Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 297
Filtrar
Más filtros

Intervalo de año de publicación
1.
Genes Dev ; 35(21-22): 1445-1460, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34711653

RESUMEN

Joubert syndrome (JS) is a recessive ciliopathy in which all affected individuals have congenital cerebellar vermis hypoplasia. Here, we report that CEP120, a JS-associated protein involved in centriole biogenesis and cilia assembly, regulates timely neuronal differentiation and the departure of granule neuron progenitors (GNPs) from their germinal zone during cerebellar development. Our results show that depletion of Cep120 perturbs GNP cell cycle progression, resulting in a delay of cell cycle exit in vivo. To dissect the potential mechanism, we investigated the association between CEP120 interactome and the JS database and identified KIAA0753 (a JS-associated protein) as a CEP120-interacting protein. Surprisingly, we found that CEP120 recruits KIAA0753 to centrioles, and that loss of this interaction induces accumulation of GNPs in the germinal zone and impairs neuronal differentiation. Importantly, the replenishment of wild-type CEP120 rescues the above defects, whereas expression of JS-associated CEP120 mutants, which hinder KIAA0753 recruitment, does not. Together, our data reveal a close interplay between CEP120 and KIAA0753 for the germinal zone exit and timely neuronal differentiation of GNPs during cerebellar development, and mutations in CEP120 and KIAA0753 may participate in the heterotopia and cerebellar hypoplasia observed in JS patients.


Asunto(s)
Centriolos , Enfermedades Renales Quísticas , Anomalías Múltiples , Ciclo Celular , Proteínas de Ciclo Celular/metabolismo , Centriolos/genética , Centriolos/metabolismo , Cerebelo/anomalías , Cerebelo/metabolismo , Anomalías del Ojo , Humanos , Enfermedades Renales Quísticas/genética , Enfermedades Renales Quísticas/metabolismo , Proteínas Asociadas a Microtúbulos , Retina/anomalías
2.
Hum Mol Genet ; 33(16): 1442-1453, 2024 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-38751342

RESUMEN

Primary cilia are antenna-like structures protruding from the surface of various eukaryotic cells, and have distinct protein compositions in their membranes. This distinct protein composition is maintained by the presence of the transition zone (TZ) at the ciliary base, which acts as a diffusion barrier between the ciliary and plasma membranes. Defects in cilia and the TZ are known to cause a group of disorders collectively called the ciliopathies, which demonstrate a broad spectrum of clinical features, such as perinatally lethal Meckel syndrome (MKS), relatively mild Joubert syndrome (JBTS), and nonsyndromic nephronophthisis (NPHP). Proteins constituting the TZ can be grouped into the MKS and NPHP modules. The MKS module is composed of several transmembrane proteins and three soluble proteins. TMEM218 was recently reported to be mutated in individuals diagnosed as MKS and JBTS. However, little is known about how TMEM218 mutations found in MKS and JBTS affect the functions of cilia. In this study, we found that ciliary membrane proteins were not localized to cilia in TMEM218-knockout cells, indicating impaired barrier function of the TZ. Furthermore, the exogenous expression of JBTS-associated TMEM218 variants but not MKS-associated variants in TMEM218-knockout cells restored the localization of ciliary membrane proteins. In particular, when expressed in TMEM218-knockout cells, the TMEM218(R115H) variant found in JBTS was able to restore the barrier function of cells, whereas the MKS variant TMEM218(R115C) could not. Thus, the severity of symptoms of MKS and JBTS individuals appears to correlate with the degree of their ciliary defects at the cellular level.


Asunto(s)
Anomalías Múltiples , Cilios , Ciliopatías , Encefalocele , Anomalías del Ojo , Enfermedades Renales Quísticas , Proteínas de la Membrana , Mutación , Retina , Cilios/metabolismo , Cilios/genética , Cilios/patología , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ciliopatías/genética , Ciliopatías/metabolismo , Ciliopatías/patología , Encefalocele/genética , Encefalocele/metabolismo , Encefalocele/patología , Enfermedades Renales Quísticas/genética , Enfermedades Renales Quísticas/metabolismo , Enfermedades Renales Quísticas/patología , Anomalías Múltiples/genética , Anomalías Múltiples/metabolismo , Anomalías Múltiples/patología , Anomalías del Ojo/genética , Anomalías del Ojo/patología , Anomalías del Ojo/metabolismo , Retina/metabolismo , Retina/anomalías , Retina/patología , Cerebelo/anomalías , Cerebelo/metabolismo , Cerebelo/patología , Enfermedades Cerebelosas/genética , Enfermedades Cerebelosas/metabolismo , Enfermedades Cerebelosas/patología , Animales , Membrana Celular/metabolismo , Ratones , Trastornos de la Motilidad Ciliar , Enfermedades Renales Poliquísticas , Retinitis Pigmentosa
3.
J Cell Sci ; 137(13)2024 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-38841887

RESUMEN

Centrosomal proteins play pivotal roles in orchestrating microtubule dynamics, and their dysregulation leads to disorders, including cancer and ciliopathies. Understanding the multifaceted roles of centrosomal proteins is vital to comprehend their involvement in disease development. Here, we report novel cellular functions of CEP41, a centrosomal and ciliary protein implicated in Joubert syndrome. We show that CEP41 is an essential microtubule-associated protein with microtubule-stabilizing activity. Purified CEP41 binds to preformed microtubules, promotes microtubule nucleation and suppresses microtubule disassembly. When overexpressed in cultured cells, CEP41 localizes to microtubules and promotes microtubule bundling. Conversely, shRNA-mediated knockdown of CEP41 disrupts the interphase microtubule network and delays microtubule reassembly, emphasizing its role in microtubule organization. Further, we demonstrate that the association of CEP41 with microtubules relies on its conserved rhodanese homology domain (RHOD) and the N-terminal region. Interestingly, a disease-causing mutation in the RHOD domain impairs CEP41-microtubule interaction. Moreover, depletion of CEP41 inhibits cell proliferation and disrupts cell cycle progression, suggesting its potential involvement in cell cycle regulation. These insights into the cellular functions of CEP41 hold promise for unraveling the impact of its mutations in ciliopathies.


Asunto(s)
Proliferación Celular , Microtúbulos , Humanos , Microtúbulos/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Asociadas a Microtúbulos/genética , Centrosoma/metabolismo , Retina/metabolismo , Retina/patología , Retina/anomalías , Ciliopatías/metabolismo , Ciliopatías/genética , Ciliopatías/patología , Cerebelo/metabolismo , Cerebelo/anomalías , Cerebelo/patología , Enfermedades Renales Quísticas/metabolismo , Enfermedades Renales Quísticas/genética , Enfermedades Renales Quísticas/patología , Cilios/metabolismo , Cilios/patología , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/genética , Animales , Anomalías Múltiples/metabolismo , Anomalías Múltiples/genética , Anomalías Múltiples/patología , Anomalías del Ojo/metabolismo , Anomalías del Ojo/genética , Anomalías del Ojo/patología , Unión Proteica , Ciclo Celular/genética , Células HEK293
4.
Cell ; 145(4): 513-28, 2011 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-21565611

RESUMEN

Nephronophthisis (NPHP), Joubert (JBTS), and Meckel-Gruber (MKS) syndromes are autosomal-recessive ciliopathies presenting with cystic kidneys, retinal degeneration, and cerebellar/neural tube malformation. Whether defects in kidney, retinal, or neural disease primarily involve ciliary, Hedgehog, or cell polarity pathways remains unclear. Using high-confidence proteomics, we identified 850 interactors copurifying with nine NPHP/JBTS/MKS proteins and discovered three connected modules: "NPHP1-4-8" functioning at the apical surface, "NPHP5-6" at centrosomes, and "MKS" linked to Hedgehog signaling. Assays for ciliogenesis and epithelial morphogenesis in 3D renal cultures link renal cystic disease to apical organization defects, whereas ciliary and Hedgehog pathway defects lead to retinal or neural deficits. Using 38 interactors as candidates, linkage and sequencing analysis of 250 patients identified ATXN10 and TCTN2 as new NPHP-JBTS genes, and our Tctn2 mouse knockout shows neural tube and Hedgehog signaling defects. Our study further illustrates the power of linking proteomic networks and human genetics to uncover critical disease pathways.


Asunto(s)
Enfermedades Renales Quísticas/genética , Proteínas de la Membrana/genética , Transducción de Señal , Animales , Ataxina-10 , Centrosoma/metabolismo , Cilios/metabolismo , Trastornos de la Motilidad Ciliar/genética , Encefalocele/genética , Proteínas Hedgehog/metabolismo , Humanos , Enfermedades Renales Quísticas/metabolismo , Ratones , Células 3T3 NIH , Proteínas del Tejido Nervioso/genética , Enfermedades Renales Poliquísticas/genética , Retinitis Pigmentosa , Pez Cebra
5.
FASEB J ; 38(17): e23875, 2024 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-39229897

RESUMEN

Polycystic kidney disease (PKD) is a common hereditary kidney disease. Although PKD occurrence is associated with certain gene mutations, its onset regulatory mechanisms are still not well understood. Here, we first report that the key enzyme geranylgeranyl diphosphate synthase (GGPPS) is specifically expressed in renal tubular epithelial cells of mouse kidneys. We aimed to explore the role of GGPPS in PKD. In this study, we established a Ggppsfl/fl:Cdh16cre mouse model and compared its phenotype with that of wild-type mice. A Ggpps-downregulation HK2 cell model was also used to further determine the role of GGPPS. We found that GGPPS was specifically expressed in renal tubular epithelial cells of mouse kidneys. Its expression also increased with age. Low GGPPS expression was observed in human ADPKD tissues. In the Ggppsfl/fl:Cdh16cre mouse model, Ggpps deletion in renal tubular epithelial cells induced the occurrence and development of renal tubule cystic dilation and caused the death of mice after birth due to abnormal renal function. Enhanced proliferation of cyst-lining epithelial cells was also observed after the knockout of Ggpps. These processes were related to the increased rate of Rheb on membrane/cytoplasm and hyperactivation of mTORC1 signaling. In conclusion, the deficiency of GGPPS in kidney tubules induced the formation of renal cysts. It may play a critical role in PKD pathophysiology. A novel therapeutic strategy could be designed according to this work.


Asunto(s)
Túbulos Renales , Animales , Ratones , Túbulos Renales/metabolismo , Túbulos Renales/patología , Humanos , Farnesiltransferasa/metabolismo , Farnesiltransferasa/genética , Células Epiteliales/metabolismo , Células Epiteliales/patología , Enfermedades Renales Poliquísticas/genética , Enfermedades Renales Poliquísticas/patología , Enfermedades Renales Poliquísticas/metabolismo , Masculino , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , Enfermedades Renales Quísticas/genética , Enfermedades Renales Quísticas/metabolismo , Enfermedades Renales Quísticas/patología , Ratones Noqueados , Línea Celular , Complejos Multienzimáticos
6.
J Cell Physiol ; 239(4): e31189, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38219074

RESUMEN

Joubert syndrome (JBTS) is a systematic developmental disorder mainly characterized by a pathognomonic mid-hindbrain malformation. All known JBTS-associated genes encode proteins involved in the function of antenna-like cellular organelle, primary cilium, which plays essential roles in cellular signal transduction and development. Here, we identified four unreported variants in ARL13B in two patients with the classical features of JBTS. ARL13B is a member of the Ras GTPase family and functions in ciliogenesis and cilia-related signaling. The two missense variants in ARL13B harbored the substitutions of amino acids at evolutionarily conserved positions. Using model cell lines, we found that the accumulations of the missense variants in cilia were impaired and the variants showed attenuated functions in ciliogenesis or the trafficking of INPP5E. Overall, these findings expanded the ARL13B pathogenetic variant spectrum of JBTS.


Asunto(s)
Factores de Ribosilacion-ADP , Anomalías Múltiples , Cerebelo , Anomalías del Ojo , Enfermedades Renales Quísticas , Retina , Humanos , Anomalías Múltiples/genética , Factores de Ribosilacion-ADP/genética , Factores de Ribosilacion-ADP/metabolismo , Cerebelo/anomalías , Cilios/genética , Anomalías del Ojo/genética , Anomalías del Ojo/metabolismo , Anomalías del Ojo/patología , Enfermedades Renales Quísticas/genética , Enfermedades Renales Quísticas/metabolismo , Enfermedades Renales Quísticas/patología , Monoéster Fosfórico Hidrolasas/metabolismo , Retina/metabolismo , Retina/anomalías , Masculino , Femenino , Lactante
7.
Hum Mol Genet ; 31(9): 1357-1369, 2022 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-34740236

RESUMEN

Nephronophthisis-related ciliopathies (NPHP-RC) comprises a group of inherited kidney diseases, caused by mutations in genes encoding proteins localizing to primary cilia. NPHP-RC represents one of the most frequent monogenic causes of renal failure within the first three decades of life, but its molecular disease mechanisms remain unclear. Here, we identified biallelic ANKS6 mutations in two affected siblings with late-onset chronic kidney disease by whole-exome sequencing. We employed patient-derived fibroblasts generating an in vitro model to study the precise biological impact of distinct human ANKS6 mutations, completed by immunohistochemistry studies on renal biopsy samples. Functional studies using patient-derived cells showed an impaired integrity of the ciliary inversin compartment with reduced cilia length. Further analyses demonstrated that ANKS6 deficiency leads to a dysregulation of Hippo-signaling through nuclear yes-associated protein (YAP) imbalance and disrupted ciliary localization of YAP. In addition, an altered transcriptional activity of canonical Wnt target genes and altered expression of non-phosphorylated (active) ß-catenin and phosphorylated glycogen synthase kinase 3ß were observed. Upon ciliation, ANKS6 deficiency revealed a deranged subcellular localization and expression of components of the endocytic recycling compartment. Our results demonstrate that ANKS6 plays a key role in regulating the Hippo pathway, and ANKS6 deficiency is linked to dysregulation of signaling pathways. Our study provides molecular clues in understanding pathophysiological mechanisms of NPHP-RC and may offer new therapeutic targets.


Asunto(s)
Ciliopatías , Enfermedades Renales Quísticas , Enfermedades Renales Poliquísticas , Insuficiencia Renal Crónica , Cilios/patología , Ciliopatías/metabolismo , Femenino , Humanos , Enfermedades Renales Quísticas/metabolismo , Masculino , Mutación , Proteínas Nucleares , Enfermedades Renales Poliquísticas/genética
8.
Cell Tissue Res ; 396(2): 255-267, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38502237

RESUMEN

Joubert syndrome (JS) is a recessively inherited congenital ataxia characterized by hypotonia, psychomotor delay, abnormal ocular movements, intellectual disability, and a peculiar cerebellar and brainstem malformation, the "molar tooth sign." Over 40 causative genes have been reported, all encoding for proteins implicated in the structure or functioning of the primary cilium, a subcellular organelle widely present in embryonic and adult tissues. In this paper, we developed an in vitro neuronal differentiation model using patient-derived induced pluripotent stem cells (iPSCs), to evaluate possible neurodevelopmental defects in JS. To this end, iPSCs from four JS patients harboring mutations in distinct JS genes (AHI1, CPLANE1, TMEM67, and CC2D2A) were differentiated alongside healthy control cells to obtain mid-hindbrain precursors and cerebellar granule cells. Differentiation was monitored over 31 days through the detection of lineage-specific marker expression by qRT-PCR, immunofluorescence, and transcriptomics analysis. All JS patient-derived iPSCs, regardless of the mutant gene, showed a similar impairment to differentiate into mid-hindbrain and cerebellar granule cells when compared to healthy controls. In addition, analysis of primary cilium count and morphology showed notable ciliary defects in all differentiating JS patient-derived iPSCs compared to controls. These results confirm that patient-derived iPSCs are an accessible and relevant in vitro model to analyze cellular phenotypes connected to the presence of JS gene mutations in a neuronal context.


Asunto(s)
Anomalías Múltiples , Diferenciación Celular , Cerebelo , Cerebelo/anomalías , Anomalías del Ojo , Células Madre Pluripotentes Inducidas , Enfermedades Renales Quísticas , Neuronas , Retina , Retina/anomalías , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/citología , Humanos , Anomalías del Ojo/genética , Anomalías del Ojo/patología , Cerebelo/patología , Cerebelo/metabolismo , Neuronas/metabolismo , Anomalías Múltiples/genética , Anomalías Múltiples/patología , Retina/metabolismo , Enfermedades Renales Quísticas/genética , Enfermedades Renales Quísticas/patología , Enfermedades Renales Quísticas/metabolismo , Masculino , Femenino , Mutación/genética , Cilios/metabolismo
9.
FASEB J ; 37(1): e22696, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36520027

RESUMEN

Mutations or deletions in transcription factor hepatocyte nuclear factor 1 homeobox ß (HNF1ß) cause renal cysts and/or malformation, maturity-onset diabetes of the young and electrolyte disturbances. Here, we applied a comprehensive bioinformatic approach on ChIP-seq, RNA-seq, and gene expression array studies to identify novel transcriptional targets of HNF1ß explaining the kidney phenotype of HNF1ß patients. We identified BAR/IMD Domain Containing Adaptor Protein 2 Like 2 (BAIAP2L2), as a novel transcriptional target of HNF1ß and validated direct transcriptional activation of the BAIAP2L2 promoter by a reporter luciferase assay. Using mass spectrometry analysis, we show that BAIAP2L2 binds to other members of the I-BAR domain-containing family: BAIAP2 and BAIAP2L1. Subsequently, the role of BAIAP2L2 in maintaining epithelial cell integrity in the kidney was assessed using Baiap2l2 knockout cell and mouse models. Kidney epithelial cells lacking functional BAIAP2L2 displayed normal F-actin distribution at cell-cell contacts and formed polarized three-dimensional spheroids with a lumen. In vivo, Baiap2l2 knockout mice displayed normal kidney and colon tissue morphology and serum and urine electrolyte concentrations were not affected. Altogether, our study is the first to characterize the function of BAIAP2L2 in the kidney in vivo and we report that mice lacking BAIAP2L2 exhibit normal electrolyte homeostasis and tissue morphology under physiological conditions.


Asunto(s)
Quistes , Enfermedades Renales Quísticas , Animales , Humanos , Ratones , Quistes/genética , Quistes/metabolismo , Electrólitos/metabolismo , Riñón/metabolismo , Enfermedades Renales Quísticas/genética , Enfermedades Renales Quísticas/metabolismo , Ratones Noqueados , Factores de Transcripción/metabolismo , Activación Transcripcional
10.
Int J Mol Sci ; 25(9)2024 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-38731991

RESUMEN

Tuberous sclerosis complex (TSC) presents with renal cysts and benign tumors, which eventually lead to kidney failure. The factors promoting kidney cyst formation in TSC are poorly understood. Inactivation of carbonic anhydrase 2 (Car2) significantly reduced, whereas, deletion of Foxi1 completely abrogated the cyst burden in Tsc1 KO mice. In these studies, we contrasted the ontogeny of cyst burden in Tsc1/Car2 dKO mice vs. Tsc1/Foxi1 dKO mice. Compared to Tsc1 KO, the Tsc1/Car2 dKO mice showed few small cysts at 47 days of age. However, by 110 days, the kidneys showed frequent and large cysts with overwhelming numbers of A-intercalated cells in their linings. The magnitude of cyst burden in Tsc1/Car2 dKO mice correlated with the expression levels of Foxi1 and was proportional to mTORC1 activation. This is in stark contrast to Tsc1/Foxi1 dKO mice, which showed a remarkable absence of kidney cysts at both 47 and 110 days of age. RNA-seq data pointed to profound upregulation of Foxi1 and kidney-collecting duct-specific H+-ATPase subunits in 110-day-old Tsc1/Car2 dKO mice. We conclude that Car2 inactivation temporarily decreases the kidney cyst burden in Tsc1 KO mice but the cysts increase with advancing age, along with enhanced Foxi1 expression.


Asunto(s)
Anhidrasa Carbónica II , Factores de Transcripción Forkhead , Enfermedades Renales Quísticas , Esclerosis Tuberosa , Animales , Ratones , Anhidrasa Carbónica II/genética , Anhidrasa Carbónica II/metabolismo , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Eliminación de Gen , Riñón/patología , Riñón/metabolismo , Enfermedades Renales Quísticas/genética , Enfermedades Renales Quísticas/patología , Enfermedades Renales Quísticas/metabolismo , Ratones Noqueados , Esclerosis Tuberosa/genética , Esclerosis Tuberosa/patología , Esclerosis Tuberosa/metabolismo , Proteína 1 del Complejo de la Esclerosis Tuberosa/genética , Proteína 1 del Complejo de la Esclerosis Tuberosa/metabolismo
11.
J Biol Chem ; 298(3): 101686, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35131266

RESUMEN

In humans, ciliary dysfunction causes ciliopathies, which present as multiple organ defects, including developmental and sensory abnormalities. Sdccag8 is a centrosomal/basal body protein essential for proper cilia formation. Gene mutations in SDCCAG8 have been found in patients with ciliopathies manifesting a broad spectrum of symptoms, including hypogonadism. Among these mutations, several that are predicted to truncate the SDCCAG8 carboxyl (C) terminus are also associated with such symptoms; however, the underlying mechanisms are poorly understood. In the present study, we identified the Sdccag8 C-terminal region (Sdccag8-C) as a module that interacts with the ciliopathy proteins, Ick/Cilk1 and Mak, which were shown to be essential for the regulation of ciliary protein trafficking and cilia length in mammals in our previous studies. We found that Sdccag8-C is essential for Sdccag8 localization to centrosomes and cilia formation in cultured cells. We then generated a mouse mutant in which Sdccag8-C was truncated (Sdccag8ΔC/ΔC mice) using a CRISPR-mediated stop codon knock-in strategy. In Sdccag8ΔC/ΔC mice, we observed abnormalities in cilia formation and ciliopathy-like organ phenotypes, including cleft palate, polydactyly, retinal degeneration, and cystic kidney, which partially overlapped with those previously observed in Ick- and Mak-deficient mice. Furthermore, Sdccag8ΔC/ΔC mice exhibited a defect in spermatogenesis, which was a previously uncharacterized phenotype of Sdccag8 dysfunction. Together, these results shed light on the molecular and pathological mechanisms underlying ciliopathies observed in patients with SDCCAG8 mutations and may advance our understanding of protein-protein interaction networks involved in cilia development.


Asunto(s)
Autoantígenos , Ciliopatías , Enfermedades Renales Quísticas , Proteínas de Neoplasias , Animales , Autoantígenos/metabolismo , Cuerpos Basales , Cilios/metabolismo , Ciliopatías/genética , Ciliopatías/metabolismo , Femenino , Homeostasis , Humanos , Enfermedades Renales Quísticas/metabolismo , Masculino , Mamíferos , Ratones , Mutación , Proteínas de Neoplasias/metabolismo , Proteínas/metabolismo
12.
Int J Mol Sci ; 24(4)2023 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-36834937

RESUMEN

Nephronophthisis (NPHP) is the most prevalent monogenic disease leading to end-stage renal failure in childhood. RhoA activation is involved in NPHP pathogenesis. This study explored the role of the RhoA activator guanine nucleotide exchange factor (GEF)-H1 in NPHP pathogenesis. We analyzed the expression and distribution of GEF-H1 in NPHP1 knockout (NPHP1KO) mice using Western blotting and immunofluorescence, followed by GEF-H1 knockdown. Immunofluorescence and renal histology were used to examine the cysts, inflammation, and fibrosis. A RhoA GTPase activation assay and Western blotting were used to detect the expression of downstream GTP-RhoA and p-MLC2, respectively. In NPHP1 knockdown (NPHP1KD) human kidney proximal tubular cells (HK2 cells), we detected the expressions of E-cadherin and α-smooth muscle actin (α-SMA). In vivo, increased expression and redistribution of GEF-H1, and higher levels of GTP-RhoA and p-MLC2 in renal tissue of NPHP1KO mice were observed, together with renal cysts, fibrosis, and inflammation. These changes were alleviated by GEF-H1 knockdown. In vitro, the expression of GEF-H1 and activation of RhoA were also increased, with increased expression of α-SMA and decreased E-cadherin. GEF-H1 knockdown reversed these changes in NPHP1KD HK2 cells. Thus, the GEF-H1/RhoA/MLC2 axis is activated in NPHP1 defects and may play a pivotal role in NPHP pathogenesis.


Asunto(s)
Quistes , Fibrosis , Enfermedades Renales Quísticas , Factores de Intercambio de Guanina Nucleótido Rho , Animales , Humanos , Ratones , Cadherinas/metabolismo , Quistes/genética , Quistes/metabolismo , Fibrosis/etiología , Fibrosis/metabolismo , Guanosina Trifosfato , Inflamación , Riñón/metabolismo , Riñón/patología , Enfermedades Renales Quísticas/genética , Enfermedades Renales Quísticas/metabolismo , Factores de Intercambio de Guanina Nucleótido Rho/metabolismo , Proteína de Unión al GTP rhoA/metabolismo
13.
J Biol Chem ; 295(43): 14723-14736, 2020 10 23.
Artículo en Inglés | MEDLINE | ID: mdl-32820051

RESUMEN

Primary cilia are generated through the extension of the microtubule-based axoneme. Centrosomal protein 104 (CEP104) localizes to the tip of the elongating axoneme, and CEP104 mutations are linked to a ciliopathy, Joubert syndrome. Thus, CEP104 has been implicated in ciliogenesis. However, the mechanism by which CEP104 regulates ciliogenesis remains elusive. We report here that CEP104 is critical for cilium elongation but not for initiating ciliogenesis. We also demonstrated that the tumor-overexpressed gene (TOG) domain of CEP104 exhibits microtubule-polymerizing activity and that this activity is essential for the cilium-elongating activity of CEP104. Knockdown/rescue experiments showed that the N-terminal jelly-roll (JR) fold partially contributes to cilium-elongating activity of CEP104, but neither the zinc-finger region nor the SXIP motif is required for this activity. CEP104 binds to a centriole-capping protein, CP110, through the zinc-finger region and to a microtubule plus-end-binding protein, EB1, through the SXIP motif, indicating that the binding of CP110 and EB1 is dispensable for the cilium-elongating activity of CEP104. Moreover, CEP104 depletion does not affect CP110 removal from the mother centriole, which suggests that CEP104 functions after the removal of CP110. Last, we also showed that CEP104 is required for the ciliary entry of Smoothened and export of GPR161 upon Hedgehog signal activation and that the TOG domain plays a critical role in this activity. Our results define the roles of the individual domains of CEP104 in its functions in cilium elongation and Hedgehog signaling and should enhance our understanding of the mechanism underlying CEP104 mutation-associated ciliopathies.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Cilios/metabolismo , Proteínas Hedgehog/metabolismo , Transducción de Señal , Anomalías Múltiples/genética , Anomalías Múltiples/metabolismo , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , Línea Celular , Cerebelo/anomalías , Cerebelo/metabolismo , Anomalías del Ojo/genética , Anomalías del Ojo/metabolismo , Células HEK293 , Humanos , Enfermedades Renales Quísticas/genética , Enfermedades Renales Quísticas/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Microtúbulos/metabolismo , Mutación , Fosfoproteínas/metabolismo , Dominios Proteicos , Retina/anomalías , Retina/metabolismo
14.
Biochem Biophys Res Commun ; 582: 57-63, 2021 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-34689106

RESUMEN

BACKGROUND: Nephronophthisis (NPHP) is a kind of ciliopathy. Interstitial fibrosis occurs at the early stage of the disease. TGF-ß/Smad is a key signaling pathway in regulating interstitial fibrosis and epithelial-mesenchymal transition (EMT). In this study, we explored the activation of the TGF-ß/Smad signaling pathway and EMT in NPHP1-defective MDCK cells to further understand the pathogenesis of NPHP. METHODS: NPHP1-knockdown (NPHP1KD) MDCK cells were constructed by recombinant lentiviral short hairpin RNA, and NPHP1-knockout (NPHP1KO) MDCK cells were constructed by using the CRISPR/Cas9 technique. The morphology and migration ability were observed under a microscope. Western blotting was used to detect the expression of E-cadherin, ß-catenin, α-smooth muscle actin (α-SMA), fibroblast-specific protein-1(FSP1), TGF-ß1, Smad2, Smad3, p-Smad3, Smad4 and Smad7. The localization of Smad3 was determined by immunofluorescence assay. RESULTS: NPHP1KD and NPHP1KO MDCK cells were spindle-shaped and presented EMT-like changes. E-cadherin and ß-catenin expression decreased, while α-SMA and FSP1 expression increased; the TGF-ß/Smad signaling pathway was activated, Smad2, Smad3, p-Smad3 and Smad4 expression increased, Smad3 translocated to nuclear and Smad7 expression decreased compared with those in wild type MDCK cells. Overexpression of Smad7 reversed these changes to different degrees. CONCLUSIONS: Our results indicate that NPHP1 defects induce the activation of the TGF-ß/Smad signaling pathway and EMT in MDCK cells. These factors may be implicated in the pathogenesis of interstitial fibrosis in NPHP.


Asunto(s)
Transición Epitelial-Mesenquimal/genética , Enfermedades Renales Quísticas/congénito , Proteína smad7/genética , Factor de Crecimiento Transformador beta/genética , Actinas/genética , Actinas/metabolismo , Animales , Cadherinas/genética , Cadherinas/metabolismo , Perros , Fibrosis , Regulación de la Expresión Génica , Enfermedades Renales Quísticas/genética , Enfermedades Renales Quísticas/metabolismo , Enfermedades Renales Quísticas/patología , Células de Riñón Canino Madin Darby , Modelos Biológicos , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteína de Unión al Calcio S100A4/genética , Proteína de Unión al Calcio S100A4/metabolismo , Transducción de Señal , Proteína smad7/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
15.
Mol Genet Genomics ; 296(1): 33-40, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-32944789

RESUMEN

Joubert syndrome (JBTS), a rare genetic disorder resulted from primary cilium defects or basal-body dysfunction, is characterized by agenesis of cerebellar vermis and abnormal brain stem. Both genotypes and phenotypes of JBTS are highly heterogeneous. The identification of pathogenic gene variation is essential for making a definite diagnosis on JBTS. Here, we found that hypoplasia of cerebellar vermis occurred in three male members in a Chinese family. Then, we performed whole exome sequencing to identify a novel missense mutation c.599T > C (p. L200P) in the OFD1 gene which is the candidate gene of X-linked JBTS (JBST10). The following analysis showed that the variant was absent in the 1000 Genomes, ExAC and the 200 female controls; the position 200 Leucine residue was highly conserved across species; the missense variant was predicted to be deleterious using PolyPhen-2, PROVEAN, SIFT and Mutation Taster. The OFD1 expression was heavily lower in the proband and an induced male fetus compared with a healthy male with a wild-type OFD1 gene. The in vitro expression analysis of transiently transfecting c.599T or c.599C plasmids into HEK-293T cells confirmed that the missense mutation caused OFD1 reduction at the protein level. And further the mutated OFD1 decreased the level of Gli1 protein, a read-out of Sonic hedgehog (SHH) signaling essential for development of central neural system. A known pathogenic variant c.515T > C (p. L172P) showed the similar results. All of these observations suggested that the missense mutation causes the loss function of OFD1, resulting in SHH signaling impairs and brain development abnormality. In addition, the three patients have Dandy-Walker malformation, macrogyria and tetralogy of Fallot, respectively, the latter two of which are firstly found in JBTS10 patients. In conclusion, our findings expand the context of genotype and phenotype in the JBTS10 patients.


Asunto(s)
Anomalías Múltiples/genética , Cerebelo/anomalías , Síndrome de Dandy-Walker/genética , Anomalías del Ojo/genética , Enfermedades Renales Quísticas/genética , Lisencefalia/genética , Mutación Missense , Proteínas/genética , Retina/anomalías , Tetralogía de Fallot/genética , Anomalías Múltiples/diagnóstico por imagen , Anomalías Múltiples/metabolismo , Anomalías Múltiples/patología , Secuencia de Aminoácidos , Tronco Encefálico/anomalías , Tronco Encefálico/diagnóstico por imagen , Tronco Encefálico/metabolismo , Vermis Cerebeloso/anomalías , Vermis Cerebeloso/diagnóstico por imagen , Vermis Cerebeloso/metabolismo , Cerebelo/diagnóstico por imagen , Cerebelo/metabolismo , Cerebelo/patología , Preescolar , Síndrome de Dandy-Walker/diagnóstico por imagen , Síndrome de Dandy-Walker/metabolismo , Síndrome de Dandy-Walker/patología , Anomalías del Ojo/diagnóstico por imagen , Anomalías del Ojo/metabolismo , Anomalías del Ojo/patología , Familia , Femenino , Expresión Génica , Genotipo , Células HEK293 , Proteínas Hedgehog/deficiencia , Proteínas Hedgehog/genética , Humanos , Enfermedades Renales Quísticas/diagnóstico por imagen , Enfermedades Renales Quísticas/metabolismo , Enfermedades Renales Quísticas/patología , Lisencefalia/diagnóstico por imagen , Lisencefalia/metabolismo , Lisencefalia/patología , Masculino , Linaje , Fenotipo , Proteínas/metabolismo , Retina/diagnóstico por imagen , Retina/metabolismo , Retina/patología , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Factores Sexuales , Transducción de Señal , Tetralogía de Fallot/diagnóstico por imagen , Tetralogía de Fallot/metabolismo , Tetralogía de Fallot/patología , Proteína con Dedos de Zinc GLI1/deficiencia , Proteína con Dedos de Zinc GLI1/genética
16.
Am J Pathol ; 190(1): 176-189, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31676329

RESUMEN

Nephronophthisis (NPHP), the leading genetic cause of end-stage renal failure in children and young adults, is a group of autosomal recessive diseases characterized by kidney-cyst degeneration and fibrosis for which no therapy is currently available. To date, mutations in >25 genes have been identified as causes of this disease that, in several cases, result in chronic DNA damage in kidney tubular cells. Among such mutations, those in the transcription factor-encoding GLIS2 cause NPHP type 7. Loss of function of mouse Glis2 causes senescence of kidney tubular cells. Senescent cells secrete proinflammatory molecules that induce progressive organ damage through several pathways, among which NF-κB signaling is prevalent. Herein, we show that the NF-κB signaling is active in Glis2 knockout kidney epithelial cells and that genetic inactivation of the toll-like receptor (TLR)/IL-1 receptor or pharmacologic elimination of senescent cells (senolytic therapy) reduces tubule damage, fibrosis, and apoptosis in the Glis2 mouse model of NPHP. Notably, in Glis2, Tlr2 double knockouts, senescence was also reduced and proliferation was increased, suggesting that loss of TLR2 activity improves the regenerative potential of tubular cells in Glis2 knockout kidneys. Our results further suggest that a combination of TLR/IL-1 receptor inhibition and senolytic therapy may delay the progression of kidney disease in NPHP type 7 and other forms of this disease.


Asunto(s)
Senescencia Celular/inmunología , Modelos Animales de Enfermedad , Inmunidad Innata/inmunología , Enfermedades Renales Quísticas/patología , Túbulos Renales/patología , Factores de Transcripción de Tipo Kruppel/fisiología , Proteínas del Tejido Nervioso/fisiología , Animales , Apoptosis , Enfermedades Renales Quísticas/inmunología , Enfermedades Renales Quísticas/metabolismo , Túbulos Renales/inmunología , Túbulos Renales/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/fisiología , Receptor Toll-Like 2/fisiología
17.
Am J Kidney Dis ; 77(3): 410-419, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33039432

RESUMEN

Primary cilia are specialized sensory organelles that protrude from the apical surface of most cell types. During the past 2 decades, they have been found to play important roles in tissue development and signal transduction, with mutations in ciliary-associated proteins resulting in a group of diseases collectively known as ciliopathies. Many of these mutations manifest as renal ciliopathies, characterized by kidney dysfunction resulting from aberrant cilia or ciliary functions. This group of overlapping and genetically heterogeneous diseases includes polycystic kidney disease, nephronophthisis, and Bardet-Biedl syndrome as the main focus of this review. Renal ciliopathies are characterized by the presence of kidney cysts that develop due to uncontrolled epithelial cell proliferation, growth, and polarity, downstream of dysregulated ciliary-dependent signaling. Due to cystic-associated kidney injury and systemic inflammation, cases result in kidney failure requiring dialysis and transplantation. Of the handful of pharmacologic treatments available, none are curative. It is important to determine the molecular mechanisms that underlie the involvement of the primary cilium in cyst initiation, expansion, and progression for the development of novel and efficacious treatments. This review updates research progress in defining key genes and molecules central to ciliogenesis and renal ciliopathies.


Asunto(s)
Síndrome de Bardet-Biedl/genética , Cilios/metabolismo , Ciliopatías/genética , Enfermedades Renales Poliquísticas/genética , Anomalías Múltiples/genética , Anomalías Múltiples/metabolismo , Anomalías Múltiples/fisiopatología , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras del Transporte Vesicular/genética , Síndrome de Bardet-Biedl/metabolismo , Síndrome de Bardet-Biedl/fisiopatología , Cerebelo/anomalías , Cerebelo/metabolismo , Cerebelo/fisiopatología , Chaperoninas/genética , Cilios/fisiología , Trastornos de la Motilidad Ciliar/genética , Trastornos de la Motilidad Ciliar/metabolismo , Trastornos de la Motilidad Ciliar/fisiopatología , Ciliopatías/metabolismo , Ciliopatías/fisiopatología , Proteínas del Citoesqueleto/genética , Encefalocele/genética , Encefalocele/metabolismo , Encefalocele/fisiopatología , Anomalías del Ojo/genética , Anomalías del Ojo/metabolismo , Anomalías del Ojo/fisiopatología , Humanos , Enfermedades Renales Quísticas/genética , Enfermedades Renales Quísticas/metabolismo , Enfermedades Renales Quísticas/fisiopatología , Amaurosis Congénita de Leber/genética , Amaurosis Congénita de Leber/metabolismo , Amaurosis Congénita de Leber/fisiopatología , Proteínas de la Membrana/genética , Proteínas Asociadas a Microtúbulos/genética , Atrofias Ópticas Hereditarias/genética , Atrofias Ópticas Hereditarias/metabolismo , Atrofias Ópticas Hereditarias/fisiopatología , Enfermedades Renales Poliquísticas/metabolismo , Enfermedades Renales Poliquísticas/fisiopatología , Proteínas/genética , Retina/anomalías , Retina/metabolismo , Retina/fisiopatología , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/metabolismo , Retinitis Pigmentosa/fisiopatología , Canales Catiónicos TRPP/genética
18.
BMC Nephrol ; 22(1): 261, 2021 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-34246230

RESUMEN

BACKGROUND: Nephronophthisis (NPHP) is a chronic tubular interstitial disorder that exhibits an autosomal recessive genetic form and causes progressive renal failure in children. Patients with NPHP rarely show urinary abnormalities, edema, or hypertension. Thus, NPHP is often detected only when renal failure becomes advanced. NPHP can be divided into three types based on the age of end-stage renal failure, i.e., infant type (approximately 5 years old), juvenile type (approximately 13-14 years old), and adolescent type (approximately 19 years old). Here, we report a case of NPHP diagnosed by genetic analysis at 26 years of age with atypical histological abnormalities. CASE PRESENTATION: A 26-year-old woman showed no growth disorders or urinary abnormalities in annual school physical examinations. However, at a check-up at 26 years old, she exhibited renal dysfunction (eGFR 26 mL/min/1.73 m2). Urine tests indicated low specific gravity of urine, but not proteinuria or microscopic hematuria. Urinary ß2-microglobulin was high (805 µg/L), and renal biopsy was performed for definitive diagnosis. Histological findings showed no significant findings in glomeruli. However, moderate fibrosis was observed in the interstitial area, and moderate atrophy was observed in the tubules. There were no significant findings in immunofluorescence analysis, and no electron dense deposits were detected by electron microscopy. Although cyst-like expansion of the tubules was unclear, tubular atrophy was dominantly found in the distal tubule by cytokeratin 7 staining. Genetic analysis of the NPHP1 gene showed complete deletion of this gene, leading to a definitive diagnosis of NPHP. CONCLUSIONS: NPHP is not merely a pediatric disease and is relatively high incidence in patients with adult onset end-stage of renal disease. In this case, typical histological abnormalities, such as cyst-like expansion of the tubular lesion, were not observed, and diagnosis was achieved by genetic analysis of the NPHP1 gene, which is responsible for the onset of NPHP. In patients with renal failure with tubular interstitial disease dominantly in the distal tubules, it is necessary to discriminate NPHP, even in adult cases.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas del Citoesqueleto/genética , Enfermedades Renales Quísticas/congénito , Túbulos Renales , Insuficiencia Renal , Adulto , Atrofia , Biopsia/métodos , Diagnóstico Diferencial , Femenino , Pruebas Genéticas/métodos , Tasa de Filtración Glomerular , Humanos , Queratina-7/metabolismo , Enfermedades Renales Quísticas/diagnóstico , Enfermedades Renales Quísticas/etiología , Enfermedades Renales Quísticas/genética , Enfermedades Renales Quísticas/metabolismo , Enfermedades Renales Quísticas/fisiopatología , Túbulos Renales/diagnóstico por imagen , Túbulos Renales/patología , Insuficiencia Renal/diagnóstico , Insuficiencia Renal/etiología , Eliminación de Secuencia
19.
Int J Mol Sci ; 22(22)2021 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-34830133

RESUMEN

The primary cilium is found in most mammalian cells and plays a functional role in tissue homeostasis and organ development by modulating key signaling pathways. Ciliopathies are a group of genetically heterogeneous disorders resulting from defects in cilia development and function. Patients with ciliopathic disorders exhibit a range of phenotypes that include nephronophthisis (NPHP), a progressive tubulointerstitial kidney disease that commonly results in end-stage renal disease (ESRD). In recent years, distal appendages (DAPs), which radially project from the distal end of the mother centriole, have been shown to play a vital role in primary ciliary vesicle docking and the initiation of ciliogenesis. Mutations in the genes encoding these proteins can result in either a complete loss of the primary cilium, abnormal ciliary formation, or defective ciliary signaling. DAPs deficiency in humans or mice commonly results in NPHP. In this review, we outline recent advances in our understanding of the molecular functions of DAPs and how they participate in nephronophthisis development.


Asunto(s)
Centrosoma/metabolismo , Cilios/metabolismo , Enfermedades Renales Quísticas/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Animales , Cuerpos Basales/metabolismo , Membrana Celular/metabolismo , Centriolos/metabolismo , Vesículas Citoplasmáticas/metabolismo , Humanos , Enfermedades Renales Quísticas/congénito , Modelos Biológicos
20.
Am J Physiol Renal Physiol ; 318(5): F1306-F1312, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-32308017

RESUMEN

Defects in the function of primary cilia are commonly associated with the development of renal cysts. On the other hand, the intact cilium appears to contribute a cystogenic signal whose effectors remain unclear. As integrin-ß1 is required for the cystogenesis caused by the deletion of the polycystin 1 gene, we asked whether it would be similarly important in the cystogenetic process caused by other ciliary defects. We addressed this question by investigating the effect of integrin-ß1 deletion in a ciliopathy genetic model in which the Ift88 gene, a component of complex B of intraflagellar transport that is required for the proper assembly of cilia, is specifically ablated in principal cells of the collecting ducts. We showed that the renal cystogenesis caused by loss of Ift88 is prevented when integrin-ß1 is simultaneously depleted. In parallel, pathogenetic manifestations of the disease, such as increased inflammatory infiltrate and fibrosis, were also significantly reduced. Overall, our data indicate that integrin-ß1 is also required for the renal cystogenesis caused by ciliary defects and point to integrin-ß1-controlled pathways as common drivers of the disease and as possible targets to interfere with the cystogenesis caused by ciliary defects.


Asunto(s)
Cilios/metabolismo , Integrina beta1/metabolismo , Enfermedades Renales Quísticas/metabolismo , Riñón/metabolismo , Animales , Acuaporina 2/genética , Acuaporina 2/metabolismo , Cilios/patología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Fibrosis , Mediadores de Inflamación/metabolismo , Integrina beta1/genética , Riñón/patología , Riñón/fisiopatología , Enfermedades Renales Quísticas/genética , Enfermedades Renales Quísticas/patología , Enfermedades Renales Quísticas/prevención & control , Macrófagos/metabolismo , Macrófagos/patología , Ratones Noqueados , Transducción de Señal , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA