Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 68
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Genes Dev ; 32(11-12): 781-793, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29891559

RESUMEN

Autosomal dominant polycystic kidney disease (ADPKD) is an inherited disorder caused by mutations in PKD1 or PKD2 and affects one in 500-1000 humans. Limited treatment is currently available for ADPKD. Here we identify the Hippo signaling effector YAP and its transcriptional target, c-Myc, as promoters of cystic kidney pathogenesis. While transgenic overexpression of YAP promotes proliferation and tubule dilation in mouse kidneys, loss of YAP/TAZ or c-Myc suppresses cystogenesis in a mouse ADPKD model resulting from Pkd1 deficiency. Through a comprehensive kinase inhibitor screen based on a novel three-dimensional (3D) culture of Pkd1 mutant mouse kidney cells, we identified a signaling pathway involving the RhoGEF (guanine nucleotide exchange factor) LARG, the small GTPase RhoA, and the RhoA effector Rho-associated kinase (ROCK) as a critical signaling module between PKD1 and YAP. Further corroborating its physiological importance, inhibition of RhoA signaling suppresses cystogenesis in 3D culture of Pkd1 mutant kidney cells as well as Pkd1 mutant mouse kidneys in vivo. Taken together, our findings implicate the RhoA-YAP-c-Myc signaling axis as a critical mediator and potential drug target in ADPKD.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Riñón/fisiopatología , Fosfoproteínas/metabolismo , Enfermedades Renales Poliquísticas/genética , Enfermedades Renales Poliquísticas/fisiopatología , Proteínas Proto-Oncogénicas c-myc/metabolismo , Transducción de Señal , Proteínas de Unión al GTP rho/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Proteínas de Ciclo Celular , Línea Celular , Células Cultivadas , Modelos Animales de Enfermedad , Células HEK293 , Humanos , Riñón/citología , Riñón/patología , Ratones , Fosfoproteínas/genética , Enfermedades Renales Poliquísticas/patología , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Señalizadoras YAP , Proteínas de Unión al GTP rho/genética , Proteína de Unión al GTP rhoA
2.
Am J Hum Genet ; 109(1): 136-156, 2022 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-34890546

RESUMEN

Autosomal dominant polycystic kidney disease (ADPKD), characterized by progressive cyst formation/expansion, results in enlarged kidneys and often end stage kidney disease. ADPKD is genetically heterogeneous; PKD1 and PKD2 are the common loci (∼78% and ∼15% of families) and GANAB, DNAJB11, and ALG9 are minor genes. PKD is a ciliary-associated disease, a ciliopathy, and many syndromic ciliopathies have a PKD phenotype. In a multi-cohort/-site collaboration, we screened ADPKD-diagnosed families that were naive to genetic testing (n = 834) or for whom no PKD1 and PKD2 pathogenic variants had been identified (n = 381) with a PKD targeted next-generation sequencing panel (tNGS; n = 1,186) or whole-exome sequencing (WES; n = 29). We identified monoallelic IFT140 loss-of-function (LoF) variants in 12 multiplex families and 26 singletons (1.9% of naive families). IFT140 is a core component of the intraflagellar transport-complex A, responsible for retrograde ciliary trafficking and ciliary entry of membrane proteins; bi-allelic IFT140 variants cause the syndromic ciliopathy, short-rib thoracic dysplasia (SRTD9). The distinctive monoallelic phenotype is mild PKD with large cysts, limited kidney insufficiency, and few liver cysts. Analyses of the cystic kidney disease probands of Genomics England 100K showed that 2.1% had IFT140 LoF variants. Analysis of the UK Biobank cystic kidney disease group showed probands with IFT140 LoF variants as the third most common group, after PKD1 and PKD2. The proximity of IFT140 to PKD1 (∼0.5 Mb) in 16p13.3 can cause diagnostic confusion, and PKD1 variants could modify the IFT140 phenotype. Importantly, our studies link a ciliary structural protein to the ADPKD spectrum.


Asunto(s)
Alelos , Proteínas Portadoras , Predisposición Genética a la Enfermedad , Mutación , Riñón Poliquístico Autosómico Dominante/genética , Adulto , Anciano , Sustitución de Aminoácidos , Bancos de Muestras Biológicas , Cilios/patología , Variaciones en el Número de Copia de ADN , Femenino , Estudios de Asociación Genética , Pruebas Genéticas , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Pruebas de Función Renal , Masculino , Persona de Mediana Edad , Linaje , Fenotipo , Riñón Poliquístico Autosómico Dominante/diagnóstico , Análisis de Secuencia de ADN , Reino Unido , Secuenciación del Exoma
3.
Gastroenterology ; 166(5): 902-914, 2024 05.
Artículo en Inglés | MEDLINE | ID: mdl-38101549

RESUMEN

BACKGROUND & AIMS: Autosomal dominant polycystic liver disease is a rare condition with a female preponderance, based mainly on pathogenic variants in 2 genes, PRKCSH and SEC63. Clinically, autosomal dominant polycystic liver disease is characterized by vast heterogeneity, ranging from asymptomatic to highly symptomatic hepatomegaly. To date, little is known about the prediction of disease progression at early stages, hindering clinical management, genetic counseling, and the design of randomized controlled trials. To improve disease prognostication, we built a consortium of European and US centers to recruit the largest cohort of patients with PRKCSH and SEC63 liver disease. METHODS: We analyzed an international multicenter cohort of 265 patients with autosomal dominant polycystic liver disease harboring pathogenic variants in PRKCSH or SEC63 for genotype-phenotype correlations, including normalized age-adjusted total liver volumes and polycystic liver disease-related hospitalization (liver event) as primary clinical end points. RESULTS: Classifying individual total liver volumes into predefined progression groups yielded predictive risk discrimination for future liver events independent of sex and underlying genetic defects. In addition, disease severity, defined by age at first liver event, was considerably more pronounced in female patients and patients with PRKCSH variants than in those with SEC63 variants. A newly developed sex-gene score was effective in distinguishing mild, moderate, and severe disease, in addition to imaging-based prognostication. CONCLUSIONS: Both imaging and clinical genetic scoring have the potential to inform patients about the risk of developing symptomatic disease throughout their lives. The combination of female sex, germline PRKCSH alteration, and rapid total liver volume progression is associated with the greatest odds of polycystic liver disease-related hospitalization.


Asunto(s)
Hospitalización , Hepatopatías , Adulto , Femenino , Humanos , Masculino , Persona de Mediana Edad , Proteínas de Unión al Calcio , Quistes/genética , Quistes/diagnóstico por imagen , Quistes/patología , Progresión de la Enfermedad , Europa (Continente) , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Genotipo , Glucosidasas/genética , Hepatomegalia/genética , Hepatomegalia/diagnóstico por imagen , Hospitalización/estadística & datos numéricos , Hígado/patología , Hígado/diagnóstico por imagen , Hepatopatías/genética , Hepatopatías/patología , Hepatopatías/diagnóstico por imagen , Chaperonas Moleculares , Tamaño de los Órganos , Pronóstico , Medición de Riesgo , Factores de Riesgo , Proteínas de Unión al ARN , Índice de Severidad de la Enfermedad , Factores Sexuales , Estados Unidos/epidemiología
4.
Physiol Genomics ; 55(11): 565-577, 2023 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-37720991

RESUMEN

Autosomal dominant polycystic kidney disease (ADPKD) is caused by mutations in polycystin genes, Pkd1 and Pkd2, but the underlying pathogenic mechanisms are poorly understood. To identify genes and pathways that operate downstream of polycystin-2 (PC2), a comprehensive gene expression database was created, cataloging changes in the transcriptome immediately following PC2 protein depletion. To explore cyst initiation processes, an immortalized mouse inner medullary collecting duct line was developed with the ability to knock out the Pkd2 gene conditionally. Genome-wide transcriptome profiling was performed using RNA sequencing in the cells immediately after PC2 was depleted and compared with isogenic control cells. Differentially expressed genes were identified, and a bioinformatic analysis pipeline was implemented. Altered expression of candidate cystogenic genes was validated in Pkd2 knockout mice. The expression of nearly 900 genes changed upon PC2 depletion. Differentially expressed genes were enriched for genes encoding components of the primary cilia, the canonical Wnt pathway, and MAPK signaling. Among the PC2-dependent ciliary genes, the transcription factor Glis3 was significantly downregulated. MAPK signaling formed a key node at the epicenter of PC2-dependent signaling networks. Activation of Wnt and MAPK signaling, concomitant with the downregulation of Glis3, was corroborated in Pkd2 knockout mice. The data identify a PC2 cilia-to-nucleus signaling axis and dysregulation of the Gli-similar subfamily of transcription factors as a potential initiator of cyst formation in ADPKD. The catalog of PC2-regulated genes should provide a valuable resource for future ADPKD research and new opportunities for drug development.NEW & NOTEWORTHY Autosomal dominant polycystic kidney disease (ADPKD) is the most common inherited kidney disease. Mutations in polycystin genes cause the disease, but the underlying mechanisms of cystogenesis are unknown. To help fill this knowledge gap, we created an inducible cell model of ADPKD and assembled a catalog of genes that respond in immediate proximity to polycystin-2 depletion using transcriptomic profiling. The catalog unveils a ciliary signaling-to-nucleus axis proximal to polycystin-2 dysfunction, highlighting Glis, Wnt, and MAPK signaling.


Asunto(s)
Quistes , Riñón Poliquístico Autosómico Dominante , Animales , Ratones , Quistes/complicaciones , Ratones Noqueados , Riñón Poliquístico Autosómico Dominante/genética , Transcriptoma/genética , Canales Catiónicos TRPP/genética
5.
PLoS Genet ; 16(12): e1009217, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33378371

RESUMEN

A unifying feature of polycystin-2 channels is their localization to both primary and motile cilia/flagella. In Drosophila melanogaster, the fly polycystin-2 homologue, Amo, is an ER protein early in sperm development but the protein must ultimately cluster at the flagellar tip in mature sperm to be fully functional. Male flies lacking appropriate Amo localization are sterile due to abnormal sperm motility and failure of sperm storage. We performed a forward genetic screen to identify additional proteins that mediate ciliary trafficking of Amo. Here we report that Drosophila homologues of KPC1 and KPC2, which comprise the mammalian KIP1 ubiquitination-promoting complex (KPC), form a conserved unit that is required for the sperm tail tip localization of Amo. Male flies lacking either KPC1 or KPC2 phenocopy amo mutants and are sterile due to a failure of sperm storage. KPC is a heterodimer composed of KPC1, an E3 ligase, and KPC2 (or UBAC1), an adaptor protein. Like their mammalian counterparts Drosophila KPC1 and KPC2 physically interact and they stabilize one another at the protein level. In flies, KPC2 is monoubiquitinated and phosphorylated and this modified form of the protein is located in mature sperm. Neither KPC1 nor KPC2 directly interact with Amo but they are detected in proximity to Amo at the tip of the sperm flagellum. In summary we have identified a new complex that is involved in male fertility in Drosophila melanogaster.


Asunto(s)
Infertilidad Masculina/genética , Complejos de Ubiquitina-Proteína Ligasa/genética , Animales , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Femenino , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Unión Proteica , Espermatogénesis , Complejos de Ubiquitina-Proteína Ligasa/metabolismo
6.
J Cell Sci ; 133(14)2020 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-32513820

RESUMEN

Cystogenesis is a morphological consequence of numerous genetic diseases of the epithelium. In the kidney, the pathogenic mechanisms underlying the program of altered cell and tubule morphology are obscured by secondary effects of cyst expansion. Here, we developed a new 3D tubuloid system to isolate the rapid changes in protein localization and gene expression that correlate with altered cell and tubule morphology during cyst initiation. Mouse renal tubule fragments were pulsed with a cell differentiation cocktail including glial-derived neurotrophic factor (GDNF) to yield collecting duct-like tubuloid structures with appropriate polarity, primary cilia, and gene expression. Using the 3D tubuloid model with an inducible Pkd2 knockout system allowed the tracking of morphological, protein, and genetic changes during cyst formation. Within hours of inactivation of Pkd2 and loss of polycystin-2, we observed significant progression in tubuloid to cyst morphology that correlated with 35 differentially expressed genes, many related to cell junctions, matrix interactions, and cell morphology previously implicated in cystogenesis.This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Riñón Poliquístico Autosómico Dominante , Animales , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Riñón , Túbulos Renales , Ratones , Morfogénesis/genética , Riñón Poliquístico Autosómico Dominante/genética , Canales Catiónicos TRPP/genética
7.
FASEB J ; 35(10): e21865, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34486178

RESUMEN

Autosomal dominant polycystic kidney disease is a common inherited renal disorder that results from mutations in either PKD1 or PKD2, encoding polycystin-1 (PC1) and polycystin-2 (PC2), respectively. Downregulation or overexpression of PKD1 or PKD2 in mouse models results in renal cyst formation, suggesting that the quantity of PC1 and PC2 needs to be maintained within a tight functional window to prevent cystogenesis. Here we show that enhanced PC2 expression is a common feature of PKD1 mutant tissues, in part due to an increase in Pkd2 mRNA. However, our data also suggest that more effective protein folding contributes to the augmented levels of PC2. We demonstrate that the unfolded protein response is activated in Pkd1 knockout kidneys and in Pkd1 mutant cells and that this is coupled with increased levels of GRP94, an endoplasmic reticulum protein that is a member of the HSP90 family of chaperones. GRP94 was found to physically interact with PC2 and depletion or chemical inhibition of GRP94 led to a decrease in PC2, suggesting that GRP94 serves as its chaperone. Moreover, GRP94 is acetylated and binds to histone deacetylase 6 (HDAC6), a known deacetylase and activator of HSP90 proteins. Inhibition of HDAC6 decreased PC2 suggesting that HDAC6 and GRP94 work together to regulate PC2 levels. Lastly, we showed that inhibition of GRP94 prevents cAMP-induced cyst formation in vitro. Taken together our data uncovered a novel HDAC6-GRP94-related axis that likely participates in maintaining elevated PC2 levels in Pkd1 mutant cells.


Asunto(s)
Quistes/patología , Retículo Endoplásmico/metabolismo , Enfermedades Renales/patología , Glicoproteínas de Membrana/metabolismo , Factor de Transcripción PAX8/fisiología , Canales Catiónicos TRPP/fisiología , Animales , Calcio/metabolismo , Quistes/etiología , Quistes/metabolismo , Enfermedades Renales/etiología , Enfermedades Renales/metabolismo , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Respuesta de Proteína Desplegada
8.
BMC Nephrol ; 23(1): 334, 2022 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-36258169

RESUMEN

BACKGROUND: Tolvaptan was approved in the United States in 2018 for patients with autosomal dominant polycystic kidney disease (ADPKD) at risk of rapid progression as assessed in a 3-year phase 3 clinical trial (TEMPO 3:4). An extension study (TEMPO 4:4) showed continued delay in progression at 2 years, and a trial in patients with later-stage disease (REPRISE) provided confirmatory evidence of efficacy. Given the relatively shorter-term duration of the clinical trials, estimating the longer-term benefit associated with tolvaptan via extrapolation of the treatment effect is an important undertaking. METHODS: A model was developed to simulate a cohort of patients with ADPKD at risk of rapid progression and predict their long-term outcomes using an algorithm organized around the Mayo Risk Classification system, which has five subclasses (1A through 1E) based on estimated kidney growth rates. The model base-case population represents 1280 patients enrolled in TEMPO 3:4 beginning in chronic kidney disease (CKD) stages G1, G2, and G3 across Mayo subclasses 1C, 1D, and 1E. The algorithm was used to predict longer-term natural history health outcomes. The estimated treatment effect of tolvaptan from TEMPO 3:4 was applied to the natural history to predict the longer-term treatment benefit of tolvaptan. For the cohort, analyzed once reflecting natural history and once assuming treatment with tolvaptan, the model estimated lifetime progression through CKD stages, end-stage renal disease (ESRD), and death. RESULTS: When treated with tolvaptan, the model cohort was predicted to experience a 3.1-year delay of ESRD (95% confidence interval: 1.8 to 4.4), approximately a 23% improvement over the estimated 13.7 years for patients not receiving tolvaptan. Patients beginning tolvaptan treatment in CKD stages G1, G2, and G3 were predicted to experience estimated delays of ESRD, compared with patients not receiving tolvaptan, of 3.8 years (21% improvement), 3.0 years (24% improvement), and 2.1 years (28% improvement), respectively. CONCLUSIONS: The model estimated that patients treated with tolvaptan versus no treatment spent more time in earlier CKD stages and had later onset of ESRD. Findings highlight the potential long-term value of early intervention with tolvaptan in patients at risk of rapid ADPKD progression.


Asunto(s)
Fallo Renal Crónico , Riñón Poliquístico Autosómico Dominante , Tolvaptán , Humanos , Antagonistas de los Receptores de Hormonas Antidiuréticas/uso terapéutico , Progresión de la Enfermedad , Fallo Renal Crónico/epidemiología , Riñón Poliquístico Autosómico Dominante/tratamiento farmacológico , Riñón Poliquístico Autosómico Dominante/patología , Factores de Tiempo , Tolvaptán/uso terapéutico , Ensayos Clínicos Fase III como Asunto
9.
J Am Soc Nephrol ; 32(8): 1913-1932, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34155062

RESUMEN

BACKGROUND: In autosomal dominant polycystic kidney disease (ADPKD), cyst development and enlargement lead to ESKD. Macrophage recruitment and interstitial inflammation promote cyst growth. TWEAK is a TNF superfamily (TNFSF) cytokine that regulates inflammatory responses, cell proliferation, and cell death, and its receptor Fn14 (TNFRSF12a) is expressed in macrophage and nephron epithelia. METHODS: To evaluate the role of the TWEAK signaling pathway in cystic disease, we evaluated Fn14 expression in human and in an orthologous murine model of ADPKD. We also explored the cystic response to TWEAK signaling pathway activation and inhibition by peritoneal injection. RESULTS: Meta-analysis of published animal-model data of cystic disease reveals mRNA upregulation of several components of the TWEAK signaling pathway. We also observed that TWEAK and Fn14 were overexpressed in mouse ADPKD kidney cysts, and TWEAK was significantly high in urine and cystic fluid from patients with ADPKD. TWEAK administration induced cystogenesis and increased cystic growth, worsening the phenotype in a murine ADPKD model. Anti-TWEAK antibodies significantly slowed the progression of ADPKD, preserved renal function, and improved survival. Furthermore, the anti-TWEAK cystogenesis reduction is related to decreased cell proliferation-related MAPK signaling, decreased NF-κB pathway activation, a slight reduction of fibrosis and apoptosis, and an indirect decrease in macrophage recruitment. CONCLUSIONS: This study identifies the TWEAK signaling pathway as a new disease mechanism involved in cystogenesis and cystic growth and may lead to a new therapeutic approach in ADPKD.


Asunto(s)
Citocina TWEAK/metabolismo , Riñón Poliquístico Autosómico Dominante/metabolismo , Riñón Poliquístico Autosómico Dominante/patología , Receptor de TWEAK/metabolismo , Adulto , Animales , Anticuerpos Neutralizantes/farmacología , Apoptosis , Proliferación Celular/efectos de los fármacos , Quistes/metabolismo , Quistes/patología , Citocina TWEAK/antagonistas & inhibidores , Citocina TWEAK/genética , Citocina TWEAK/farmacología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Fibrosis , Expresión Génica , Humanos , Activación de Macrófagos/efectos de los fármacos , Macrófagos , Masculino , Ratones , Persona de Mediana Edad , FN-kappa B/metabolismo , Riñón Poliquístico Autosómico Dominante/fisiopatología , Transducción de Señal , Receptor de TWEAK/genética
10.
Kidney Int ; 100(3): 684-696, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34186056

RESUMEN

Autosomal dominant polycystic kidney disease (ADPKD) is characterized by growth of kidney cysts and glomerular filtration rate (GFR) decline. Metformin was found to impact cystogenesis in preclinical models of polycystic disease, is generally considered safe and may be a promising candidate for clinical investigation in ADPKD. In this phase 2 two-year trial, we randomly assigned 97 patients, 18-60 years of age, with ADPKD and estimated GFR over 50 ml/min/1.73 m2, in a 1:1 ratio to receive metformin or placebo twice daily. Primary outcomes were medication safety and tolerability. Secondary outcomes included estimated GFR decline, and total kidney volume growth. Thirty-eight metformin and 39 placebo participants still received study product at 24-months. Twenty-one participants in the metformin arm reduced drug dose due to inability to tolerate, compared with 14 in the placebo arm (not significant). Proportions of participants experiencing serious adverse events was similar between the groups. The Gastrointestinal Symptoms Rating Scale score was low at baseline and did not significantly change over time. The annual change for estimated GFR was -1.71 with metformin and -3.07 ml/min/1.73m2 per year with placebo (mean difference 1.37 {-0.70, 3.44} ml/min/1.73m2), while mean annual percent change in height-adjusted total kidney volume was 3.87% in metformin and 2.16% per year in placebo, (mean difference 1.68% {-2.11, 5.62}). Thus, metformin in adults with ADPKD was found to be safe and tolerable while slightly reducing estimated GFR decline but not to a significant degree. Hence, evaluation of efficacy requires a larger trial, with sufficient power to detect differences in endpoints.


Asunto(s)
Quistes , Metformina , Riñón Poliquístico Autosómico Dominante , Adulto , Progresión de la Enfermedad , Tasa de Filtración Glomerular , Humanos , Riñón , Metformina/efectos adversos , Riñón Poliquístico Autosómico Dominante/tratamiento farmacológico
11.
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
12.
Am J Hum Genet ; 98(6): 1193-1207, 2016 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-27259053

RESUMEN

Autosomal-dominant polycystic kidney disease (ADPKD) is a common, progressive, adult-onset disease that is an important cause of end-stage renal disease (ESRD), which requires transplantation or dialysis. Mutations in PKD1 or PKD2 (∼85% and ∼15% of resolved cases, respectively) are the known causes of ADPKD. Extrarenal manifestations include an increased level of intracranial aneurysms and polycystic liver disease (PLD), which can be severe and associated with significant morbidity. Autosomal-dominant PLD (ADPLD) with no or very few renal cysts is a separate disorder caused by PRKCSH, SEC63, or LRP5 mutations. After screening, 7%-10% of ADPKD-affected and ∼50% of ADPLD-affected families were genetically unresolved (GUR), suggesting further genetic heterogeneity of both disorders. Whole-exome sequencing of six GUR ADPKD-affected families identified one with a missense mutation in GANAB, encoding glucosidase II subunit α (GIIα). Because PRKCSH encodes GIIß, GANAB is a strong ADPKD and ADPLD candidate gene. Sanger screening of 321 additional GUR families identified eight further likely mutations (six truncating), and a total of 20 affected individuals were identified in seven ADPKD- and two ADPLD-affected families. The phenotype was mild PKD and variable, including severe, PLD. Analysis of GANAB-null cells showed an absolute requirement of GIIα for maturation and surface and ciliary localization of the ADPKD proteins (PC1 and PC2), and reduced mature PC1 was seen in GANAB(+/-) cells. PC1 surface localization in GANAB(-/-) cells was rescued by wild-type, but not mutant, GIIα. Overall, we show that GANAB mutations cause ADPKD and ADPLD and that the cystogenesis is most likely driven by defects in PC1 maturation.


Asunto(s)
Quistes/genética , Hepatopatías/genética , Mutación/genética , Riñón Poliquístico Autosómico Dominante/genética , alfa-Glucosidasas/genética , Adulto , Anciano , Secuencia de Aminoácidos , Sistemas CRISPR-Cas , Células Cultivadas , Niño , Femenino , Técnica del Anticuerpo Fluorescente , Humanos , Inmunoprecipitación , Masculino , Microscopía Confocal , Persona de Mediana Edad , Linaje , Riñón Poliquístico Autosómico Dominante/patología , Homología de Secuencia de Aminoácido
13.
BMC Nephrol ; 20(1): 386, 2019 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-31653199

RESUMEN

BACKGROUND: Patients with Autosomal Dominant Polycystic Kidney Disease (ADPKD) often develop hypertension in childhood or early adulthood. Although this could result in left ventricular hypertrophy (LVH), a major risk factor for cardiovascular morbidity and mortality, prior studies of LVH in ADPKD have yielded conflicting results. We estimated the prevalence of LVH using consensus echocardiography criteria and examined the independent association of ADPKD severity with LV mass in a contemporary cohort of ADPKD patients. METHODS: Adults with ADPKD and eGFR> 15 ml/min/1.73m2 were enrolled in a single-center study. Left Ventricular Mass (LVM) was quantified using 2D echocardiography, and LVH was defined using gender-specific cut-points of LVM and LVM indexed to body surface area (LVMI) from consensus guidelines. Total Kidney Volume (TKV) was quantified using Magnetic Resonance Imaging, and GFR was estimated from serum creatinine using the CKD-Epi equation. Multiple linear regression was used to estimate the association of TKV and eGFR with LVM and LVMI, adjusting for potential confounders. RESULTS: Among 126 participants (78% with hypertension), median age was 46 years, median eGFR 63 ml/min/1.73 m2, and median [IQR] systolic blood pressure was 125 [116-133] mmHg. Prevalence of LVH was 21.4% as defined by LVMI and was not significantly different (p = 0.8) between those with and without HTN, and was similar (21.4%) after excluding those (N = 21) with known cardiac disease. Greater TKV and lower eGFR were directly correlated with greater LVMI (p = .016 and p < .001, respectively). In multiple linear regression models accounting for potential confounders including blood pressure, greater TKV was positively associated with LVM ([Formula: see text] =0.19, p = 0.04). CONCLUSIONS: In a contemporary cohort of ADPKD patients with well-controlled blood pressure, the prevalence of LVH is high, and ADPKD severity as reflected by TKV is independently associated with greater LV mass. These results may suggest a relationship between ADPKD pathophysiology and increased LV mass.


Asunto(s)
Hipertrofia Ventricular Izquierda/epidemiología , Riñón Poliquístico Autosómico Dominante/epidemiología , Riñón Poliquístico Autosómico Dominante/patología , Adulto , Presión Sanguínea , Estudios de Cohortes , Comorbilidad , Ecocardiografía , Femenino , Tasa de Filtración Glomerular , Humanos , Hipertensión/epidemiología , Hipertrofia Ventricular Izquierda/diagnóstico por imagen , Masculino , Persona de Mediana Edad , Tamaño de los Órganos , Gravedad del Paciente , Riñón Poliquístico Autosómico Dominante/diagnóstico por imagen , Riñón Poliquístico Autosómico Dominante/fisiopatología , Prevalencia
14.
BMC Nephrol ; 20(1): 324, 2019 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-31419965

RESUMEN

BACKGROUND: Patients with autosomal dominant polycystic kidney disease (ADPKD) have an increased risk of cardiovascular morbidity and mortality. Impaired left ventricular (LV) global longitudinal strain (GLS) can be a sign of subclinical cardiac dysfunction even in patients with otherwise preserved ejection fraction (EF). Transmitral early filling velocity to early diastolic strain rate (E/SRe) is a novel measure of LV filling pressure, which is often affected early in cardiac disease. METHODS: A total of 110 ADPKD patients not on dialysis were included in this prospective study. All patients underwent an extensive echocardiographic examination including two-dimensional speckle tracking. GLS and strain rates were measured. The distribution of GLS and E/SRe was determined and patient characteristics were compared by median levels of GLS (- 17.8%) and E/SRe (91.4 cm). Twenty healthy participants were included as control group. RESULTS: There was a significantly worse GLS in the ADPKD patients (mean: - 17.8 ± 2.5%) compared to the healthy controls (mean: - 21.9 ± 1.9%), p < 0.001. The same was true for E/SRe (mean: 10.0 ± 0.3 cm) compared to the control group (mean: 6.5 ± 0.3 cm), p < 0.001. In simple logistic regression, male gender (OR: 4.74 [2.10-10.71], p < 0.001), fasting glucose (odds ratio (OR) 1.05 [1.01-1.10], p = 0.024), htTKV (OR: 1.07 [1.01-1.13], p = 0.013), HDL cholesterol (OR: 0.97 [0.94, 0.996], p = 0.025), triglycerides (OR: 1.01 [1.00-1.02], p = 0.039), hemoglobin (OR: 1.50 [1.11-2.04], p = 0.009), and ß-blocker use (OR: 1.07 [1.01, 1.13], p = 0.013) were all associated with higher GLS. After multivariate logistic regression with backward model selection, only male gender (OR: 5.78 [2.27-14.71], p < 0.001) and ß-blocker use (OR: 14.00 [1.60, 122.51], p = 0.017) remained significant. In simple logistic regression models, BMI (OR: 1.11 [1.02-1.20], p = 0.015), systolic blood pressure (OR: 1.03 [1.00-1.06], p = 0.027) and ß-blocker use (OR: 17.12 [2.15-136.20], p = 0.007) were associated with higher E/SRe - a novel measure of left ventricular filling pressure. After backward elimination, only ß-blocker use (OR: 17.22 [2.16, 137.14], p = 0.007) remained significant. CONCLUSION: Higher GLS and E/SRe are common in ADPKD patients, even in patients with preserved eGFR and normal left ventricular EF. GLS and E/SRe may aid in cardiovascular risk stratification in patients with ADPKD as they represent early markers of cardiac dysfunction.


Asunto(s)
Contracción Miocárdica/fisiología , Riñón Poliquístico Autosómico Dominante/fisiopatología , Disfunción Ventricular Izquierda/fisiopatología , Adulto , Estudios de Casos y Controles , Estudios Transversales , Ecocardiografía , Femenino , Humanos , Masculino , Persona de Mediana Edad , Riñón Poliquístico Autosómico Dominante/sangre , Riñón Poliquístico Autosómico Dominante/complicaciones , Estudios Prospectivos , Análisis de Regresión , Factores Sexuales , Volumen Sistólico , Disfunción Ventricular Izquierda/sangre , Disfunción Ventricular Izquierda/diagnóstico por imagen
15.
J Am Soc Nephrol ; 29(10): 2458-2470, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30228150

RESUMEN

In the past, the treatment of autosomal dominant polycystic kidney disease (ADPKD) has been limited to the management of its symptoms and complications. Recently, the US Food and Drug Administration (FDA) approved tolvaptan as the first drug treatment to slow kidney function decline in adults at risk of rapidly progressing ADPKD. Full prescribing information approved by the FDA provides helpful guidelines but does not address practical questions that are being raised by nephrologists, internists, and general practitioners taking care of patients with ADPKD, and by the patients themselves. In this review, we provide practical guidance and discuss steps that require consideration before and after prescribing tolvaptan to patients with ADPKD to ensure that this treatment is implemented safely and effectively. These steps include confirmation of diagnosis; identification of rapidly progressive disease; implementation of basic renal protective measures; counseling of patients on potential benefits and harms; exclusions to use; education of patients on aquaresis and its expected consequences; initiation, titration, and optimization of tolvaptan treatment; prevention of aquaresis-related complications; evaluation and management of liver enzyme elevations; and monitoring of treatment efficacy. Our recommendations are made on the basis of published evidence and our collective experiences during the randomized, clinical trials and open-label extension studies of tolvaptan in ADPKD.


Asunto(s)
Antagonistas de los Receptores de Hormonas Antidiuréticas/uso terapéutico , Riñón Poliquístico Autosómico Dominante/tratamiento farmacológico , Tolvaptán/uso terapéutico , Algoritmos , Antagonistas de los Receptores de Hormonas Antidiuréticas/administración & dosificación , Antagonistas de los Receptores de Hormonas Antidiuréticas/efectos adversos , Protocolos Clínicos , Deshidratación/inducido químicamente , Deshidratación/prevención & control , Progresión de la Enfermedad , Diuresis/efectos de los fármacos , Femenino , Tasa de Filtración Glomerular , Humanos , Hígado/enzimología , Imagen por Resonancia Magnética , Masculino , Selección de Paciente , Riñón Poliquístico Autosómico Dominante/diagnóstico , Riñón Poliquístico Autosómico Dominante/fisiopatología , Factores de Tiempo , Tolvaptán/administración & dosificación , Tolvaptán/efectos adversos , Resultado del Tratamiento
17.
Am J Nephrol ; 47(5): 352-360, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29779024

RESUMEN

BACKGROUND: Metformin inhibits cyclic AMP generation and activates AMP-activated protein kinase (AMPK), which inhibits the cystic fibrosis transmembrane conductance regulator and Mammalian Target of Rapamycin pathways. Together these effects may reduce cyst growth in autosomal dominant polycystic kidney disease (ADPKD). METHODS: A phase II, double-blinded randomized placebo-controlled trial of 26 months duration. Participants will include nondiabetic adults (n = 96) aged 18-60 years, with an estimated glomerular filtration rate (eGFR) ≥50 mL/min/1.73 m2 and ADPKD, recruited from university-based practices in Baltimore and Boston. Participants will be randomized in 1: 1 ratio to metformin or placebo at 500 mg once daily, increased every 2 weeks to a maximum of 1,000 mg twice daily as tolerated. Dose is decreased if eGFR falls to 30-45 mL/min/1.73 m2 and discontinued at eGFR < 30 mL/min/1.73 m2. RESULTS: The primary outcomes are safety, assessed by the rates of hypoglycemia, elevated lactic acid levels, adverse events, and tolerability assessed by the Gastrointestinal Severity Rating Scale and maximum tolerated dose of study medication. Secondary outcomes include changes in total kidney and liver volumes, pain, and health-related quality of life, and changes in urinary metabolomic biomarkers. CONCLUSIONS: Results of this trial will provide important information on the feasibility, safety, and tolerability of long-term use of metformin in patients with -ADPKD and provide preliminary information regarding its efficacy in slowing disease progression. Furthermore, results may support or refute the hypothesis that metformin effects on disease progression are mediated through the activation of the AMPK pathway. These results will be essential for the justification and design of a full-scale efficacy trial.


Asunto(s)
Quistes/tratamiento farmacológico , Hepatopatías/tratamiento farmacológico , Metformina/administración & dosificación , Riñón Poliquístico Autosómico Dominante/tratamiento farmacológico , Adolescente , Adulto , Ensayos Clínicos Fase II como Asunto , Quistes/etiología , Quistes/patología , Progresión de la Enfermedad , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Estudios de Factibilidad , Estudios de Seguimiento , Enfermedades Gastrointestinales/inducido químicamente , Enfermedades Gastrointestinales/epidemiología , Tasa de Filtración Glomerular , Humanos , Hipoglucemia/inducido químicamente , Hipoglucemia/epidemiología , Riñón/diagnóstico por imagen , Riñón/efectos de los fármacos , Riñón/patología , Hígado/diagnóstico por imagen , Hígado/efectos de los fármacos , Hígado/patología , Hepatopatías/etiología , Hepatopatías/patología , Imagen por Resonancia Magnética , Dosis Máxima Tolerada , Metformina/efectos adversos , Persona de Mediana Edad , Estudios Multicéntricos como Asunto , Tamaño de los Órganos/efectos de los fármacos , Placebos/administración & dosificación , Placebos/efectos adversos , Riñón Poliquístico Autosómico Dominante/complicaciones , Riñón Poliquístico Autosómico Dominante/patología , Ensayos Clínicos Controlados Aleatorios como Asunto , Proyectos de Investigación , Adulto Joven
18.
Kidney Int ; 92(5): 1130-1144, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28729032

RESUMEN

Autosomal recessive polycystic kidney disease (OMIM 263200) is a serious condition of the kidney and liver caused by mutations in a single gene, PKHD1. This gene encodes fibrocystin/polyductin (FPC, PD1), a large protein shown by in vitro studies to undergo Notch-like processing. Its cytoplasmic tail, reported to include a ciliary targeting sequence, a nuclear localization signal, and a polycystin-2 binding domain, is thought to traffic to the nucleus after cleavage. We now report a novel mouse line with a triple HA-epitope "knocked-in" to the C-terminus along with lox P sites flanking exon 67, which encodes most of the C-terminus (Pkhd1Flox67HA). The triple HA-epitope has no functional effect as assayed by phenotype and allows in vivo tracking of Fibrocystin. We used the HA tag to identify previously predicted Fibrocystin cleavage products in tissue. In addition, we found that Polycystin-2 fails to co-precipitate with Fibrocystin in kidney samples. Immunofluorescence studies with anti-HA antibodies demonstrate that Fibrocystin is primarily present in a sub-apical location the in kidney, biliary duct, and pancreatic ducts, partially overlapping with the Golgi. In contrast to previous studies, the endogenous protein in the primary cilia was not detectable in mouse tissues. After Cre-mediated deletion, homozygous Pkhd1Δ67 mice are completely normal. Thus, Pkhd1Flox67HA is a valid model to track Pkhd1-derived products containing the C-terminus. Significantly, exon 67 containing the nuclear localization signal and the polycystin-2 binding domain is not essential for Fibrocystin function in our model.


Asunto(s)
Riñón/metabolismo , Riñón Poliquístico Autosómico Recesivo/genética , Dominios Proteicos/genética , Receptores de Superficie Celular/genética , Canales Catiónicos TRPP/metabolismo , Animales , Cilios/metabolismo , Modelos Animales de Enfermedad , Epítopos/genética , Exones/genética , Femenino , Técnica del Anticuerpo Fluorescente , Técnicas de Sustitución del Gen , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Humanos , Riñón/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación , Señales de Localización Nuclear/genética , Señales de Localización Nuclear/metabolismo , Fragmentos de Péptidos/genética , Fenotipo , Riñón Poliquístico Autosómico Recesivo/metabolismo , Receptores de Superficie Celular/metabolismo
19.
Kidney Int ; 89(4): 949-55, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26924047

RESUMEN

Autosomal dominant polycystic kidney disease (ADPKD) is the most common monogenic cause of end-stage renal disease. The molecular pathogenesis of ADPKD is not completely known, and there is no approved therapy. To date, there is limited knowledge concerning the molecular consequences of specific disease-causing mutations. Here we show that the ADPKD missense variant TRPP2(D511V) greatly reduces TRPP2 protein stability, and that TRPP2(D511V) function can be rescued in vivo by small molecules targeting the TRPP2 degradation pathway. Expression of the TRPP2(D511V) protein was significantly reduced compared to wild-type TRPP2. Inhibition of lysosomal degradation of TRPP2(D511V) by the US Food and Drug Administration (FDA)-approved drug chloroquine strongly increased TRPP2 protein levels in vitro. The validation of these results in vivo requires appropriate animal models. However, there are currently no mouse models harboring human PKD2 missense mutations, and screening for chemical rescue of patient mutations in rodent models is time-consuming and expensive. Therefore, we developed a Drosophila melanogaster model expressing the ortholog of TRPP2(D511V) to test chemical rescue of mutant TRPP2 in vivo. Notably, chloroquine was sufficient to improve the phenotype of flies expressing mutant TRPP2. Thus, this proof-of-concept study highlights the potential of directed therapeutic approaches for ADPKD, and provides a rapid-throughput experimental model to screen PKD2 patient mutations and small molecules in vivo.


Asunto(s)
Antirreumáticos/uso terapéutico , Cloroquina/uso terapéutico , Riñón Poliquístico Autosómico Dominante/genética , Canales Catiónicos TRPP/genética , Animales , Antirreumáticos/farmacología , Cloroquina/farmacología , Drosophila melanogaster , Evaluación Preclínica de Medicamentos , Femenino , Células HEK293 , Células HeLa , Humanos , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Masculino , Mutación Missense , Riñón Poliquístico Autosómico Dominante/tratamiento farmacológico , Riñón Poliquístico Autosómico Dominante/metabolismo , Estabilidad Proteica , Canales Catiónicos TRPP/metabolismo
20.
Kidney Int ; 90(1): 90-9, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27165822

RESUMEN

Abnormal proliferation of cyst-lining epithelium and increased intracystic fluid secretion via the cystic fibrosis transmembrane conductance regulator (CFTR) are thought to contribute to cyst growth in autosomal dominant polycystic kidney disease (ADPKD). Histone deacetylase 6 (HDAC6) expression and activity are increased in certain cancers, neurodegenerative diseases, and in Pkd1-mutant renal epithelial cells. Inhibition of HDAC6 activity with specific inhibitors slows cancer growth. Here we studied the effect of tubacin, a specific HDAC6 inhibitor, on cyst growth in polycystic kidney disease. Treatment with tubacin prevented cyst formation in MDCK cells, an in vitro model of cystogenesis. Cyclic AMP stimulates cell proliferation and activates intracystic CFTR-mediated chloride secretion in ADPKD. Treatment with tubacin downregulated cyclic AMP levels, inhibited cell proliferation, and inhibited cyclic AMP-activated CFTR chloride currents in MDCK cells. We also found that tubacin reduced cyst growth by inhibiting proliferation of cyst-lining epithelial cells, downregulated cyclic AMP levels, and improved renal function in a Pkd1-conditional mouse model of ADPKD. Thus, HDAC6 could play a role in cyst formation and could serve as a potential therapeutic target in ADPKD.


Asunto(s)
Anilidas/farmacología , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Células Epiteliales/fisiología , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/metabolismo , Ácidos Hidroxámicos/farmacología , Riñón/efectos de los fármacos , Riñón Poliquístico Autosómico Dominante/metabolismo , Animales , Proliferación Celular/efectos de los fármacos , Cloruros/sangre , Cloruros/metabolismo , AMP Cíclico/sangre , Modelos Animales de Enfermedad , Perros , Regulación hacia Abajo , Células Epiteliales/metabolismo , Femenino , Histona Desacetilasa 6 , Histona Desacetilasas/genética , Humanos , Riñón/enzimología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Riñón Poliquístico Autosómico Dominante/genética , Canales Catiónicos TRPP/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA