Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Biochem Biophys Res Commun ; 584: 19-25, 2021 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-34753064

RESUMEN

The primary cilium is a sensory organelle at the cell surface with integral functions in cell signaling. It contains a microtubular axoneme that is rooted in the basal body (BB) and serves as a scaffold for the movement of intraflagellar transport (IFT) particles by Kinesin-2 along the cilium. Ift88, a member of the anterograde moving IFT-B1 complex, as well as the Kinesin-2 subunit Kif3a are required for cilia formation. To facilitate signaling, the cilium restricts the access of molecules to its membrane ("ciliary gate"). This is thought to be mediated by cytoskeletal barriers ("subciliary domains") originating from the BB subdistal/distal appendages, the periciliary membrane compartment (PCMC) as well as the transition fibers and zone (TF/TZ). The PCMC is a poorly characterized membrane domain surrounding the ciliary base with exclusion of certain apical membrane proteins. Here we describe that Ift88, but not Kinesin-2, is required for the establishment of the PCMC in MDCK cells. Likewise, in C. elegans mutants of the Ift88 ortholog osm-5 fail to establish the PCMC, while Kinesin-2 deficient osm-3 mutants form PCMCs normally. Furthermore, disruption of IFT-B1 into two subcomplexes, while disrupting ciliogenesis, does not interfere with PCMC formation. Our findings suggest that cilia are not a prerequisite for the formation of the PCMC, and that separate machineries with partially overlapping functions are required for the establishment of each.


Asunto(s)
Membrana Celular/metabolismo , Cilios/metabolismo , Células Epiteliales/metabolismo , Cinesinas/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Animales , Cuerpos Basales/metabolismo , Caenorhabditis elegans/citología , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Citoesqueleto/metabolismo , Perros , Células de Riñón Canino Madin Darby , Microscopía Fluorescente , Proteínas del Tejido Nervioso/metabolismo , Transducción de Señal
2.
PLoS Biol ; 16(8): e2005651, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30080851

RESUMEN

Cilia are organelles specialized in movement and signal transduction. The ciliary transient receptor potential ion channel polycystin-2 (TRPP2) controls elementary cilia-mediated physiological functions ranging from male fertility and kidney development to left-right patterning. However, the molecular components translating TRPP2 channel-mediated Ca2+ signals into respective physiological functions are unknown. Here, we show that the Ca2+-regulated mitochondrial ATP-Mg/Pi solute carrier 25 A 25 (SLC25A25) acts downstream of TRPP2 in an evolutionarily conserved metabolic signaling pathway. We identify SLC25A25 as an essential component in this cilia-dependent pathway using a genome-wide forward genetic screen in Drosophila melanogaster, followed by a targeted analysis of SLC25A25 function in zebrafish left-right patterning. Our data suggest that TRPP2 ion channels regulate mitochondrial SLC25A25 transporters via Ca2+ establishing an evolutionarily conserved molecular link between ciliary signaling and mitochondrial metabolism.


Asunto(s)
Sistemas de Transporte de Aminoácidos Acídicos/metabolismo , Proteínas de Unión al Calcio/metabolismo , Cilios/metabolismo , Canales Catiónicos TRPP/metabolismo , Animales , Antiportadores/metabolismo , Calcio/metabolismo , Canales de Calcio/metabolismo , Drosophila melanogaster/metabolismo , Heterocigoto , Humanos , Canales Iónicos/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Proteínas Mitocondriales/metabolismo , Transducción de Señal , Pez Cebra
3.
Proc Natl Acad Sci U S A ; 114(23): E4621-E4630, 2017 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-28536193

RESUMEN

Podocytes form the outer part of the glomerular filter, where they have to withstand enormous transcapillary filtration forces driving glomerular filtration. Detachment of podocytes from the glomerular basement membrane precedes most glomerular diseases. However, little is known about the regulation of podocyte adhesion in vivo. Thus, we systematically screened for podocyte-specific focal adhesome (FA) components, using genetic reporter models in combination with iTRAQ-based mass spectrometry. This approach led to the identification of FERM domain protein EPB41L5 as a highly enriched podocyte-specific FA component in vivo. Genetic deletion of Epb41l5 resulted in severe proteinuria, detachment of podocytes, and development of focal segmental glomerulosclerosis. Remarkably, by binding and recruiting the RhoGEF ARGHEF18 to the leading edge, EPB41L5 directly controls actomyosin contractility and subsequent maturation of focal adhesions, cell spreading, and migration. Furthermore, EPB41L5 controls matrix-dependent outside-in signaling by regulating the focal adhesome composition. Thus, by linking extracellular matrix sensing and signaling, focal adhesion maturation, and actomyosin activation EPB41L5 ensures the mechanical stability required for podocytes at the kidney filtration barrier. Finally, a diminution of EPB41L5-dependent signaling programs appears to be a common theme of podocyte disease, and therefore offers unexpected interventional therapeutic strategies to prevent podocyte loss and kidney disease progression.


Asunto(s)
Actomiosina/metabolismo , Proteínas del Citoesqueleto/metabolismo , Adhesiones Focales/metabolismo , Proteínas de la Membrana/metabolismo , Podocitos/metabolismo , Animales , Proteínas del Citoesqueleto/deficiencia , Proteínas del Citoesqueleto/genética , Femenino , Adhesiones Focales/patología , Técnicas de Inactivación de Genes , Glomeruloesclerosis Focal y Segmentaria/etiología , Glomeruloesclerosis Focal y Segmentaria/metabolismo , Glomeruloesclerosis Focal y Segmentaria/patología , Humanos , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Síndrome Nefrótico/etiología , Síndrome Nefrótico/metabolismo , Síndrome Nefrótico/patología , Podocitos/patología , Embarazo , Proteómica , Factores de Intercambio de Guanina Nucleótido Rho/metabolismo , Transducción de Señal
4.
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
5.
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
6.
Nephrol Dial Transplant ; 31(4): 663-71, 2016 04.
Artículo en Inglés | MEDLINE | ID: mdl-26610596

RESUMEN

BACKGROUND: ABO-incompatible kidney transplantation (ABOi KTx) expands the living donor transplantation options. However, long-term outcome data, especially in comparison with ABO-compatible kidney transplantation (ABOc KTx), remain limited. Since the first ABOi KTx in Germany on 1 April 2004 at our centre, we have followed 100 ABOi KTx over up to 10 years. METHODS: One hundred ABOi KTx and 248 ABOc KTx from 1 April 2004 until 28 October 2014 were analysed in this observational, single-centre study. Three ABOi KTx and 141 ABOc KTx were excluded because of cyclosporine A-based immunosuppression, and 1 ABOc KTx was lost to follow-up. RESULTS: Median estimated 10-year patient and graft survival in ABOi KTx was 99 and 94%, respectively, and surpassed ABOc-KTx patient and graft survival of 80 and 88%, respectively. The incidence rate of antibody-mediated rejections was 10 and 8%, and that of T-cell-mediated rejections was 17 and 20% in ABOi KTx and ABOc KTx, respectively. Infectious and malignant complications in ABOi KTx were not more common than in ABOc KTx. However, postoperative lymphoceles occurred more frequently in ABOi KTx. Subgroup analysis of ABOi-KTx patients revealed that patients with high-titre isohaemagglutinins before transplantation had equal long-term results compared with low-titre isohaemagglutinin patients. CONCLUSION: Taken together, long-term outcome of ABOi KTx is not inferior to ABOc KTx. Incidences of rejection episodes, infectious complications and malignancies are not increased, despite the more vigorous immunosuppression in ABOi KTx. Our data provide further evidence that ABOi KTx with living donation is a safe, successful and reasonable option to reduce the organ shortage.


Asunto(s)
Sistema del Grupo Sanguíneo ABO/inmunología , Incompatibilidad de Grupos Sanguíneos/inmunología , Rechazo de Injerto/epidemiología , Infecciones/epidemiología , Trasplante de Riñón , Adolescente , Adulto , Anciano , Femenino , Alemania/epidemiología , Supervivencia de Injerto , Humanos , Terapia de Inmunosupresión , Masculino , Persona de Mediana Edad , Complicaciones Posoperatorias , Estudios Prospectivos , Factores de Tiempo , Adulto Joven
7.
Nephrol Dial Transplant ; 31(2): 262-9, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26420894

RESUMEN

BACKGROUND: Chronic kidney disease (CKD) in children is characterized by rapid progression and a high incidence of end-stage renal disease and therefore constitutes an important health problem. While unbiased genetic screens have identified common risk variants influencing renal function and CKD in adults, the presence and identity of such variants in pediatric CKD are unknown. METHODS: The international Pediatric Investigation for Genetic Factors Linked with Renal Progression (PediGFR) Consortium comprises three pediatric CKD cohorts: Chronic Kidney Disease in Children (CKiD), Effect of Strict Blood Pressure Control and ACE Inhibition on the Progression of CRF in Pediatric Patients (ESCAPE) and Cardiovascular Comorbidity in Children with CKD (4C). Clean genotype data from > 10 million genotyped or imputed single-nucleotide polymorphisms (SNPs) were available for 1136 patients with measurements of serum creatinine at study enrollment. Genome-wide association studies were conducted to relate the SNPs to creatinine-based estimated glomerular filtration rate (eGFR crea) and proteinuria (urinary albumin- or protein-to-creatinine ratio ≥ 300 and ≥ 500 mg/g, respectively). In addition, European-ancestry PediGFR patients (cases) were compared with 1347 European-ancestry children without kidney disease (controls) to identify genetic variants associated with the presence of CKD. RESULTS: SNPs with suggestive association P-values < 1 × 10(-5) were identified in 10 regions for eGFR crea, four regions for proteinuria and six regions for CKD including some plausible biological candidates. No SNP was associated at genome-wide significance (P < 5 × 10(-8)). Investigation of the candidate genes for proteinuria in adults from the general population provided support for a region on chromosome 15 near RSL24D1/UNC13C/RAB27A. Conversely, targeted investigation of genes harboring GFR-associated variants in adults from the general population did not reveal significantly associated SNPs in children with CKD. CONCLUSIONS: Our findings suggest that larger collaborative efforts will be needed to draw reliable conclusions about the presence and identity of common variants associated with eGFR, proteinuria and CKD in pediatric populations.


Asunto(s)
Estudio de Asociación del Genoma Completo/métodos , Tasa de Filtración Glomerular/fisiología , Polimorfismo de Nucleótido Simple , Insuficiencia Renal Crónica/genética , Adolescente , Niño , Preescolar , Progresión de la Enfermedad , Europa (Continente)/epidemiología , Femenino , Sitios Genéticos , Genotipo , Humanos , Lactante , Masculino , Morbilidad/tendencias , Insuficiencia Renal Crónica/epidemiología , Insuficiencia Renal Crónica/fisiopatología , Factores de Riesgo
8.
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
9.
Transpl Int ; 28(4): 387-97, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25387763

RESUMEN

ABO-incompatible kidney transplantation is nowadays a well-established procedure to expand living donor transplantation to blood group incompatible donor/recipient constellations. In the last two decades, transplantation protocols evolved to more specific isohaemagglutinin elimination techniques and established competent antirejection protection protocols without the need of splenectomy. ABOi kidney transplantation associated accommodation despite isohaemagglutinin reappearance, C4d positivity of peritubular capillaries as well as the increased incidence of bleeding complications is currently under intense investigation. However, most recent data show excellent graft survival rates equivalent to ABO-compatible kidney transplantation outcome.


Asunto(s)
Sistema del Grupo Sanguíneo ABO , Trasplante de Riñón , Inmunología del Trasplante , Protocolos Clínicos , Complemento C4/inmunología , Rechazo de Injerto/prevención & control , Hemaglutininas/sangre , Hemaglutininas/aislamiento & purificación , Humanos , Inmunoglobulinas Intravenosas , Factores Inmunológicos/uso terapéutico , Rituximab/uso terapéutico
10.
Hum Mol Genet ; 20(16): 3119-28, 2011 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-21596840

RESUMEN

NPHP4 mutations cause nephronophthisis, an autosomal recessive cystic kidney disease associated with renal fibrosis and kidney failure. The NPHP4 gene product nephrocystin-4 interacts with other nephrocystins, cytoskeletal and ciliary proteins; however, the molecular and cellular functions of nephrocystin-4 have remained elusive. Here we demonstrate that nephrocystin-4 is required for normal cloaca formation during zebrafish embryogenesis. Time-lapse imaging of the developing zebrafish pronephros revealed that tubular epithelial cells at the distal pronephros actively migrate between the yolk sac extension and the blood island towards the ventral fin fold to join the proctodeum and to form the cloaca. Nphp4-deficient pronephric duct cells failed to connect with their ectodermal counterparts, and instead formed a vesicle at the obstructed end of the pronephric duct. Nephrocystin-4 interacts with nephrocystin-1 and Par6. Depletion of zebrafish NPHP1 (nphp1) increased the incidence of cyst formation and randomization of the normal body axis, but did not augment cloaca malformation in nphp4-deficient zebrafish embryos. However, simultaneous depletion of zebrafish Par6 (pard6) aggravated cloaca formation defects in nphp4-depleted embryos, suggesting that nphp4 orchestrates directed cell migration and cloaca formation through interaction with the Par protein complex.


Asunto(s)
Cloaca/embriología , Nefronas/embriología , Proteínas de Pez Cebra/metabolismo , Pez Cebra/embriología , Secuencia de Aminoácidos , Animales , Movimiento Celular , Cilios/metabolismo , Cloaca/metabolismo , Cloaca/patología , Clonación Molecular , Embrión no Mamífero/metabolismo , Embrión no Mamífero/patología , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Datos de Secuencia Molecular , Nefronas/metabolismo , Nefronas/patología , Fenotipo , Pez Cebra/genética , Proteínas de Pez Cebra/química , Proteínas de Pez Cebra/deficiencia , Proteínas de Pez Cebra/genética
12.
Sci Rep ; 13(1): 17647, 2023 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-37848494

RESUMEN

CLIC5 belongs to a family of ion channels with six members reported so far. In vertebrates, the CLIC5 gene encodes two different isoforms, CLIC5A and CLIC5B. In addition to its ion channel activity, there is evidence for further functions of CLIC5A, such as the remodeling of the actin cytoskeleton during the formation of a functional glomerulus in the vertebrate kidney. However, its specific role is still incompletely understood and a specific functional role for CLIC5B has not been described yet. Here we report our findings on the differential expression and functions of Clic5a and Clic5b during zebrafish kidney development. Whole-mount in situ hybridization studies revealed specific expression of clic5a in the eye and pronephric glomerulus, and clic5b is expressed in the gut, liver and the pronephric tubules. Clic5 immunostainings revealed that Clic5b is localized in the cilia. Whereas knockdown of Clic5a resulted in leakiness of the glomerular filtration barrier, Clic5b deficient embryos displayed defective ciliogenesis, leading to ciliopathy-associated phenotypes such as ventral body curvature, otolith deposition defects, altered left-right asymmetry and formation of hydrocephalus and pronephric cysts. In addition, Clic5 deficiency resulted in dysregulation of cilia-dependent Wnt signalling pathway components. Mechanistically, we identified a Clic5-dependent activation of the membrane-cytoskeletal linker proteins Ezrin/Radixin/Moesin (ERM) in the pronephric tubules of zebrafish. In conclusion, our in vivo data demonstrates a novel role for Clic5 in regulating essential ciliary functions and identified Clic5 as a positive regulator of ERM phosphorylation.


Asunto(s)
Canales de Cloruro , Cloruros , Cilios , Glomérulos Renales , Proteínas de Microfilamentos , Pez Cebra , Animales , Citoesqueleto de Actina/metabolismo , Canales de Cloruro/genética , Canales de Cloruro/metabolismo , Cloruros/metabolismo , Cilios/genética , Cilios/metabolismo , Glomérulos Renales/metabolismo , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Pez Cebra/genética , Pez Cebra/metabolismo
13.
Hum Mol Genet ; 19(1): 16-24, 2010 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-19801576

RESUMEN

Autosomal dominant polycystic liver disease (PCLD) is caused by mutations of either PRKCSH or Sec63, two proteins associated with the endoplasmic reticulum (ER). Both proteins are involved in carbohydrate processing, folding and translocation of newly synthesized glycoproteins. It is postulated that defective quality control of proteins initiates endoplasmic reticulum-associated degradation (ERAD), which disrupts hepatic homeostasis in patients with PRKCSH or Sec63 mutations. However, the precise molecular mechanisms are not known. Here, we show that over-expression or depletion of PRKCSH in zebrafish embryos leads to pronephric cysts, abnormal body curvature and situs inversus. Identical phenotypic changes are induced by depletion or over-expression of TRPP2. Increased PRKCSH levels ameliorate developmental abnormalities caused by over-expressed TRPP2, whereas excess TRPP2 can compensate the loss PRKCSH, indicating that the proteins share a common signaling pathway. PRKCSH binds the C-terminal domain of TRPP2, and both proteins co-localize within the ER. Furthermore, PRKCSH interacts with Herp, and inhibits Herp-mediated ubiquitination of TRPP2. Our findings suggest that PRKCSH functions as a chaperone-like molecule, which prevents ERAD of TRPP2. Dysequilibrium between TRPP2 and PRKCSH may lead to cyst formation in PCLD patients with PRKCSH mutations, and thereby account for the overlapping manifestations observed in PCLD and autosomal dominant polycystic kidney disease.


Asunto(s)
Proteínas Portadoras/metabolismo , Chaperonas Moleculares/metabolismo , Mutación/genética , Riñón Poliquístico Autosómico Dominante/metabolismo , Procesamiento Proteico-Postraduccional , Canales Catiónicos TRPP/metabolismo , Ubiquitinas/metabolismo , Proteínas de Pez Cebra/metabolismo , Animales , Proteínas Portadoras/genética , Perros , Embrión no Mamífero/anomalías , Embrión no Mamífero/efectos de los fármacos , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/metabolismo , Técnicas de Silenciamiento del Gen , Humanos , Chaperonas Moleculares/genética , Nefronas/efectos de los fármacos , Nefronas/metabolismo , Nefronas/patología , Oligonucleótidos Antisentido/farmacología , Unión Proteica/efectos de los fármacos , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Canales Catiónicos TRPP/genética , Ubiquitinación/efectos de los fármacos , Ubiquitinas/genética , Pez Cebra/embriología , Proteínas de Pez Cebra/genética
14.
Hum Mol Genet ; 19(12): 2347-59, 2010 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-20233749

RESUMEN

Mutations of the immunoglobulin superfamily proteins nephrin and Neph1 lead to congenital nephrotic syndrome in humans or mice. Neph proteins are three closely related molecules that are evolutionarily conserved and mediate cell recognition. Their importance for morphogenetic processes including the formation of the kidney filtration barrier in vertebrates and synaptogenesis in Caenorhabditis elegans has recently been uncovered. However, the individual morphogenetic function of mammalian Neph1-3 isoforms remained elusive. We demonstrate now that the Neph/nephrin family proteins can form cell-cell adhesion modules across species. Expression of all three mammalian Neph isoforms partially rescued mutant C. elegans lacking their Neph homolog syg-1 and restored synapse formation, suggesting a functional redundancy between the three isoforms. Strikingly, the rescue of defective synaptic connectivity was prevented by deletion of the highly conserved cytoplasmic PSD95/Dlg/ZO-1-binding motif of SYG-1/Neph proteins, indicating the critical role of this intracellular signaling motif for SYG-1/Neph-dependent morphogenetic events. To determine the significance of Neph isoform redundancy for vertebrate kidney development, we analyzed the expression pattern and the functional role of Neph proteins in zebrafish. In situ hybridizations identified zNeph1 and zNeph2 as glomerular proteins. Morpholino knockdown of either zNeph1 or zNeph2 resulted in loss of slit diaphragms and leakiness of the glomerular filtration barrier. This is the first report utilizing C. elegans to study mammalian Neph/nephrin protein function and to demonstrate a functional overlap of Neph1-3 proteins. Furthermore, we identify Neph2 as a novel critical regulator of glomerular function, indicating that both Neph1 and Neph2 are required for glomerular maintenance and development.


Asunto(s)
Inmunoglobulinas/fisiología , Riñón/crecimiento & desarrollo , Proteínas de la Membrana/fisiología , Neuronas/fisiología , Secuencia de Aminoácidos , Animales , Caenorhabditis elegans/genética , Caenorhabditis elegans/crecimiento & desarrollo , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/clasificación , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/fisiología , Adhesión Celular , Células HeLa , Humanos , Inmunoglobulinas/clasificación , Inmunoglobulinas/genética , Proteínas de la Membrana/clasificación , Proteínas de la Membrana/genética , Ratones , Modelos Animales , Morfogénesis/genética , Dominios PDZ , Filogenia
15.
Proc Natl Acad Sci U S A ; 106(21): 8579-84, 2009 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-19439659

RESUMEN

Spatial organization of cells and their appendages is controlled by the planar cell polarity pathway, a signaling cascade initiated by the protocadherin Fat in Drosophila. Vertebrates express 4 Fat molecules, Fat1-4. We found that depletion of Fat1 caused cyst formation in the zebrafish pronephros. Knockdown of the PDZ domain containing the adaptor protein Scribble intensified the cyst-promoting phenotype of Fat1 depletion, suggesting that Fat1 and Scribble act in overlapping signaling cascades during zebrafish pronephros development. Supporting the genetic interaction with Fat1, Scribble recognized the PDZ-binding site of Fat1. Depletion of Yes-associated protein 1 (YAP1), a transcriptional co-activator inhibited by Hippo signaling, ameliorated the cyst formation in Fat1-deficient zebrafish, whereas Scribble inhibited the YAP1-induced cyst formation. Thus, reduced Hippo signaling and subsequent YAP1 disinhibition seem to play a role in the development of pronephric cysts after depletion of Fat1 or Scribble. We hypothesize that Hippo signaling is required for normal pronephros development in zebrafish and that Scribble is a candidate link between Fat and the Hippo signaling cascade in vertebrates.


Asunto(s)
Riñón/embriología , Riñón/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Proteínas de Pez Cebra/metabolismo , Pez Cebra/embriología , Pez Cebra/metabolismo , Animales , Animales Modificados Genéticamente , Cadherinas/genética , Cadherinas/metabolismo , Línea Celular , Embrión no Mamífero/embriología , Embrión no Mamífero/metabolismo , Humanos , Proteínas de la Membrana/genética , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Regiones Promotoras Genéticas/genética , Unión Proteica , Proteínas Serina-Treonina Quinasas/genética , Serina-Treonina Quinasa 3 , Pez Cebra/genética , Proteínas de Pez Cebra/genética
16.
Hum Mol Genet ; 17(23): 3655-62, 2008 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-18723859

RESUMEN

Nephronophthisis (NPHP) is an autosomal recessive cystic kidney disease, caused by mutations of at least nine different genes. Several extrarenal manifestations characterize this disorder, including cerebellar defects, situs inversus and retinitis pigmentosa. While the clinical manifestations vary significantly in NPHP, mutations of NPHP5 and NPHP6 are always associated with progressive blindness. This clinical finding suggests that the gene products, nephrocystin-5 and nephrocystin-6, participate in overlapping signaling pathways to maintain photoreceptor homeostasis. To analyze the genetic interaction between these two proteins in more detail, we studied zebrafish embryos after depletion of NPHP5 and NPHP6. Knockdown of zebrafish zNPHP5 and zNPHP6 produced similar phenotypes, and synergistic effects were observed after the combined knockdown of zNPHP5 and zNPHP6. The N-terminal domain of nephrocystin-6-bound nephrocystin-5, and mapping studies delineated the interacting site from amino acid 696 to 896 of NPHP6. In Xenopus laevis, knockdown of NPHP5 caused substantial neural tube closure defects. This phenotype was copied by expression of the nephrocystin-5-binding fragment of nephrocystin-6, and rescued by co-expression of nephrocystin-5, supporting a physical interaction between both gene products in vivo. Since the N- and C-terminal fragments of nephrocystin-6 engage in the formation of homo- and heteromeric protein complexes, conformational changes seem to regulate the interaction of nephrocystin-6 with its binding partners.


Asunto(s)
Proteínas de Unión a Calmodulina/genética , Proteínas de Unión a Calmodulina/metabolismo , Enfermedades Renales Quísticas/metabolismo , Pez Cebra/genética , Pez Cebra/metabolismo , Secuencias de Aminoácidos , Animales , Proteínas de Unión a Calmodulina/química , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Enfermedades Renales Quísticas/complicaciones , Enfermedades Renales Quísticas/embriología , Enfermedades Renales Quísticas/genética , Masculino , Microinyecciones , Tubo Neural/embriología , Tubo Neural/crecimiento & desarrollo , Tubo Neural/metabolismo , Fenotipo , Unión Proteica , Estructura Terciaria de Proteína , Eliminación de Secuencia , Proteínas de Xenopus/genética , Proteínas de Xenopus/metabolismo , Xenopus laevis/embriología , Xenopus laevis/genética , Xenopus laevis/crecimiento & desarrollo , Xenopus laevis/metabolismo , Pez Cebra/embriología , Pez Cebra/crecimiento & desarrollo
17.
Exp Cell Res ; 315(7): 1157-70, 2009 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-19331813

RESUMEN

Mutations of PKD1 cause autosomal dominant polycystic kidney disease (ADPKD), a syndrome characterized by kidney cysts and progressive renal failure. Polycystin-1, the protein encoded by PKD1, is a large integral membrane protein with a short carboxy-terminal cytoplasmic domain that appears to initiate multiple cellular programs. We report now that this polycystin-1 domain contains a novel motif responsible for rearrangements of intermediate filaments, microtubules and the endoplasmic reticulum (ER). This motif reveals homology to CLIMP-63, a microtubule-binding protein that rearranges the ER. Our findings suggest that polycystin-1 influences the shape and localization of both the microtubular network and the ER.


Asunto(s)
Retículo Endoplásmico/metabolismo , Microtúbulos/metabolismo , Canales Catiónicos TRPP/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Ecdisterona/análogos & derivados , Ecdisterona/metabolismo , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Datos de Secuencia Molecular , Riñón Poliquístico Autosómico Dominante/genética , Riñón Poliquístico Autosómico Dominante/metabolismo , Estructura Terciaria de Proteína , Alineación de Secuencia , Canales Catiónicos TRPP/genética
18.
J Am Soc Nephrol ; 19(7): 1342-51, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18417723

RESUMEN

TRPP2, also known as polycystin-2, is a calcium permeable nonselective cation channel that is mutated in autosomal dominant polycystic kidney disease but has also been implicated in the regulation of cardiac development, renal tubular differentiation, and left-to-right (L-R) axis determination. For obtaining further insight into how TRPP2 exerts tissue-specific functions, this study took advantage of PACS-dependent trafficking of TRPP2 in zebrafish larvae. PACS proteins recognize an acidic cluster within the carboxy-terminal domain of TRPP2 that undergoes phosphorylation and mediate retrieval of TRPP2 to the Golgi and endoplasmic reticulum (ER). The interaction of human TRPP2 with PACS proteins can be inhibited by a Ser812Ala mutation (TRPP2(S812A)), thereby allowing TRPP2 to reach other subcellular compartments, and enhanced by a Ser812Asp mutation (TRPP2(S812D)), thereby trapping TRPP2 in the ER. It was found that the TRPP2(S812A) mutant rescued cyst formation of TRPP2-deficient zebrafish larvae to the same degree as wild-type TRPP2, whereas the TRPP2(S812D) mutant was significantly more effective in normalizing the distorted body axis of TRPP2-deficient fish. Surprisingly, the TRPP2(S812D) mutant rescued the abnormalities of L-R asymmetry more effectively than either wild-type or TRPP2(S812A), suggesting that the ER localization of TRPP2 plays an important role in the development of normal L-R asymmetry. Taken together, these findings support the hypothesis that TRPP2 assumes distinct subcellular localizations to exert tissue-specific functions.


Asunto(s)
Riñón/anomalías , Situs Inversus/metabolismo , Canales Catiónicos TRPP/metabolismo , Pez Cebra/anomalías , Secuencia de Aminoácidos , Animales , Secuencia Conservada , Retículo Endoplásmico/metabolismo , Evolución Molecular , Expresión Génica , Aparato de Golgi/metabolismo , Humanos , Datos de Secuencia Molecular , Mutación Missense , Fenotipo , Situs Inversus/genética , Pez Cebra/genética , Pez Cebra/metabolismo
19.
Nat Commun ; 9(1): 3660, 2018 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-30202007

RESUMEN

Kidney injury is a common complication of severe disease. Here, we report that injuries of the zebrafish embryonal kidney are rapidly repaired by a migratory response in 2-, but not in 1-day-old embryos. Gene expression profiles between these two developmental stages identify cxcl12a and myca as candidates involved in the repair process. Zebrafish embryos with cxcl12a, cxcr4b, or myca deficiency display repair abnormalities, confirming their role in response to injury. In mice with a kidney-specific knockout, Cxcl12 and Myc gene deletions suppress mitochondrial metabolism and glycolysis, and delay the recovery after ischemia/reperfusion injury. Probing these observations in zebrafish reveal that inhibition of glycolysis slows fast migrating cells and delays the repair after injury, but does not affect the slow cell movements during kidney development. Our findings demonstrate that Cxcl12 and Myc facilitate glycolysis to promote fast migratory responses during development and repair, and potentially also during tumor invasion and metastasis.


Asunto(s)
Quimiocina CXCL12/metabolismo , Regulación del Desarrollo de la Expresión Génica , Enfermedades Renales/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra/genética , Animales , Animales Modificados Genéticamente , Movimiento Celular , Metabolismo Energético , Eliminación de Gen , Perfilación de la Expresión Génica , Glucólisis , Homeostasis , Riñón/lesiones , Riñón/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Transducción de Señal , Tretinoina/química
20.
Biochem Biophys Res Commun ; 364(4): 861-6, 2007 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-18273441

RESUMEN

Inherited mutations in genes encoding for ciliary proteins lead to a broad spectrum of human diseases, such as polycystic kidney disease (PKD), situs inversus and retinitis pigmentosa. In the human kidney, autosomal dominant PKD (ADPKD) is caused by mutations in PKD1 (PC1), or PKD2 (TRPP2). Both are necessary for ciliary mechanotransduction, whereby bending of the cilium elicits a calcium response in the cell. We have previously shown that overexpression of mutated forms of the chemosensor kidney injury molecule 1 (Kim1) abolishes the flow response in ciliated MDCK cells. Here we identify Kim1 as an endogenous ciliary protein. Kim1 co-precipitates with TRPP2. Mutational analysis reveals that the interaction between Kim1 and TRPP2 requires the ciliary sorting motif in the N-terminus of TRPP2, and the presence of a highly conserved tyrosine in the intracellular tail of Kim1, which has previously been shown to play a role in ciliary flow sensing. These data support the notion that TRPP2 functionally interacts with ciliary chemosensors.


Asunto(s)
Glicoproteínas de Membrana/metabolismo , Receptores Virales/metabolismo , Canales Catiónicos TRPP/metabolismo , Línea Celular , Cilios/metabolismo , Receptor Celular 1 del Virus de la Hepatitis A , Humanos , Glicoproteínas de Membrana/genética , Fosforilación , Unión Proteica , Receptores Virales/genética , Tirosina/genética , Tirosina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA