de Dominio T Box/genética , Uréter/embriología , Sistema Urinario/anomalías , Secuencia de Bases , Ensayo de Cambio de Movilidad Electroforética , Exoma/genética , Células HEK293 , Humanos , Inmunohistoquímica , Inmunoprecipitación , Microscopía Fluorescente , Datos de Secuencia Molecular , Linaje , Análisis de Secuencia de ADN
14.
Development ; 142(1): 174-84, 2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25516973

RESUMEN

Cilia are microtubule-based organelles that are present on most cells and are required for normal tissue development and function. Defective cilia cause complex syndromes with multiple organ manifestations termed ciliopathies. A crucial step during ciliogenesis in multiciliated cells (MCCs) is the association of future basal bodies with the apical plasma membrane, followed by their correct spacing and planar orientation. Here, we report a novel role for ELMO-DOCK1, which is a bipartite guanine nucleotide exchange factor complex for the small GTPase Rac1, and for the membrane-cytoskeletal linker Ezrin, in regulating centriole/basal body migration, docking and spacing. Downregulation of each component results in ciliopathy-related phenotypes in zebrafish and disrupted ciliogenesis in Xenopus epidermal MCCs. Subcellular analysis revealed a striking impairment of basal body docking and spacing, which is likely to account for the observed phenotypes. These results are substantiated by showing a genetic interaction between elmo1 and ezrin b. Finally, we provide biochemical evidence that the ELMO-DOCK1-Rac1 complex influences Ezrin phosphorylation and thereby probably serves as an important molecular switch. Collectively, we demonstrate that the ELMO-Ezrin complex orchestrates ciliary basal body migration, docking and positioning in vivo.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Cuerpos Basales/metabolismo , Cilios/metabolismo , Proteínas del Citoesqueleto/metabolismo , Proteínas de Xenopus/metabolismo , Proteínas de Pez Cebra/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Animales , Axonema/metabolismo , Axonema/ultraestructura , Membrana Celular/metabolismo , Cilios/ultraestructura , Embrión no Mamífero/metabolismo , Embrión no Mamífero/ultraestructura , Proteínas de la Membrana/metabolismo , Proteínas de Microfilamentos/metabolismo , Modelos Biológicos , Fosforilación , Unión Proteica , Xenopus laevis , Pez Cebra/embriología , Proteínas de Unión al GTP rac
15.
J Am Soc Nephrol ; 28(8): 2364-2376, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28381549

RESUMEN

Congenital anomalies of the kidney and urinary tract (CAKUT) are the most common cause of CKD in the first three decades of life. However, for most patients with CAKUT, the causative mutation remains unknown. We identified a kindred with an autosomal dominant form of CAKUT. By whole-exome sequencing, we identified a heterozygous truncating mutation (c.279delG, p.Trp93fs*) of the nuclear receptor interacting protein 1 gene (NRIP1) in all seven affected members. NRIP1 encodes a nuclear receptor transcriptional cofactor that directly interacts with the retinoic acid receptors (RARs) to modulate retinoic acid transcriptional activity. Unlike wild-type NRIP1, the altered NRIP1 protein did not translocate to the nucleus, did not interact with RARα, and failed to inhibit retinoic acid-dependent transcriptional activity upon expression in HEK293 cells. Notably, we also showed that treatment with retinoic acid enhanced NRIP1 binding to RARα RNA in situ hybridization confirmed Nrip1 expression in the developing urogenital system of the mouse. In explant cultures of embryonic kidney rudiments, retinoic acid stimulated Nrip1 expression, whereas a pan-RAR antagonist strongly reduced it. Furthermore, mice heterozygous for a null allele of Nrip1 showed a CAKUT-spectrum phenotype. Finally, expression and knockdown experiments in Xenopus laevis confirmed an evolutionarily conserved role for NRIP1 in renal development. These data indicate that dominant NRIP1 mutations can cause CAKUT by interference with retinoic acid transcriptional signaling, shedding light on the well documented association between abnormal vitamin A levels and renal malformations in humans, and suggest a possible gene-environment pathomechanism in this disease.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Mutación , Proteínas Nucleares/genética , Transducción de Señal/genética , Tretinoina/fisiología , Sistema Urinario/anomalías , Animales , Ratones , Proteína de Interacción con Receptores Nucleares 1
16.
Cell Tissue Res ; 369(1): 143-157, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28401306

RESUMEN

Xenopus is a versatile model organism increasingly used to study organogenesis and genetic diseases. The rapid embryonic development, targeted injections, loss- and gain-of-function experiments and an increasing supply of tools for functional in vivo analysis are unique advantages of the Xenopus system. Here, we review the vast array of methods available that have facilitated its transition into a translational model. We will focus primarily on how these methods have been employed in the study of kidney development, renal function and kidney disease. Future advances in the fields of genome editing, imaging and quantitative 'omics approaches are likely to enable exciting and novel applications for Xenopus to deepen our understanding of core principles of renal development and molecular mechanisms of human kidney disease. Thus, using Xenopus in clinically relevant research diversifies the narrowing pool of "standard" model organisms and provides unique opportunities for translational research.


Asunto(s)
Enfermedades Renales , Riñón/embriología , Modelos Biológicos , Organogénesis , Animales , Enfermedades Renales/embriología , Enfermedades Renales/genética , Larva , Xenopus laevis
17.
Cell Tissue Res ; 369(1): 185-197, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28560692

RESUMEN

Growing knowledge of how cell identity is determined at the molecular level has enabled the generation of diverse tissue types, including renal cells from pluripotent or somatic cells. Recently, several in vitro protocols involving either directed differentiation or transcription-factor-based reprogramming to kidney cells have been established. Embryonic stem cells or induced pluripotent stem cells can be guided towards a kidney fate by exposing them to combinations of growth factors or small molecules. Here, renal development is recapitulated in vitro resulting in kidney cells or organoids that show striking similarities to mammalian embryonic nephrons. In addition, culture conditions are also defined that allow the expansion of renal progenitor cells in vitro. Another route towards the generation of kidney cells is direct reprogramming. Key transcription factors are used to directly impose renal cell identity on somatic cells, thus circumventing the pluripotent stage. This complementary approach to stem-cell-based differentiation has been demonstrated to generate renal tubule cells and nephron progenitors. In-vitro-generated renal cells offer new opportunities for modelling inherited and acquired renal diseases on a patient-specific genetic background. These cells represent a potential source for developing novel models for kidney diseases, drug screening and nephrotoxicity testing and might represent the first steps towards kidney cell replacement therapies. In this review, we summarize current approaches for the generation of renal cells in vitro and discuss the advantages of each approach and their potential applications.


Asunto(s)
Técnicas de Reprogramación Celular/métodos , Reprogramación Celular , Regulación de la Expresión Génica , Riñón , Ingeniería de Tejidos/métodos , Factores de Transcripción , Animales , Humanos , Riñón/citología , Riñón/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
18.
J Biol Chem ; 289(12): 8390-401, 2014 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-24500717

RESUMEN

Although the two ciliopathies Bardet-Biedl syndrome and nephronophthisis share multiple clinical manifestations, the molecular basis for this overlap remains largely unknown. Both BBS11 and NPHP7 are unusual members of their respective gene families. Although BBS11/TRIM32 represents a RING finger E3 ubiquitin ligase also involved in hereditary forms of muscular dystrophy, NPHP7/Glis2 is a Gli-like transcriptional repressor that localizes to the nucleus, deviating from the ciliary localization of most other ciliopathy-associated gene products. We found that BBS11/TRIM32 and NPHP7/Glis2 can physically interact with each other, suggesting that both proteins form a functionally relevant protein complex in vivo. This hypothesis was further supported by the genetic interaction and synergist cyst formation in the zebrafish pronephros model. However, contrary to our expectation, the E3 ubiquitin ligase BBS11/TRIM32 was not responsible for the short half-life of NPHP7/Glis2 but instead promoted the accumulation of mixed Lys(48)/Lys(63)-polyubiquitylated NPHP7/Glis2 species. This modification not only prolonged the half-life of NPHP7/Glis2, but also altered the subnuclear localization and the transcriptional activity of NPHP7/Glis2. Thus, physical and functional interactions between NPHP and Bardet-Biedl syndrome gene products, demonstrated for Glis2 and TRIM32, may help to explain the phenotypic similarities between these two syndromes.


Asunto(s)
Síndrome de Bardet-Biedl/metabolismo , Enfermedades Renales Quísticas/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Factores de Transcripción/metabolismo , Animales , Síndrome de Bardet-Biedl/genética , Células HEK293 , Humanos , Enfermedades Renales Quísticas/genética , Factores de Transcripción de Tipo Kruppel/análisis , Factores de Transcripción de Tipo Kruppel/genética , Mapas de Interacción de Proteínas , Transporte de Proteínas , Factores de Transcripción/genética , Activación Transcripcional , Proteínas de Motivos Tripartitos , Ubiquitina-Proteína Ligasas , Pez Cebra
19.
J Biol Chem ; 289(38): 26344-26356, 2014 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-25100726

RESUMEN

Tight regulation of Wnt/ß-catenin signaling is critical for vertebrate development and tissue maintenance, and deregulation can lead to a host of disease phenotypes, including developmental disorders and cancer. Proteins associated with primary cilia and centrosomes have been demonstrated to negatively regulate canonical Wnt signaling in interphase cells. The plant homeodomain zinc finger protein Jade-1 can act as an E3 ubiquitin ligase-targeting ß-catenin for proteasomal degradation and concentrates at the centrosome and ciliary basal body in addition to the nucleus in interphase cells. We demonstrate that the destruction complex component casein kinase 1α (CK1α) phosphorylates Jade-1 at a conserved SLS motif and reduces the ability of Jade-1 to inhibit ß-catenin signaling. Consistently, Jade-1 lacking the SLS motif is more effective than wild-type Jade-1 in reducing ß-catenin-induced secondary axis formation in Xenopus laevis embryos in vivo. Interestingly, CK1α also phosphorylates ß-catenin and the destruction complex component adenomatous polyposis coli at a similar SLS motif to the effect that ß-catenin is targeted for degradation. The opposing effect of Jade-1 phosphorylation by CK1α suggests a novel example of the dual functions of CK1α activity to either oppose or promote canonical Wnt signaling in a context-dependent manner.


Asunto(s)
Caseína Quinasa Ialfa/fisiología , Proteínas de Homeodominio/metabolismo , Procesamiento Proteico-Postraduccional , Proteínas Supresoras de Tumor/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Secuencia Conservada , Represión Enzimática , Expresión Génica , Células HEK293 , Humanos , Datos de Secuencia Molecular , Fosforilación , Vía de Señalización Wnt , Xenopus laevis , beta Catenina/metabolismo
20.
Kidney Int ; 87(6): 1191-200, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25671767

RESUMEN

Nephronophthisis (NPH) is a heterogenetic autosomal recessive disorder associated with kidney cysts and multiple extrarenal manifestations. The disease-associated gene products (NPHPs) typically contain domains involved in protein-protein interactions, and appear to exert their tissue-specific functions in large protein complexes. Most NPHPs localize to the cilium and/or basal body; however, their precise molecular functions remain largely unknown. We have recently identified the SAM-domain containing protein Anks3 as a potential ANKS6/NPHP16-interacting protein, and report now that Anks3 interacts with several NPHPs as well as with Bicc1 and the oxygen-sensitive asparaginyl hydroxylase HIF1AN. Knockdown of anks3 in zebrafish embryos was associated with NPH-typical manifestations, including ciliary abnormalities, cyst formation, and laterality defects. In multi-ciliated epidermal cells, GFP-tagged Anks3 localizes to the cilium, but forms large aggregates in the absence of NPHP1, indicating that the negatively charged NPHP1 curtails the polymerization of Anks3. Collectively, these findings suggest that Anks3 is a cilia-associated molecule that partners with the ANKS6- and via NPHP1 to the NPHP1-4-8 module. Thus, developmental defects associated with Anks3 depletion in zebrafish suggest that ANKS3 mutations may cause NPH or NPH-like disease in humans.


Asunto(s)
Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Riñón/embriología , Proteínas de Xenopus/metabolismo , Proteínas de Pez Cebra/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Cilios/metabolismo , Proteínas del Citoesqueleto , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Riñón/anomalías , Enfermedades Renales Quísticas/metabolismo , Proteínas de la Membrana/metabolismo , Oxigenasas de Función Mixta/metabolismo , Quinasas Relacionadas con NIMA , Proteínas Nucleares/metabolismo , Polimerizacion , Proteínas Quinasas/metabolismo , Proteínas/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteínas Represoras/metabolismo , Situs Inversus/genética , Xenopus , Proteínas de Xenopus/genética , Pez Cebra , Proteínas de Pez Cebra/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA