Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
World J Nephrol ; 7(5): 108-116, 2018 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-30211029

RESUMEN

AIM: To delineate changes in miRNA expression localized to the peri-cystic local microenvironment (PLM) in an orthologous mouse model of autosomal dominant polycystic kidney disease (ADPKD) (mcwPkd1(nl/nl) ). METHODS: We profiled miRNA expression in the whole kidney and laser captured microdissection (LCM) samples from PLM in mcwPkd1(nl/nl) kidneys with Qiagen miScript 384 HC miRNA PCR arrays. The three times points used are: (1) post-natal (PN) day 21, before the development of trichrome-positive areas; (2) PN28, the earliest sign of trichrome staining; and (3) PN42 following the development of progressive fibrosis. PN21 served as appropriate controls and as the reference time point for comparison of miRNA expression profiles. RESULTS: LCM samples revealed three temporally upregulated miRNAs [2 to 2.75-fold at PN28 and 2.5 to 4-fold (P ≤ 0.05) at PN42] and four temporally downregulated miRNAs [2 to 2.75 fold at PN28 and 2.75 to 5-fold (P ≤ 0.05) at PN42]. Expression of twenty-six miRNAs showed no change until PN42 [six decreased (2.25 to 3.5-fold) (P ≤ 0.05) and 20 increased (2 to 4-fold) (P ≤ 0.05)]. Many critical miRNA changes seen in the LCM samples from PLM were not seen in the contralateral whole kidney. CONCLUSION: Precise sampling with LCM identifies miRNA changes that occur with the initiation and progression of renal interstitial fibrosis (RIF). Identification of the target proteins regulated by these miRNAs will provide new insight into the process of fibrosis and identify unique therapeutic targets to prevent or slow the development and progression of RIF in ADPKD.

2.
World J Nephrol ; 6(4): 188-200, 2017 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-28729967

RESUMEN

AIM: To investigate the therapeutic potential of tesevatinib (TSV), a unique multi-kinase inhibitor currently in Phase II clinical trials for autosomal dominant polycystic kidney disease (ADPKD), in well-defined rodent models of autosomal recessive polycystic kidney disease (ARPKD). METHODS: We administered TSV in daily doses of 7.5 and 15 mg/kg per day by I.P. to the well characterized bpk model of polycystic kidney disease starting at postnatal day (PN) 4 through PN21 to assess efficacy and toxicity in neonatal mice during postnatal development and still undergoing renal maturation. We administered TSV by oral gavage in the same doses to the orthologous PCK model (from PN30 to PN90) to assess efficacy and toxicity in animals where developmental processes are complete. The following parameters were assessed: Body weight, total kidney weight; kidney weight to body weight ratios; and morphometric determination of a cystic index and a measure of hepatic disease. Renal function was assessed by: Serum BUN; creatinine; and a 12 h urinary concentrating ability. Validation of reported targets including the level of angiogenesis and inhibition of angiogenesis (active VEGFR2/KDR) was assessed by Western analysis. RESULTS: This study demonstrates that: (1) in vivo pharmacological inhibition of multiple kinase cascades with TSV reduced phosphorylation of key mediators of cystogenesis: EGFR, ErbB2, c-Src and KDR; and (2) this reduction of kinase activity resulted in significant reduction of renal and biliary disease in both bpk and PCK models of ARPKD. The amelioration of disease by TSV was not associated with any apparent toxicity. CONCLUSION: The data supports the hypothesis that this multi-kinase inhibitor TSV may provide an effective clinical therapy for human ARPKD.

3.
Front Pediatr ; 5: 77, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28473970

RESUMEN

Cystic kidney diseases comprise a varied collection of hereditary disorders, where renal cysts comprise a major element of their pleiotropic phenotype. In pediatric patients, the term polycystic kidney disease (PKD) commonly refers to two specific hereditary diseases, autosomal recessive polycystic kidney disease (ARPKD) and autosomal dominant polycystic kidney disease (ADPKD). Remarkable progress has been made in understanding the complex molecular and cellular mechanisms of renal cyst formation in ARPKD and ADPKD. One of the most important discoveries is that both the genes and proteins products of ARPKD and ADPKD interact in a complex network of genetic and functional interactions. These interactions and the shared phenotypic abnormalities of ARPKD and ADPKD, the "cystic phenotypes" suggest that many of the therapies developed and tested for ADPKD may be effective in ARPKD as well. Successful therapeutic interventions for childhood PKD will, therefore, be guided by knowledge of these molecular interactions, as well as a number of clinical parameters, such as the stage of the disease and the rate of disease progression.

4.
Pediatr Res ; 75(1-2): 148-57, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24336431

RESUMEN

Autosomal dominant polycystic kidney disease (ADPKD) and autosomal recessive polycystic kidney disease (ARPKD) are significant causes of morbidity and mortality in children and young adults. ADPKD, with an incidence of 1:400 to 1:1,000, affects more than 13 million individuals worldwide and is a major cause of end-stage renal disease in adults. However, symptomatic disease is increasingly recognized in children. ARPKD is a dual-organ hepatorenal disease with an incidence of 1:20,000 to 1:40,000 and a heterozygote carrier rate of 1 in 70. Currently, no clinically significant disease-specific therapy exists for ADPKD or ARPKD. The genetic basis of both ADPKD and ARPKD have been identified, and delineation of the basic molecular and cellular pathophysiology has led to the discovery that abnormal ADPKD and ARPKD gene products interact to create "polycystin complexes" located at multiple sites within affected cells. The extracellular matrix and vessels produce a variety of soluble factors that affect the biology of adjacent cells in many dynamic ways. This review will focus on the molecular and cellular bases of the abnormal cystic phenotype and discuss the clinical translation of such basic data into new therapies that promise to alter the natural history of disease for children with genetic PKDs.


Asunto(s)
Riñón/fisiopatología , Riñón Poliquístico Autosómico Dominante/fisiopatología , Riñón Poliquístico Autosómico Recesivo/fisiopatología , Adolescente , Factores de Edad , Animales , Niño , Preescolar , Predisposición Genética a la Enfermedad , Humanos , Incidencia , Lactante , Riñón/metabolismo , Fenotipo , Riñón Poliquístico Autosómico Dominante/epidemiología , Riñón Poliquístico Autosómico Dominante/genética , Riñón Poliquístico Autosómico Dominante/metabolismo , Riñón Poliquístico Autosómico Dominante/terapia , Riñón Poliquístico Autosómico Recesivo/epidemiología , Riñón Poliquístico Autosómico Recesivo/genética , Riñón Poliquístico Autosómico Recesivo/metabolismo , Riñón Poliquístico Autosómico Recesivo/terapia , Pronóstico , Factores de Riesgo , Transducción de Señal
5.
J Am Soc Nephrol ; 24(12): 2010-22, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23990677

RESUMEN

Past efforts to pharmacologically disrupt the development and growth of renal cystic lesions focused primarily on normalizing the activity of a specific signaling molecule, but the effects of stimulating apoptosis in the proliferating epithelial cells have not been well studied. Although benign, ADPKD renal cysts created by the sustained proliferation of epithelial cells resemble tumors, and malignant cell death can be achieved by cotreatment with TNF-α and a mimetic of second mitochondria-derived activator of caspase (Smac). Notably, TNF-α accumulates to high levels in ADPKD cyst fluid. Here, we report that an Smac-mimetic selectively induces TNF-α-dependent cystic renal epithelial cell death, leading to the removal of cystic epithelial cells from renal tissues and delaying cyst formation. In vitro, a Smac-mimetic (GT13072) induced the degradation of cIAP1 that is required but not sufficient for cell death. Cotreatment with TNF-α augmented the formation and activation of the RIPK1-dependent death complex and the degradation and cleavage of FLIP, an inhibitor of caspase-8, in renal cystic epithelial cells. This approach produced death specifically in Pkd1 mutant epithelial cells, with no effect on normal renal epithelial cells. Moreover, treatment with the Smac-mimetic slowed cyst and kidney enlargement and preserved renal function in two genetic strains of mice with Pkd1 mutations. Thus, our mechanistic data characterize an apoptotic pathway, activated by the selective synergy of an Smac-mimetic and TNF-α in renal cyst fluid, that attenuates cyst development, providing an innovative translational platform for the rational development of novel therapeutics for ADPKD.


Asunto(s)
Apoptosis/efectos de los fármacos , Proteínas Portadoras/fisiología , Dipéptidos/farmacología , Células Epiteliales/efectos de los fármacos , Indoles/farmacología , Péptidos y Proteínas de Señalización Intracelular/fisiología , Proteínas Mitocondriales/fisiología , Riñón Poliquístico Autosómico Dominante/tratamiento farmacológico , Riñón Poliquístico Autosómico Dominante/patología , Animales , Apoptosis/fisiología , Proteínas Reguladoras de la Apoptosis , Proteínas Portadoras/antagonistas & inhibidores , Proteínas Portadoras/genética , Células Cultivadas , Células Epiteliales/citología , Femenino , Humanos , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Péptidos y Proteínas de Señalización Intracelular/genética , Túbulos Renales Proximales/citología , Ratones , Ratones Mutantes , Proteínas Mitocondriales/antagonistas & inhibidores , Proteínas Mitocondriales/genética , FN-kappa B/metabolismo , Riñón Poliquístico Autosómico Dominante/genética , Embarazo , Canales Catiónicos TRPP/genética , Factor de Necrosis Tumoral alfa/fisiología
6.
J Clin Invest ; 123(7): 3084-98, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23778143

RESUMEN

Autosomal-dominant polycystic kidney disease (ADPKD) is caused by mutations in either PKD1 or PKD2 and is characterized by the development of multiple bilateral renal cysts that replace normal kidney tissue. Here, we used Pkd1 mutant mouse models to demonstrate that the nicotinamide adenine dinucleotide-dependent (NAD-dependent) protein deacetylase sirtuin 1 (SIRT1) is involved in the pathophysiology of ADPKD. SIRT1 was upregulated through c-MYC in embryonic and postnatal Pkd1-mutant mouse renal epithelial cells and tissues and could be induced by TNF-α, which is present in cyst fluid during cyst development. Double conditional knockouts of Pkd1 and Sirt1 demonstrated delayed renal cyst formation in postnatal mouse kidneys compared with mice with single conditional knockout of Pkd1. Furthermore, treatment with a pan-sirtuin inhibitor (nicotinamide) or a SIRT1-specific inhibitor (EX-527) delayed cyst growth in Pkd1 knockout mouse embryonic kidneys, Pkd1 conditional knockout postnatal kidneys, and Pkd1 hypomorphic kidneys. Increased SIRT1 expression in Pkd1 mutant renal epithelial cells regulated cystic epithelial cell proliferation through deacetylation and phosphorylation of Rb and regulated cystic epithelial cell death through deacetylation of p53. This newly identified role of SIRT1 signaling in cystic renal epithelial cells provides the opportunity to develop unique therapeutic strategies for ADPKD.


Asunto(s)
Carbazoles/farmacología , Riñón/patología , Niacinamida/farmacología , Riñón Poliquístico Autosómico Dominante/tratamiento farmacológico , Sirtuina 1/antagonistas & inhibidores , Acetilación , Animales , Apoptosis , Carbazoles/uso terapéutico , Proliferación Celular , Células Cultivadas , Evaluación Preclínica de Medicamentos , Células Epiteliales/efectos de los fármacos , Células Epiteliales/enzimología , Femenino , Regulación Enzimológica de la Expresión Génica , Riñón/efectos de los fármacos , Riñón/enzimología , Masculino , Ratones , Ratones Noqueados , Fosforilación , Riñón Poliquístico Autosómico Dominante/enzimología , Proteína Quinasa C/genética , Procesamiento Proteico-Postraduccional , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteína de Retinoblastoma/metabolismo , Sirtuina 1/genética , Sirtuina 1/metabolismo , Factor de Necrosis Tumoral alfa/fisiología , Proteína p53 Supresora de Tumor/metabolismo
7.
Pediatr Transplant ; 17(4): 328-35, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23593929

RESUMEN

Improved neonatal medical care and renal replacement technology have improved the long-term survival of patients with ARPKD. Ten-yr survival of those surviving the first year of life is reported to be 82% and is continuing to improve further. However, despite increases in overall survival and improved treatment of systemic hypertension and other complications of their renal disease, nearly 50% of survivors will develop ESRD within the first decade of life. In addition to renal pathology, patients with ARPKD develop ductal plate malformations with cystic dilation of intra- and extrahepatic bile ducts resulting in CHF and Caroli syndrome. Many patients with CHF will develop portal hypertension with resulting esophageal varices, splenomegaly, hypersplenism, protein losing enteropathy, and gastrointestinal bleeding. Management of portal hypertension may require EBL of esophageal varices or porto-systemic shunting. Complications of hepatic involvement can include ascending cholangitis, cholestasis with malabsorption of fat-soluble vitamins, and rarely benign or malignant liver tumors. Patients with ARPKD who eventually reach ESRD, and ultimately require kidney transplantation, present a unique set of complications related to their underlying hepato-biliary disease. In this review, we focus on new approaches to these challenging patients, including the indications for liver transplantation in ARPKD patients with severe chronic kidney disease awaiting kidney transplant. While survival in patients with ARPKD and isolated kidney transplant is comparable to that of age-matched pediatric patients who have received kidney transplants due to other primary renal diseases, 64-80% of the mortality occurring in ARPKD kidney transplant patients is attributed to cholangitis/sepsis, which is related to their hepato-biliary disease. Recent data demonstrate that surgical mortality among pediatric liver transplant recipients is decreased to <10% at one yr. The immunosuppressive regimen used for kidney transplant recipients is adequate for most liver transplant recipients. We therefore suggest that in a select group of ARPKD patients with recurrent cholangitis or complications of portal hypertension, combined liver-kidney transplant is a viable option. Although further study is necessary to confirm our approach, we believe that combined liver-kidney transplantation can potentially decrease overall mortality and morbidity in carefully selected ARPKD patients with ESRD and clinically significant CHF.


Asunto(s)
Trasplante de Riñón/métodos , Trasplante de Hígado/métodos , Riñón Poliquístico Autosómico Recesivo/terapia , Enfermedad de Caroli/complicaciones , Niño , Colangitis/complicaciones , Colestasis/complicaciones , Humanos , Hipertensión Portal/complicaciones , Inmunosupresores/uso terapéutico , Fallo Renal Crónico/complicaciones , Fallo Renal Crónico/terapia , Hepatopatías/complicaciones , Fallo Hepático/complicaciones , Fallo Hepático/terapia , Neoplasias Hepáticas/complicaciones , Selección de Paciente , Riñón Poliquístico Autosómico Recesivo/complicaciones , Riesgo
8.
Proc Natl Acad Sci U S A ; 109(52): 21462-7, 2012 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-23236168

RESUMEN

Polycystic kidney diseases are the most common genetic diseases that affect the kidney. There remains a paucity of information regarding mechanisms by which G proteins are regulated in the context of polycystic kidney disease to promote abnormal epithelial cell expansion and cystogenesis. In this study, we describe a functional role for the accessory protein, G-protein signaling modulator 1 (GPSM1), also known as activator of G-protein signaling 3, to act as a modulator of cyst progression in an orthologous mouse model of autosomal dominant polycystic kidney disease (ADPKD). A complete loss of Gpsm1 in the Pkd1(V/V) mouse model of ADPKD, which displays a hypomorphic phenotype of polycystin-1, demonstrated increased cyst progression and reduced renal function compared with age-matched cystic Gpsm1(+/+) and Gpsm1(+/-) mice. Electrophysiological studies identified a role by which GPSM1 increased heteromeric polycystin-1/polycystin-2 ion channel activity via Gßγ subunits. In summary, the present study demonstrates an important role for GPSM1 in controlling the dynamics of cyst progression in an orthologous mouse model of ADPKD and presents a therapeutic target for drug development in the treatment of this costly disease.


Asunto(s)
Proteínas Portadoras/metabolismo , Progresión de la Enfermedad , Riñón Poliquístico Autosómico Dominante/metabolismo , Riñón Poliquístico Autosómico Dominante/patología , Animales , Modelos Animales de Enfermedad , Fenómenos Electrofisiológicos , Técnica del Anticuerpo Fluorescente , Genotipo , Inhibidores de Disociación de Guanina Nucleótido , Riñón/metabolismo , Riñón/patología , Riñón/fisiopatología , Pruebas de Función Renal , Ratones , Riñón Poliquístico Autosómico Dominante/fisiopatología , Transporte de Proteínas , Canales Catiónicos TRPP/metabolismo
9.
PLoS One ; 7(11): e49418, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23152903

RESUMEN

We present for the first time that histone deacetylase 6 (HDAC6) regulates EGFR degradation and trafficking along microtubules in Pkd1 mutant renal epithelial cells. HDAC6, the microtubule-associated α-tubulin deacetylase, demonstrates increased expression and activity in Pkd1 mutant mouse embryonic kidney cells. Targeting HDAC6 with a general HDAC inhibitor, trichostatin (TSA), or a specific HDAC6 inhibitor, tubacin, increased the acetylation of α-tubulin and downregulated the expression of EGFR in Pkd1 mutant renal epithelial cells. HDAC6 was co-localized with EGF induced endocytic EGFR and endosomes, respectively. Inhibition of the activity of HDAC6 accelerated the trafficking of EGFR from early endosomes to late endosomes along the microtubules. Without EGF stimulation EGFR was randomly distributed while after stimulation with EGF for 30 min, EGFR was accumulated around α-tubulin labeled microtubule bundles. These data suggested that the Pkd1 mutation induced upregulation of HDAC6 might act to slow the trafficking of EGFR from early endosomes to late endosomes along the microtubules for degradation through deacetylating α-tubulin. In addition, inhibition of HDAC activity decreased the phosphorylation of ERK1/2, the downstream target of EGFR axis, and normalized EGFR localization from apical to basolateral in Pkd1 knockout mouse kidneys. Thus, targeting HDAC6 to downregulate EGFR activity may provide a potential therapeutic approach to treat polycystic kidney disease.


Asunto(s)
Endocitosis , Células Epiteliales/citología , Receptores ErbB/metabolismo , Histona Desacetilasas/metabolismo , Riñón/citología , Proteolisis , Anilidas/farmacología , Animales , Endocitosis/efectos de los fármacos , Endosomas/efectos de los fármacos , Endosomas/metabolismo , Factor de Crecimiento Epidérmico/farmacología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/enzimología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Células HEK293 , Histona Desacetilasa 6 , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Ácidos Hidroxámicos/farmacología , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Ratones , Microtúbulos/efectos de los fármacos , Microtúbulos/metabolismo , Nocodazol/farmacología , Fosforilación/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Proteolisis/efectos de los fármacos , ARN Interferente Pequeño/metabolismo , Canales Catiónicos TRPP/genética , Tubulina (Proteína)/metabolismo , Regulación hacia Arriba/efectos de los fármacos
10.
Physiol Genomics ; 44(15): 741-53, 2012 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-22669842

RESUMEN

Human data and animal models of autosomal recessive polycystic kidney disease (ARPKD) suggest that genetic factors modulate the onset and severity of the disease. We report here for the first time that ARPKD susceptibility is attenuated by introgressing the mutated Pkhd1 disease allele from the polycystic kidney (PCK) rat onto the FHH (Fawn-Hooded Hypertensive) genetic background. Compared with PCK, the FHH.Pkhd1 strain had significantly decreased renal cyst formation that coincided with a threefold reduction in mean kidney weights. Further analysis revealed that the FHH. Pkhd1 is protected from increased blood pressure as well as elevated plasma creatinine and blood urea nitrogen levels. On the other hand, liver weight and biliary cystogenesis revealed no differences between PCK and FHH.Pkdh1, indicating that genes within the FHH genetic background prevent the development of renal, but not hepatic, manifestations of ARPKD. Microarray expression analysis of kidneys from 30-day-old PCK rats revealed increased expression of genes previously identified in PKD renal expression profiles, such as inflammatory response, extracellular matrix synthesis, and cell proliferation genes among others, whereas the FHH.Pkhd1 did not show activation of these common markers of disease. This newly developed strain can serve as a tool to map modifier genes for renal disease in ARPKD and provides further insight into disease variability and pathophysiology.


Asunto(s)
Modelos Animales de Enfermedad , Genes Modificadores/genética , Riñón Poliquístico Autosómico Recesivo/genética , Animales , Presión Sanguínea , Regulación hacia Abajo/genética , Femenino , Perfilación de la Expresión Génica , Redes Reguladoras de Genes/genética , Humanos , Riñón/metabolismo , Riñón/patología , Pruebas de Función Renal , Hígado/metabolismo , Hígado/patología , Masculino , Mutación/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Tamaño de los Órganos , Riñón Poliquístico Autosómico Recesivo/fisiopatología , Ratas , Receptores de Superficie Celular/genética , Programas Informáticos , Regulación hacia Arriba/genética
12.
FASEB J ; 25(6): 1844-55, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21343176

RESUMEN

The intracellular mechanisms underlying renal tubular epithelial cell proliferation and tubular repair following ischemia-reperfusion injury (IRI) remain poorly understood. In this report, we demonstrate that activator of G-protein signaling 3 (AGS3), an unconventional receptor-independent regulator of heterotrimeric G-protein function, influences renal tubular regeneration following IRI. In rat kidneys exposed to IRI, there was a temporal induction in renal AGS3 protein expression that peaked 72 h after reperfusion and corresponded to the repair and recovery phase following ischemic injury. Renal AGS3 expression was localized predominantly to the recovering outer medullary proximal tubular cells and was highly coexpressed with Ki-67, a marker of cell proliferation. Kidneys from mice deficient in the expression of AGS3 exhibited impaired renal tubular recovery 7 d following IRI compared to wild-type AGS3-expressing mice. Mechanistically, genetic knockdown of endogenous AGS3 mRNA and protein in renal tubular epithelial cells reduced cell proliferation in vitro. Similar reductions in renal tubular epithelial cell proliferation were observed following incubation with gallein, a selective inhibitor of Gßγ subunit activity, and lentiviral overexpression of the carboxyl-terminus of G-protein-coupled receptor kinase 2 (GRK2ct), a scavenger of Gßγ subunits. In summary, these data suggest that AGS3 acts through a novel receptor-independent mechanism to facilitate renal tubular epithelial cell proliferation and renal tubular regeneration.


Asunto(s)
Lesión Renal Aguda/metabolismo , Proteínas Portadoras/metabolismo , Túbulos Renales/fisiología , Regeneración/fisiología , Daño por Reperfusión/metabolismo , Lesión Renal Aguda/patología , Animales , Proteínas Portadoras/genética , Regulación de la Expresión Génica , Genotipo , Inhibidores de Disociación de Guanina Nucleótido , Antígeno Ki-67/genética , Antígeno Ki-67/metabolismo , Túbulos Renales/metabolismo , Túbulos Renales/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratas , Ratas Sprague-Dawley , Daño por Reperfusión/genética , Factores de Tiempo
13.
Pediatr Nephrol ; 26(5): 675-92, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21046169

RESUMEN

A number of syndromic disorders have renal cysts as a component of their phenotypes. These disorders can generally be distinguished from autosomal dominant polycystic kidney disease (ADPKD) and autosomal recessive polycystic kidney disease (ARPKD) by imaging studies of their characteristic, predominantly non-renal associated abnormalities. Therefore, a major distinction in the differential diagnosis of enlarge echogenic kidneys is delineating ARPKD from ADPKD. ADPKD and ARPKD can be diagnosed by imaging the kidney with ultrasound, computed tomography, or magnetic resonance imaging (MRI), although ultrasound is still the method of choice for diagnosis in utero and in young children due to ease of use, cost, and safety. Differences in ultrasound characteristics, the presence or absence of associated extrarenal abnormalities, and the screening of the parents >40 years of age usually allow the clinician to make an accurate diagnosis. Early diagnosis of ADPKD and ARPKD affords the opportunity for maximal anticipatory care (i.e. blood pressure control) and in the not-too-distant future, the opportunity to benefit from new therapies currently being developed. If results are equivocal, genetic testing is available for both ARPKD and ADPKD. Specialized centers are now offering preimplantation genetic diagnosis and in vitro fertilization for parents who have previously had a child with ARPKD. For ADPKD patients, a number of therapeutic interventions are currently in clinical trial and may soon be available.


Asunto(s)
Enfermedades Renales Poliquísticas/diagnóstico , Enfermedades Renales Poliquísticas/terapia , Niño , Diagnóstico Precoz , Humanos
14.
J Assist Reprod Genet ; 27(7): 397-407, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20490649

RESUMEN

PURPOSE: To develop a reliable preimplantation genetic diagnosis protocol for couples who both carry a mutant PKHD1 gene wishing to conceive children unaffected with autosomal recessive polycystic kidney disease (ARPKD). METHODS: Development of a unique protocol for preimplantation genetic testing using whole genome amplification of single blastomeres by multiple displacement amplification (MDA), and haplotype analysis with novel short tandem repeat (STR) markers from the PKHD1 gene and flanking sequences, and a case report of successful utilization of the protocol followed by successful IVF resulting in the birth of an infant unaffected with ARPKD. RESULTS: We have developed 20 polymorphic STR markers suitable for linkage analysis of ARPKD. These linked STR markers have enabled unambiguous identification of the PKHD1 haplotypes of embryos produced by at-risk couples. CONCLUSIONS: We have developed a reliable protocol for preimplantation genetic diagnosis of ARPKD using single-cell MDA products for PKHD1 haplotyping.


Asunto(s)
Riñón Poliquístico Autosómico Recesivo/diagnóstico , Reacción en Cadena de la Polimerasa/métodos , Diagnóstico Preimplantación/métodos , Receptores de Superficie Celular/genética , Femenino , Marcadores Genéticos/genética , Haplotipos , Humanos , Lactante , Repeticiones de Microsatélite/genética , Riñón Poliquístico Autosómico Recesivo/genética , Embarazo
15.
Am J Physiol Renal Physiol ; 297(3): F662-70, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19570883

RESUMEN

20-Hydroxyeicosatetraenoic acid (20-HETE) has been reported to promote mitogenicity in a variety of cell types, including renal epithelial cells. However, the signal transduction pathways activated by 20-HETE have not been fully defined. The present study evaluated the effects of 20-HETE and its more stable agonist analogs 20-hydroxyeicosa-5(Z),14(Z)-dienoic acid (5,14-20-HEDE) and N-[20-hydroxyeicosa-5(Z),14(Z)-dienoyl]glycine (5,14-20-HEDGE) on the Raf/MEK/ERK and phosphatidylinositol 3-kinase (PI3K)-Akt pathway in LLC-PK(1) renal epithelial cells. 20-HETE (20 microM) increased phosphorylation of Raf-1 (2.5 +/- 0.2-fold), MEK1/2 (6.3 +/- 1.6-fold), and ERK1/2 (5.8 +/- 0.3-fold) compared with vehicle-treated cells. Similarly, the 20-HETE analogs also strongly activated ERK1/2 in a Raf-1- and MEK1/2-dependent manner. Moreover, 5,14-20-HEDE increased Akt phosphorylation by 2.2 +/- 0.3-fold. 20-HETE and 5,14-20-HEDE also promoted activation (Y1086) of epidermal growth factor receptor (EGFR; Y1086) by 1.9 +/- 0.2- and 2.5 +/- 0.2-fold, respectively. These effects were completely blocked by the EGFR inhibitor EKB-569 (0.1 microM). Moreover, EKB-569 (0.1 microM), as well as a c-Src inhibitor, SKI-606 (0.05 microM), completely abolished the 20-HETE-mediated activation of the Raf/MEK/ERK and PI3K-Akt pathways. Blockade of PKC with bisindolylmaleimide I had no effect on 20-HETE-induced ERK1/2 activation. This study demonstrated that 20-HETE activated the Raf/MEK/ERK and Akt pathways in renal epithelial cells secondary to the activation of c-Src and EGFR.


Asunto(s)
Células Epiteliales/enzimología , Receptores ErbB/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Ácidos Hidroxieicosatetraenoicos/metabolismo , Riñón/enzimología , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Proteínas Proto-Oncogénicas c-raf/metabolismo , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo , Transducción de Señal , Animales , Células Epiteliales/efectos de los fármacos , Receptores ErbB/antagonistas & inhibidores , Riñón/efectos de los fármacos , Células LLC-PK1 , Lipopéptidos/metabolismo , MAP Quinasa Quinasa 1/metabolismo , MAP Quinasa Quinasa 2/metabolismo , Masculino , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas pp60(c-src)/antagonistas & inhibidores , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Porcinos , Factores de Tiempo
16.
Am J Physiol Renal Physiol ; 296(3): F575-82, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19129252

RESUMEN

20-Hydroxyeicosatetraenoic acid (20-HETE) has been implicated as a potential mediator in epithelial cell proliferation and cyst formation in polycystic kidney disease (PKD). In the present study, we studied the effects of chronic blockade of 20-HETE synthesis in an orthologous rodent model of autosomal recessive polycystic kidney disease (ARPKD), the PCK rat. RT-PCR analysis indicated that the expression of CYP4A1, CYP4A2, CYP4A3, and CYP4A8 mRNA was increased two- to fourfold in cystic PCK compared with noncystic Sprague-Dawley rat kidneys. Daily administration of a 20-HETE synthesis inhibitor, HET-0016 (10 mg x kg(-1) x day(-1) ip) for 4-7 wk significantly reduced kidney size by 24% from 4.95 +/- 0.19 g in vehicle-treated PCK rats to 3.76 +/- 0.15 g (n = 4). Collecting tubule morphometric cystic indices were reduced in HET-0016-treated PCK rats (2.1 +/- 0.2; n = 4) compared with vehicle-treated PCK rats (4.4 +/- 0.1; n = 4). The cellular mechanism by which 20-HETE may play a role in cyst formation has not been well characterized, but there was a significantly lower (P < 0.05) level of intracellular cAMP and decreased phosphorylation (activation) of ERK1/2 protein in PCK rat kidneys (n = 3) treated with HET-0016 . These studies indicate a potential role of 20-HETE in cyst formation in the orthologous rodent PCK model of ARPKD.


Asunto(s)
Ácidos Hidroxieicosatetraenoicos/metabolismo , Riñón/patología , Riñón Poliquístico Autosómico Recesivo/metabolismo , Ácido 8,11,14-Eicosatrienoico/análogos & derivados , Ácido 8,11,14-Eicosatrienoico/metabolismo , Amidinas/farmacología , Animales , AMP Cíclico/metabolismo , Citocromo P-450 CYP4A/antagonistas & inhibidores , Modelos Animales de Enfermedad , Activación Enzimática , Compuestos Epoxi/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Masculino , Microsomas/metabolismo , Tamaño de los Órganos , Riñón Poliquístico Autosómico Recesivo/patología , Isoformas de Proteínas/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley
17.
J Am Soc Nephrol ; 19(10): 1929-39, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18596124

RESUMEN

Polycystic kidney diseases are characterized by abnormal proliferation of renal epithelial cells. In this study, the role of 20-hydroxyeicosatetraenoic acid (20-HETE), an endogenous cytochrome P450 metabolite of arachidonic acid with mitogenic properties, was evaluated in cystic renal disease. Daily administration of HET-0016, an inhibitor of 20-HETE synthesis, significantly reduced kidney size by half in the BPK mouse model of autosomal recessive polycystic kidney disease. In addition, compared with untreated BPK mice, this treatment significantly reduced collecting tubule cystic indices and approximately doubled survival. For evaluation of the role of 20-HETE as a mediator of epithelial cell proliferation, principal cells isolated from cystic BPK and noncystic Balb/c mice were genetically modified using lentiviral vectors. Noncystic Balb/c cells overproducing Cyp4a12 exhibited a four- to five-fold increase in cell proliferation compared with control Balb/c cells, and this increase was completely abolished when 20-HETE synthesis was inhibited; therefore, this study suggests that 20-HETE mediates proliferation of epithelial cells in the formation of renal cysts.


Asunto(s)
Proliferación Celular , Células Epiteliales/fisiología , Ácidos Hidroxieicosatetraenoicos/fisiología , Enfermedades Renales Poliquísticas/etiología , Enfermedades Renales Poliquísticas/patología , Amidinas/farmacología , Animales , Técnicas de Cultivo de Célula , Proliferación Celular/efectos de los fármacos , Sistema Enzimático del Citocromo P-450/metabolismo , Sistema Enzimático del Citocromo P-450/fisiología , Familia 4 del Citocromo P450 , Modelos Animales de Enfermedad , Ácidos Hidroxieicosatetraenoicos/antagonistas & inhibidores , Ratones , Ratones Endogámicos BALB C , Enfermedades Renales Poliquísticas/metabolismo
18.
J Am Soc Nephrol ; 19(7): 1331-41, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18385429

RESUMEN

Despite identification of the genes responsible for autosomal dominant polycystic kidney disease (PKD) and autosomal recessive PKD (ARPKD), the precise functions of their cystoprotein products remain unknown. Recent data suggested that multimeric cystoprotein complexes initiate aberrant signaling cascades in PKD, and common components of these signaling pathways may be therapeutic targets. This study identified c-Src (pp60(c-Src)) as one such common signaling intermediate and sought to determine whether Src activity plays a role in cyst formation. With the use of the nonorthologous BPK murine model and the orthologous PCK rat model of ARPKD, greater Src activity was found to correlate with disease progression. Inhibition of Src activity with the pharmacologic inhibitor SKI-606 resulted in amelioration of renal cyst formation and biliary ductal abnormalities in both models. Furthermore, the effects of Src inhibition in PCK kidneys suggest that the ErbB2 and B-Raf/MEK/ERK pathways are involved in Src-mediated signaling in ARPKD and that this occurs without reducing elevated cAMP. These data suggest that Src inhibition may provide therapeutic benefit in PKD.


Asunto(s)
Compuestos de Anilina/uso terapéutico , Receptores ErbB/metabolismo , Glicoproteínas/metabolismo , Nitrilos/uso terapéutico , Riñón Poliquístico Autosómico Recesivo/metabolismo , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo , Quinolinas/uso terapéutico , Compuestos de Anilina/farmacología , Compuestos de Anilina/toxicidad , Animales , AMP Cíclico/metabolismo , Progresión de la Enfermedad , Receptores ErbB/antagonistas & inhibidores , Glicoproteínas/antagonistas & inhibidores , Riñón/patología , Pruebas de Función Renal , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos BALB C , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Nitrilos/farmacología , Nitrilos/toxicidad , Tamaño de los Órganos/efectos de los fármacos , Riñón Poliquístico Autosómico Recesivo/tratamiento farmacológico , Riñón Poliquístico Autosómico Recesivo/patología , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Proteínas Proto-Oncogénicas pp60(c-src)/antagonistas & inhibidores , Quinolinas/farmacología , Quinolinas/toxicidad , Ratas , Ratas Sprague-Dawley , Receptor ErbB-2
19.
Pediatr Clin North Am ; 53(5): 889-909, ix, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17027616

RESUMEN

This article cannot comprehensively cover the enormous strides made in defining the molecular and cellular basis of renal cystic diseases over the last decade. Therefore, it provides a brief overview and categorization of inherited, developmental, and acquired renal cystic diseases, providing a relevant, up-to-date bibliography as well as a useful list of informative Internet Web sites. Its major focus is the translational biology of polycystic kidney disease. It demonstrates how emerging molecular and cellular knowledge of the pathophysiology of particular diseases such as autosomal dominant polycystic kidney disease (ADPKD) and autosomal recessive polycystic kidney disease (ADPKD) can translate into innovative therapeutic insights.


Asunto(s)
Enfermedades Renales Quísticas/genética , Enfermedades Renales Quísticas/fisiopatología , Animales , Apoptosis , División Celular/genética , División Celular/fisiología , Líquido Quístico/metabolismo , Receptores ErbB/metabolismo , Matriz Extracelular/metabolismo , Matriz Extracelular/patología , Sustancias de Crecimiento/biosíntesis , Humanos , Internet , Enfermedades Renales Quísticas/diagnóstico , Proteínas Proto-Oncogénicas/biosíntesis , Transducción de Señal/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...