Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Am J Physiol Gastrointest Liver Physiol ; 324(5): G404-G414, 2023 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-36880660

RESUMEN

Systemic and portal hypertension, liver fibrosis, and hepatomegaly are manifestations associated with autosomal recessive polycystic kidney disease (ARPKD), which is caused by malfunctions of fibrocystin/polyductin (FPC). The goal is to understand how liver pathology occurs and to devise therapeutic strategies to treat it. We injected 5-day-old Pkhd1del3-4/del3-4 mice for 1 mo with the cystic fibrosis transmembrane conductance regulator (CFTR) modulator VX-809 designed to rescue processing and trafficking of CFTR folding mutants. We used immunostaining and immunofluorescence techniques to evaluate liver pathology. We assessed protein expression via Western blotting. We detected abnormal biliary ducts consistent with ductal plate abnormalities, as well as a greatly increased proliferation of cholangiocytes in the Pkhd1del3-4/del3-4 mice. CFTR was present in the apical membrane of cholangiocytes and increased in the Pkhd1del3-4/del3-4 mice, consistent with a role for apically located CFTR in enlarged bile ducts. Interestingly, we also found CFTR in the primary cilium, in association with polycystin (PC2). Localization of CFTR and PC2 and overall length of the cilia were increased in the Pkhd1del3-4/del3-4 mice. In addition, several of the heat shock proteins; 27, 70, and 90 were upregulated, suggesting that global changes in protein processing and trafficking had occurred. We found that a deficit of FPC leads to bile duct abnormalities, enhanced cholangiocyte proliferation, and misregulation of heat shock proteins, which all returned toward wild type (WT) values following VX-809 treatment. These data suggest that CFTR correctors can be useful as therapeutics for ARPKD. Given that these drugs are already approved for use in humans, they can be fast-tracked for clinical use.NEW & NOTEWORTHY ARPKD is a multiorgan genetic disorder resulting in newborn morbidity and mortality. There is a critical need for new therapies to treat this disease. We show that persistent cholangiocytes proliferation occurs in a mouse model of ARPKD along with mislocalized CFTR and misregulated heat shock proteins. We found that VX-809, a CFTR modulator, inhibits proliferation and limits bile duct malformation. The data provide a therapeutic pathway for strategies to treat ADPKD.


Asunto(s)
Riñón Poliquístico Autosómico Recesivo , Humanos , Ratones , Animales , Riñón Poliquístico Autosómico Recesivo/tratamiento farmacológico , Riñón Poliquístico Autosómico Recesivo/genética , Riñón Poliquístico Autosómico Recesivo/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Receptores de Superficie Celular/metabolismo , Cirrosis Hepática/complicaciones , Proteínas de Choque Térmico/metabolismo
2.
BMC Nephrol ; 24(1): 33, 2023 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-36782137

RESUMEN

PURPOSE: Autosomal recessive polycystic kidney disease (ARPKD) is a hereditary condition characterized by massive kidney enlargement and developmental liver defects. Potential consequences during childhood include the need for kidney replacement therapy (KRT). We report the design of 2 ongoing clinical trials (Study 204, Study 307) to evaluate safety, tolerability, and efficacy of tolvaptan in children with ARPKD. METHODS: Both trials are of multinational, multicenter, open-label design. Age range at enrollment is 28 days to < 12 weeks in Study 204 and 28 days to < 18 years in Study 307. Subjects in both studies must have a clinical diagnosis of ARPKD, and those in Study 204 must additionally have signs indicative of risk of rapid progression to KRT, namely, all of: nephromegaly, multiple kidney cysts or increased kidney echogenicity suggesting microcysts, and oligohydramnios or anhydramnios. Target enrollment is 20 subjects for Study 204 and ≥ 10 subjects for Study 307. RESULTS: Follow-up is 24 months in Study 204 (with optional additional treatment up to 36 months) and 18 months in Study 307. Outcomes include safety, tolerability, change in kidney function, and percentage of subjects requiring KRT relative to historical data. Regular safety assessments monitor for possible adverse effects of treatment on parameters such as liver function, kidney function, fluid balance, electrolyte levels, and growth trajectory, with increased frequency of monitoring following tolvaptan initiation or dose escalation. CONCLUSIONS: These trials will provide data on tolvaptan safety and efficacy in a population without disease-specific treatment options. TRIAL REGISTRATION: Study 204: EudraCT 2020-005991-36; Study 307: EudraCT 2020-005992-10.


Asunto(s)
Quistes , Riñón Poliquístico Autosómico Dominante , Riñón Poliquístico Autosómico Recesivo , Humanos , Niño , Recién Nacido , Tolvaptán/uso terapéutico , Riñón Poliquístico Autosómico Recesivo/diagnóstico por imagen , Riñón Poliquístico Autosómico Recesivo/tratamiento farmacológico , Riñón , Estudios Longitudinales , Quistes/tratamiento farmacológico , Antagonistas de los Receptores de Hormonas Antidiuréticas/efectos adversos
3.
Physiol Rep ; 10(21): e15510, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36353932

RESUMEN

Autosomal recessive polycystic kidney disease (ARPKD) is an inherited pathology caused mainly by mutations of the polycystic kidney and hepatic disease 1 (PKHD1) gene, which usually leads to end-stage renal disease. Previous studies suggested that the P2X purinoreceptor 4 (P2X4 R) may play an important role in the progression of ARPKD. To test this hypothesis, we assessed the chronic effects of ivermectin (P2X4 R allosteric modulator) and 5-BDBD (P2X4 R antagonist) on the development of ARPKD in PCK/CrljCrl-Pkhd1pck/CRL (PCK) rats. Our data indicated that activation of ATP-mediated P2X4 R signaling with ivermectin for 6 weeks in high dose (50 mg/L; water supplementation) decreased the total body weight of PCK rats while the heart and kidney weight remained unaffected. Smaller doses of ivermectin (0.5 or 5 mg/L, 6 weeks) or the inhibition of P2X4 R signaling with 5-BDBD (18 mg/kg/day, food supplement for 8 weeks) showed no effect on electrolyte balance or the basic physiological parameters. Furthermore, cystic index analysis for kidneys and liver revealed no effect of smaller doses of ivermectin (0.5 or 5 mg/L) and 5-BDBD on the cyst development of PCK rats. We observed a slight increase in the cystic liver index on high ivermectin dose, possibly due to the cytotoxicity of the drug. In conclusion, this study revealed that pharmacological modulation of P2X4 R by ivermectin or 5-BDBD does not affect the development of ARPKD in PCK rats, which may provide insights for future studies on investigating the therapeutic potential of adenosine triphosphate (ATP)-P2 signaling in PKD diseases.


Asunto(s)
Riñón Poliquístico Autosómico Recesivo , Ratas , Animales , Riñón Poliquístico Autosómico Recesivo/tratamiento farmacológico , Riñón Poliquístico Autosómico Recesivo/genética , Riñón Poliquístico Autosómico Recesivo/patología , Ivermectina/farmacología , Ivermectina/uso terapéutico , Ratas Sprague-Dawley , Modelos Animales de Enfermedad , Adenosina Trifosfato
5.
Nat Commun ; 12(1): 6138, 2021 10 22.
Artículo en Inglés | MEDLINE | ID: mdl-34686668

RESUMEN

To investigate the pathogenesis of a congenital form of hepatic fibrosis, human hepatic organoids were engineered to express the most common causative mutation for Autosomal Recessive Polycystic Kidney Disease (ARPKD). Here we show that these hepatic organoids develop the key features of ARPKD liver pathology (abnormal bile ducts and fibrosis) in only 21 days. The ARPKD mutation increases collagen abundance and thick collagen fiber production in hepatic organoids, which mirrors ARPKD liver tissue pathology. Transcriptomic and other analyses indicate that the ARPKD mutation generates cholangiocytes with increased TGFß pathway activation, which are actively involved stimulating myofibroblasts to form collagen fibers. There is also an expansion of collagen-producing myofibroblasts with markedly increased PDGFRB protein expression and an activated STAT3 signaling pathway. Moreover, the transcriptome of ARPKD organoid myofibroblasts resemble those present in commonly occurring forms of liver fibrosis. PDGFRB pathway involvement was confirmed by the anti-fibrotic effect observed when ARPKD organoids were treated with PDGFRB inhibitors. Besides providing insight into the pathogenesis of congenital (and possibly acquired) forms of liver fibrosis, ARPKD organoids could also be used to test the anti-fibrotic efficacy of potential anti-fibrotic therapies.


Asunto(s)
Cirrosis Hepática/patología , Modelos Biológicos , Organoides/patología , Enfermedades de los Conductos Biliares/genética , Enfermedades de los Conductos Biliares/metabolismo , Enfermedades de los Conductos Biliares/patología , Colágeno/metabolismo , Células Epiteliales/patología , Humanos , Células Madre Pluripotentes Inducidas/citología , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Cirrosis Hepática/tratamiento farmacológico , Cirrosis Hepática/genética , Cirrosis Hepática/metabolismo , Mutación , Miofibroblastos/metabolismo , Miofibroblastos/patología , Organoides/efectos de los fármacos , Organoides/metabolismo , Riñón Poliquístico Autosómico Recesivo/tratamiento farmacológico , Riñón Poliquístico Autosómico Recesivo/genética , Riñón Poliquístico Autosómico Recesivo/metabolismo , Riñón Poliquístico Autosómico Recesivo/patología , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo
6.
Pediatr Nephrol ; 36(9): 2639-2658, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33474686

RESUMEN

Autosomal dominant polycystic kidney disease (ADPKD) and autosomal recessive polycystic kidney disease (ARPKD) are characterized by bilateral cystic kidney disease leading to progressive kidney function decline. These diseases also have distinct liver manifestations. The range of clinical presentation and severity of both ADPKD and ARPKD is much wider than was once recognized. Pediatric and adult nephrologists are likely to care for individuals with both diseases in their lifetimes. This article will review genetic, clinical, and imaging predictors of kidney and liver disease progression in ADPKD and ARPKD and will briefly summarize pharmacologic therapies to prevent progression.


Asunto(s)
Riñón Poliquístico Autosómico Dominante , Riñón Poliquístico Autosómico Recesivo , Adulto , Niño , Progresión de la Enfermedad , Humanos , Riñón Poliquístico Autosómico Dominante/diagnóstico por imagen , Riñón Poliquístico Autosómico Dominante/tratamiento farmacológico , Riñón Poliquístico Autosómico Dominante/genética , Riñón Poliquístico Autosómico Dominante/patología , Riñón Poliquístico Autosómico Recesivo/diagnóstico por imagen , Riñón Poliquístico Autosómico Recesivo/tratamiento farmacológico , Riñón Poliquístico Autosómico Recesivo/genética , Riñón Poliquístico Autosómico Recesivo/patología
7.
Semin Cell Dev Biol ; 110: 139-148, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32475690

RESUMEN

Polycystic kidney disease (PKD), comprising autosomal dominant polycystic kidney disease (ADPKD) and autosomal recessive polycystic kidney disease (ARPKD), is characterized by incessant cyst formation in the kidney and liver. ADPKD and ARPKD represent the leading genetic causes of renal disease in adults and children, respectively. ADPKD is caused by mutations in PKD1 encoding polycystin1 (PC1) and PKD2 encoding polycystin 2 (PC2). PC1/2 are multi-pass transmembrane proteins that form a complex localized in the primary cilium. Predominant ARPKD cases are caused by mutations in polycystic kidney and hepatic disease 1 (PKHD1) gene that encodes the Fibrocystin/Polyductin (FPC) protein, whereas a small subset of cases are caused by mutations in DAZ interacting zinc finger protein 1 like (DZIP1L) gene. FPC is a type I transmembrane protein, localizing to the cilium and basal body, in addition to other compartments, and DZIP1L encodes a transition zone/basal body protein. Apparently, PC1/2 and FPC are signaling molecules, while the mechanism that cilia employ to govern renal tubule morphology and prevent cyst formation is unclear. Nonetheless, recent genetic and biochemical studies offer a glimpse of putative physiological malfunctions and the pathomechanisms underlying both disease entities. In this review, I summarize the results of genetic studies that deduced the function of PC1/2 on cilia and of cilia themselves in cyst formation in ADPKD, and I discuss studies regarding regulation of polycystin biogenesis and cilia trafficking. I also summarize the synergistic genetic interactions between Pkd1 and Pkhd1, and the unique tissue patterning event controlled by FPC, but not PC1. Interestingly, while DZIP1L mutations generate compromised PC1/2 cilia expression, FPC deficiency does not affect PC1/2 biogenesis and ciliary localization, indicating that divergent mechanisms could lead to cyst formation in ARPKD. I conclude by outlining promising areas for future PKD research and highlight rationales for potential therapeutic interventions for PKD treatment.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Cilios/metabolismo , Riñón Poliquístico Autosómico Dominante/genética , Riñón Poliquístico Autosómico Recesivo/genética , Receptores de Superficie Celular/genética , Canales Catiónicos TRPP/genética , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Adulto , Cuerpos Basales/efectos de los fármacos , Cuerpos Basales/metabolismo , Cuerpos Basales/patología , Niño , Cilios/efectos de los fármacos , Cilios/patología , Medicamentos Herbarios Chinos/farmacología , Flavonoides/farmacología , Expresión Génica , Humanos , Riñón/efectos de los fármacos , Riñón/metabolismo , Riñón/patología , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Mutación , Riñón Poliquístico Autosómico Dominante/tratamiento farmacológico , Riñón Poliquístico Autosómico Dominante/metabolismo , Riñón Poliquístico Autosómico Dominante/patología , Riñón Poliquístico Autosómico Recesivo/tratamiento farmacológico , Riñón Poliquístico Autosómico Recesivo/metabolismo , Riñón Poliquístico Autosómico Recesivo/patología , Receptores de Superficie Celular/deficiencia , Transducción de Señal , Canales Catiónicos TRPP/deficiencia
8.
Int J Mol Sci ; 21(17)2020 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-32847032

RESUMEN

Autosomal recessive and autosomal dominant polycystic kidney disease (ARPKD, ADPKD) are systemic disorders with pronounced hepatorenal phenotypes. While the main underlying genetic causes of both ARPKD and ADPKD have been well-known for years, the exact molecular mechanisms resulting in the observed clinical phenotypes in the different organs, remain incompletely understood. Recent research has identified cellular metabolic changes in PKD. These findings are of major relevance as there may be an immediate translation into clinical trials and potentially clinical practice. Here, we review important results in the field regarding metabolic changes in PKD and their modulation as a potential target of systemic treatment.


Asunto(s)
Terapia Molecular Dirigida/tendencias , Enfermedades Renales Poliquísticas/tratamiento farmacológico , Enfermedades Renales Poliquísticas/metabolismo , Humanos , Metaboloma/fisiología , Terapia Molecular Dirigida/métodos , Enfermedades Renales Poliquísticas/genética , Riñón Poliquístico Autosómico Dominante/tratamiento farmacológico , Riñón Poliquístico Autosómico Dominante/genética , Riñón Poliquístico Autosómico Dominante/metabolismo , Riñón Poliquístico Autosómico Recesivo/tratamiento farmacológico , Riñón Poliquístico Autosómico Recesivo/genética , Riñón Poliquístico Autosómico Recesivo/metabolismo
9.
J. bras. nefrol ; 41(2): 300-303, Apr.-June 2019. graf
Artículo en Inglés | LILACS | ID: biblio-1012547

RESUMEN

ABSTRACT A 16-year-old female patient previously diagnosed with autosomal recessive polycystic kidney disease (ARPKD) presented with acute bilateral pneumonia, upper gastrointestinal bleeding caused by ruptured esophageal varices, ascites, and lower limb edema. She required intensive care and an endoscopic procedure to treat the gastrointestinal bleeding. The analysis of the differential diagnosis for chronic liver disease indicated she had a spontaneous splenorenal shunt. Ultrasound-guided biopsy revealed the patient had cirrhosis, as characteristically seen in individuals with ARPKD. She had no symptoms at discharge and was referred for review for a combined transplant.


RESUMO Relato de caso de uma paciente adolescente de 16 anos de idade com diagnóstico prévio de doença renal policística autossômica recessiva (DRPAR), que apresentou quadro agudo de pneumonia bilateral e hemorragia digestiva alta por ruptura de varizes esofágicas, bem como ascite e edema de membros inferiores. Necessitou de estabilização clínica intensiva e tratamento endoscópico do sangramento digestivo. Após investigação dos diagnósticos diferenciais da hepatopatia crônica, diagnosticou-se shunt esplenorrenal espontâneo, e realizou-se biópsia hepática guiada por ecografia com diagnóstico de cirrose, espectro típico da DRPAR. Recebeu alta hospitalar assintomática e foi encaminhada para avaliação de transplante duplo.


Asunto(s)
Humanos , Femenino , Adolescente , Anastomosis Arteriovenosa/patología , Riñón Poliquístico Autosómico Recesivo/complicaciones , Enfermedad de Caroli/complicaciones , Cirrosis Hepática/complicaciones , Anastomosis Arteriovenosa/diagnóstico por imagen , Derivación y Consulta , Venas Renales/diagnóstico por imagen , Biopsia , Brasil , Inhibidores de la Enzima Convertidora de Angiotensina/uso terapéutico , Unidades de Cuidado Intensivo Pediátrico , Resultado del Tratamiento , Riñón Poliquístico Autosómico Recesivo/tratamiento farmacológico , Riñón Poliquístico Autosómico Recesivo/diagnóstico por imagen , Enfermedad de Caroli/patología , Enfermedad de Caroli/tratamiento farmacológico , Angiografía por Resonancia Magnética , Agonistas Adrenérgicos beta/uso terapéutico , Diuréticos Conservadores de Potasio/uso terapéutico , Cirrosis Hepática/patología , Cirrosis Hepática/tratamiento farmacológico
10.
J Bras Nefrol ; 41(2): 300-303, 2019.
Artículo en Inglés, Portugués | MEDLINE | ID: mdl-30199558

RESUMEN

A 16-year-old female patient previously diagnosed with autosomal recessive polycystic kidney disease (ARPKD) presented with acute bilateral pneumonia, upper gastrointestinal bleeding caused by ruptured esophageal varices, ascites, and lower limb edema. She required intensive care and an endoscopic procedure to treat the gastrointestinal bleeding. The analysis of the differential diagnosis for chronic liver disease indicated she had a spontaneous splenorenal shunt. Ultrasound-guided biopsy revealed the patient had cirrhosis, as characteristically seen in individuals with ARPKD. She had no symptoms at discharge and was referred for review for a combined transplant.


Asunto(s)
Anastomosis Arteriovenosa/patología , Enfermedad de Caroli/complicaciones , Cirrosis Hepática/complicaciones , Riñón Poliquístico Autosómico Recesivo/complicaciones , Adolescente , Antagonistas Adrenérgicos beta/uso terapéutico , Inhibidores de la Enzima Convertidora de Angiotensina/uso terapéutico , Anastomosis Arteriovenosa/diagnóstico por imagen , Biopsia , Brasil , Enfermedad de Caroli/tratamiento farmacológico , Enfermedad de Caroli/patología , Diuréticos Conservadores de Potasio/uso terapéutico , Femenino , Humanos , Unidades de Cuidado Intensivo Pediátrico , Cirrosis Hepática/tratamiento farmacológico , Cirrosis Hepática/patología , Angiografía por Resonancia Magnética , Riñón Poliquístico Autosómico Recesivo/diagnóstico por imagen , Riñón Poliquístico Autosómico Recesivo/tratamiento farmacológico , Derivación y Consulta , Venas Renales/diagnóstico por imagen , Venas Renales/patología , Resultado del Tratamiento , Vena Cava Inferior/diagnóstico por imagen , Vena Cava Inferior/patología
11.
Saudi J Kidney Dis Transpl ; 29(6): 1475-1479, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30588981

RESUMEN

The mutations associated with polycystic kidney disease are closely aligned with that of tuberous sclerosis (TSC) in chromosome 16. Occasionally, the presence of these mutations in an individual can lead to a presence of a disease phenotype with a combination of polycystic kidney disease and TSC (contiguous gene syndrome). We present a case report of a young girl who presented with skin lesions, central nervous system tubers, and cystic disease of the kidneys. She was treated with mammalian target of rapamycin inhibitors with a favorable outcome.


Asunto(s)
Deleción Cromosómica , Riñón Poliquístico Autosómico Recesivo/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/uso terapéutico , Sirolimus/uso terapéutico , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Proteína 2 del Complejo de la Esclerosis Tuberosa/genética , Esclerosis Tuberosa/tratamiento farmacológico , Adolescente , Femenino , Predisposición Genética a la Enfermedad , Humanos , Imagen por Resonancia Magnética , Fenotipo , Riñón Poliquístico Autosómico Recesivo/diagnóstico , Riñón Poliquístico Autosómico Recesivo/enzimología , Riñón Poliquístico Autosómico Recesivo/genética , Serina-Treonina Quinasas TOR/metabolismo , Tomografía Computarizada por Rayos X , Resultado del Tratamiento , Esclerosis Tuberosa/diagnóstico , Esclerosis Tuberosa/enzimología , Esclerosis Tuberosa/genética
12.
PLoS One ; 12(5): e0177934, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28542433

RESUMEN

Increased intracellular cyclic AMP (cAMP) in renal tubular epithelia accelerates the progression of polycystic kidney disease (PKD). Thus, decreasing cAMP levels by an adenylyl cyclase inhibitory G protein activator is considered to be an effective approach in ameliorating PKD. In fact, pasireotide (PAS) was effective in reducing disease progression in animal models of PKD. However, hyperglycemia caused by the administration of PAS is an adverse effect in its clinical use. Whereas, co-administration of octreotide (OCT) with PAS did not increase serum glucose in normal rats. In the current study, we examined the efficacy of combined treatment with OCT and PAS in PCK rats, an autosomal recessive PKD model. Four-week-old PCK males were treated with the long-acting release type of OCT, PAS, or a combination of both (OCT/PAS) for 12 weeks. After termination, serum and renal tissue were used for analyses. Kidney weight, kidney weight per body weight, renal cyst area, renal Ki67 expression, and serum urea nitrogen were significantly decreased either in the PAS or OCT/PAS group, compared with vehicle. Renal tissue cAMP content was significantly decreased by PAS or OCT/PAS treatment, but not OCT, compared with vehicle. As a marker of cellular mTOR signaling activity, renal phospho-S6 kinase expression was significantly decreased by OCT/PAS treatment compared with vehicle, OCT, or PAS. Serum glucose was significantly increased by PAS administration, whereas no difference was shown between vehicle and OCT/PAS, possibly because serum glucagon was decreased either by the treatment of OCT alone or co-application of OCT/PAS. In conclusion, since serum glucose levels are increased by the use of PAS, its combination with OCT may reduce the risk of hyperglycemia associated with PAS monotherapy against PKD progression.


Asunto(s)
Octreótido/farmacología , Riñón Poliquístico Autosómico Recesivo/tratamiento farmacológico , Somatostatina/análogos & derivados , Animales , Glucemia/metabolismo , Peso Corporal/efectos de los fármacos , AMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Interacciones Farmacológicas , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Glucagón/sangre , Corazón/efectos de los fármacos , Corazón/fisiología , Humanos , Hidrocortisona/sangre , Hiperglucemia/inducido químicamente , Insulina/sangre , Factor I del Crecimiento Similar a la Insulina/metabolismo , Antígeno Ki-67/metabolismo , Riñón/efectos de los fármacos , Riñón/metabolismo , Riñón/patología , Riñón/fisiopatología , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Masculino , Octreótido/efectos adversos , Octreótido/uso terapéutico , Fosfoproteínas/metabolismo , Riñón Poliquístico Autosómico Recesivo/metabolismo , Riñón Poliquístico Autosómico Recesivo/patología , Riñón Poliquístico Autosómico Recesivo/fisiopatología , Ratas , Receptor IGF Tipo 1/sangre , Proteínas Quinasas S6 Ribosómicas/metabolismo , Somatostatina/efectos adversos , Somatostatina/farmacología , Somatostatina/uso terapéutico
13.
J Pediatr Gastroenterol Nutr ; 60(4): 467-73, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25822773

RESUMEN

OBJECTIVE: In polycystic liver disease (PCLD), multiple cysts cause liver enlargement, structural damage, and loss of function. Soy protein and dietary ω-3 polyunsaturated fatty acids (n-3 PUFAs) have been found to decrease cyst proliferation and inflammation in polycystic kidney disease. Therefore, the aim of the study was to investigate whether soy protein and n-3 PUFA supplementation attenuates PCLD. METHODS: Young (age 28 days) female PCK rats were fed (n = 12 per group) either casein + corn oil (casein + CO), casein + soybean oil (casein + SO), soy protein isolate + soybean oil (SPI + SO), or SPI + 1:1 soybean/salmon oil blend (SPI + SB) diet for 12 weeks. Liver histology, gene expression by real-time quantitative polymerase chain reaction, and serum markers of liver injury were determined. RESULTS: Diet had no effect on PCLD progression as indicated by no significant differences in liver weight and hepatic proliferation gene expression between diet groups. PCK rats fed SPI + SB diet, however, had the greatest (P < 0.05) histological evidence of hepatic cyst obstruction, portal inflammation, steatosis, and upregulation (P = 0.03) of fibrosis-related genes. Rats fed SPI + SB diet also had the lowest (P < 0.001) serum cholesterol and higher (P < 0.05) serum alkaline phosphatase and bilirubin concentrations. CONCLUSIONS: Feeding young female PCK rats SPI and n-3 PUFA failed to attenuate PCLD progression. Furthermore, feeding SPI + SB diet resulted in complications of hepatic steatosis attributable to cysts obstruction of bile duct and hepatic vein. Based on the results, it was concluded that diet intervention alone was not effective at attenuating PCLD associated with autosomal recessive polycystic kidney disease.


Asunto(s)
Quistes , Suplementos Dietéticos , Ácidos Grasos Omega-3/farmacología , Hepatopatías , Hígado/efectos de los fármacos , Riñón Poliquístico Autosómico Recesivo/patología , Proteínas de Soja/farmacología , Fosfatasa Alcalina/sangre , Animales , Conductos Biliares/efectos de los fármacos , Conductos Biliares/patología , Bilirrubina/sangre , Colesterol/sangre , Quistes/tratamiento farmacológico , Quistes/etiología , Dieta , Progresión de la Enfermedad , Ácidos Grasos Omega-3/efectos adversos , Ácidos Grasos Omega-3/uso terapéutico , Hígado Graso/sangre , Hígado Graso/etiología , Femenino , Venas Hepáticas/efectos de los fármacos , Venas Hepáticas/patología , Inflamación/tratamiento farmacológico , Inflamación/etiología , Cirrosis Hepática/sangre , Cirrosis Hepática/etiología , Cirrosis Hepática/genética , Hepatopatías/tratamiento farmacológico , Hepatopatías/etiología , Riñón Poliquístico Autosómico Recesivo/tratamiento farmacológico , Ratas , Proteínas de Soja/efectos adversos , Proteínas de Soja/uso terapéutico
14.
PLoS One ; 8(12): e81480, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24324698

RESUMEN

Human autosomal recessive polycystic kidney disease (ARPKD) produces kidneys which are massively enlarged due to multiple cysts, hypertension, and congenital hepatic fibrosis characterized by dilated bile ducts and portal hypertension. The PCK rat is an orthologous model of human ARPKD with numerous fluid-filled cysts caused by stimulated cellular proliferation in the renal tubules and hepatic bile duct epithelia, with interstitial fibrosis developed in the liver. We previously reported that a peroxisome proliferator activated receptor (PPAR)-γ full agonist ameliorated kidney and liver disease in PCK rats. Telmisartan is an angiotensin receptor blocker (ARB) used widely as an antihypertensive drug and shows partial PPAR-γ agonist activity. It also has nephroprotective activity in diabetes and renal injury and prevents the effects of drug-induced hepatotoxicity and hepatic fibrosis. In the present study, we determined whether telmisartan ameliorates progression of polycystic kidney and fibrocystic liver disease in PCK rats. Five male and 5 female PCK and normal control (+/+) rats were orally administered 3 mg/kg telmisartan or vehicle every day from 4 to 20 weeks of age. Treatment with telmisartan decreased blood pressure in both PCK and +/+ rats. Blood levels of aspartate amino transferase, alanine amino transferase and urea nitrogen were unaffected by telmisartan treatment. There was no effect on kidney disease progression, but liver weight relative to body weight, liver cystic area, hepatic fibrosis index, expression levels of Ki67 and TGF-ß, and the number of Ki67- and TGF-ß-positive interstitial cells in the liver were significantly decreased in telmisartan-treated PCK rats. Therefore, telmisartan ameliorates congenital hepatic fibrosis in ARPKD, possibly through the inhibition of signaling cascades responsible for cellular proliferation and interstitial fibrosis in PCK rats. The present results support the potential therapeutic use of ARBs for the treatment of fibrocystic liver disease in ARPKD patients.


Asunto(s)
Bencimidazoles/uso terapéutico , Benzoatos/uso terapéutico , Quistes/tratamiento farmacológico , Hepatopatías/tratamiento farmacológico , Riñón Poliquístico Autosómico Recesivo/tratamiento farmacológico , Angiotensina II/metabolismo , Animales , Bencimidazoles/farmacología , Benzoatos/farmacología , Presión Sanguínea/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Quistes/patología , Quistes/fisiopatología , Modelos Animales de Enfermedad , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Hepatocitos/efectos de los fármacos , Hepatocitos/patología , Humanos , Pruebas de Función Renal , Hígado/efectos de los fármacos , Hígado/enzimología , Hígado/patología , Hepatopatías/patología , Hepatopatías/fisiopatología , Pruebas de Función Hepática , Masculino , Tamaño de los Órganos/efectos de los fármacos , Riñón Poliquístico Autosómico Recesivo/patología , Riñón Poliquístico Autosómico Recesivo/fisiopatología , Ratas , Ratas Sprague-Dawley , Receptor de Angiotensina Tipo 1/metabolismo , Transducción de Señal/efectos de los fármacos , Telmisartán , Factor de Crecimiento Transformador beta/metabolismo
15.
Gastroenterology ; 142(3): 622-633.e4, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22155366

RESUMEN

BACKGROUND & AIMS: In polycystic kidney disease and polycystic liver disease (PLD), the normally nonproliferative hepato-renal epithelia acquire a proliferative, cystic phenotype that is linked to overexpression of cell division cycle 25 (Cdc25)A phosphatase and cell-cycle deregulation. We investigated the effects of Cdc25A inhibition in mice and rats via genetic and pharmacologic approaches. METHODS: Cdc25A(+/-) mice (which have reduced levels of Cdc25A) were cross-bred with polycystic kidney and hepatic disease 1 (Pkhd1(del2/del2)) mice (which have increased levels of Cdc25A and develop hepatic cysts). Cdc25A expression was analyzed in livers of control and polycystic kidney (PCK) rats, control and polycystic kidney 2 (Pkd2(ws25/-)) mice, healthy individuals, and patients with PLD. We examined effects of pharmacologic inhibition of Cdc25A with vitamin K3 (VK3) on the cell cycle, proliferation, and cyst expansion in vitro; hepato-renal cystogenesis in PCK rats and Pkd2(ws25/-)mice; and expression of Cdc25A and the cell-cycle proteins regulated by Cdc25A. We also examined the effects of the Cdc25A inhibitor PM-20 on hepato-renal cystogenesis in Pkd2(ws25/-) mice. RESULTS: Liver weights and hepatic and fibrotic areas were decreased by 32%-52% in Cdc25A(+/-):Pkhd1(del2/del2) mice, compared with Pkhd1(del2/del2) mice. VK3 altered the cell cycle and reduced proliferation of cultured cholangiocytes by 32%-83% and decreased growth of cultured cysts by 23%-67%. In PCK rats and Pkd2(ws25/-) mice, VK3 reduced liver and kidney weights and hepato-renal cystic and fibrotic areas by 18%-34%. PM-20 decreased hepato-renal cystogenesis in Pkd2(ws25/-) mice by 15%. CONCLUSIONS: Cdc25A inhibitors block cell-cycle progression and proliferation, reduce liver and kidney weights and cyst growth in animal models of polycystic kidney disease and PLD, and might be developed as therapeutics for these diseases.


Asunto(s)
Quistes/tratamiento farmacológico , Inhibidores Enzimáticos/farmacología , Riñón/efectos de los fármacos , Hepatopatías/tratamiento farmacológico , Hígado/efectos de los fármacos , Riñón Poliquístico Autosómico Recesivo/tratamiento farmacológico , Vitamina K 3/farmacología , Fosfatasas cdc25/antagonistas & inhibidores , Animales , Conductos Biliares Intrahepáticos/efectos de los fármacos , Conductos Biliares Intrahepáticos/enzimología , Conductos Biliares Intrahepáticos/patología , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Quistes/enzimología , Quistes/genética , Quistes/patología , Modelos Animales de Enfermedad , Humanos , Riñón/enzimología , Riñón/patología , Hígado/enzimología , Hígado/patología , Hepatopatías/enzimología , Hepatopatías/genética , Hepatopatías/patología , Ratones , Ratones Noqueados , Tamaño de los Órganos/efectos de los fármacos , Riñón Poliquístico Autosómico Recesivo/enzimología , Riñón Poliquístico Autosómico Recesivo/genética , Riñón Poliquístico Autosómico Recesivo/patología , Ratas , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Canales Catiónicos TRPP/genética , Canales Catiónicos TRPP/metabolismo , Factores de Tiempo , Regulación hacia Arriba , Fosfatasas cdc25/deficiencia , Fosfatasas cdc25/genética , Fosfatasas cdc25/metabolismo
16.
Kidney Int ; 80(6): 612-9, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21633408

RESUMEN

The development and progression of renal cysts appears to be driven by reduced cellular calcium and increased cyclic adenosine monophosphate (cAMP) from G-protein-coupled receptors. To test whether treatment with a calcimimetic that stimulates the G-protein-coupled calcium-sensing receptor might normalize cystic epithelial cell intracellular calcium and cAMP, thereby inhibiting cyst progression, we used pcy mice. These animals develop cysts principally in the collecting duct, as do humans with nephronophthisis (NPHP). We administered the calcimimetic R-568 mixed in their food at early or late stages in the pathogenesis of cyst formation. The treatment reduced cyst enlargement, and the early treatment inhibited development of renal fibrosis. Although the effect of later treatment was more modest, both stages of the disease responded positively to treatment. Additionally, R-568 decreased total kidney cAMP in the pcy mice and, in vitro, decreased cAMP levels and cell proliferation, while increasing intracellular calcium in immortalized human autosomal recessive polycystic kidney disease renal epithelial cells. The latter two effects were unique to R-568 and not replicated by raising extracellular calcium. Thus, treating pcy mice with R-568 was effective in reducing cyst progression in this rodent model of NPHP. Direct studies will be needed to determine whether these results can be applied to the human disease.


Asunto(s)
Compuestos de Anilina/farmacología , Calcimiméticos/farmacología , Enfermedades Renales Quísticas/patología , Enfermedades Renales Quísticas/prevención & control , Animales , Línea Celular , Proliferación Celular/efectos de los fármacos , AMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Riñón/efectos de los fármacos , Riñón/metabolismo , Riñón/patología , Enfermedades Renales Quísticas/genética , Enfermedades Renales Quísticas/metabolismo , Cinesinas/genética , Masculino , Ratones , Ratones Mutantes , Mutación Missense , Fenetilaminas , Riñón Poliquístico Autosómico Recesivo/tratamiento farmacológico , Riñón Poliquístico Autosómico Recesivo/metabolismo , Riñón Poliquístico Autosómico Recesivo/patología , Propilaminas , Receptores Sensibles al Calcio/agonistas
17.
Am J Physiol Renal Physiol ; 300(2): F465-74, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21147840

RESUMEN

In autosomal recessive polycystic kidney disease (ARPKD), progressive enlargement of fluid-filled cysts is due to aberrant proliferation of tubule epithelial cells and transepithelial fluid secretion leading to extensive nephron loss and interstitial fibrosis. Congenital hepatic fibrosis associated with biliary cysts/dilatations is the most common extrarenal manifestation in ARPKD and can lead to massive liver enlargement. Peroxisome proliferator-activated receptor γ (PPAR-γ), a member of the ligand-dependent nuclear receptor superfamily, is expressed in a variety of tissues, including the kidneys and liver, and plays important roles in cell proliferation, fibrosis, and inflammation. In the current study, we determined that pioglitazone (PIO), a PPAR-γ agonist, decreases polycystic kidney and liver disease progression in the polycystic kidney rat, an orthologous model of human ARPKD. Daily treatment with 10 mg/kg PIO for 16 wk decreased kidney weight (% of body weight), renal cystic area, serum urea nitrogen, and the number of Ki67-, pERK1/2-, and pS6-positive cells in the kidney. There was also a decrease in liver weight (% of body weight), liver cystic area, fibrotic index, and the number of Ki67-, pERK1/2-, pERK5-, and TGF-ß-positive cells in the liver. Taken together, these data suggest that PIO inhibits the progression of polycystic kidney and liver disease in a model of human ARPKD by inhibiting cell proliferation and fibrosis. These findings suggest that PPAR-γ agonists may have therapeutic value in the treatment of the renal and hepatic manifestations of ARPKD.


Asunto(s)
Hepatopatías/tratamiento farmacológico , PPAR gamma/agonistas , Riñón Poliquístico Autosómico Recesivo/tratamiento farmacológico , Tiazolidinedionas/uso terapéutico , Animales , Nitrógeno de la Urea Sanguínea , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Antígeno Ki-67/análisis , Cirrosis Hepática/tratamiento farmacológico , Masculino , Quinasas de Proteína Quinasa Activadas por Mitógenos/análisis , Pioglitazona , Ratas , Ratas Sprague-Dawley , Factor de Crecimiento Transformador beta/análisis
18.
Pediatr Nephrol ; 25(6): 1139-46, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20229187

RESUMEN

Angiotensin converting enzyme (ACE) inhibition is a common therapeutic modality in the treatment of autosomal recessive polycystic kidney disease (ARPKD). This study was designed to investigate whether chronic inhibition of ACE would have a therapeutic effect in attenuating the progression of renal cystogenesis in an orthologous rat model of ARPKD, the polycystic kidney (PCK) rat. Lisinopril (3 mg/kg per day) was administered orally for a period of 12 weeks, beginning at post-natal week 4. Lisinopril treatment resulted in an approximately 30% improvement in the collecting duct cystic indices (CT CI) of PCK animals. Activation of extracellular signal-regulated kinase 1 (ERK1) and 2 (ERK2), proliferative signaling markers, and proliferating cell nuclear antigen (PCNA), an end-point marker for proliferation, was reduced following chronic treatment with lisinopril compared to that in vehicle-treated PCK rats. To assess whether apoptotic pathways were altered due to chronic ACE inhibition, we examined p38 mitogen activated protein kinase (MAPK) and stress-activated protein kinase/c-Jun N-terminal kinase (SAPK/JNK), which are markers of apoptotic signaling cascades. p38 MAPK was significantly reduced (P < 0.0001) following chronic treatment with lisinopril, but no change in the activation of SAPK/JNK could be detected by immunoblot analysis. Lisinopril treatment resulted in a significant reduction (P < 0.01) in cleaved caspase-7 levels, but not caspase-3 activity, in PCK rat kidneys compared to the vehicle-treated PCK rat kidneys. Proteinuria was completely ameliorated in the presence of chronic ACE inhibition in the lisinopril-treated rats compared with the vehicle-treated PCK rats. In all, these findings demonstrated that chronic ACE inhibition can beneficially alter proliferative and apoptotic pathways to promote therapeutic reductions in renal cyst development in ARPKD.


Asunto(s)
Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Lisinopril/farmacología , Riñón Poliquístico Autosómico Recesivo/tratamiento farmacológico , Animales , Western Blotting , Modelos Animales de Enfermedad , Masculino , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos
19.
Nephrol Dial Transplant ; 24(2): 526-34, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18826972

RESUMEN

BACKGROUND: Antagonists of relevant Gs protein-coupled and agonists of relevant Gi protein-coupled receptors lower renal cAMP and inhibit growth of renal cysts in animal models of human ARPKD (PCK rat) and/or ADPKD (Pkd2(-/WS25) mouse). A calcium-sensing receptor (CaR) is expressed in various tubular segments and couples to Gq, thereby activating phospholipase Cgamma, InsP3 generation and calcium mobilization from intracellular stores, and Gi proteins. By both mechanisms, CaR activation could lower intracellular cAMP and inhibit renal cyst growth. METHODS: PCK rat and Pkd2(-/WS25) mouse littermates were fed rodent chow without or with R-568, a type 2 calcimimetic, at a concentration of 0.05% or 0.1% between 3 and 10 or 16 weeks of age. Histomorphometric analysis was performed with Meta-Morph software. Western analysis and immunohistochemical staining were performed using antibodies for aquaporin-2, urea transporter UT-A1 and CaR. Northern blot hybridization was used to quantify the expression of vasopressin V2 receptor and aquaporin 2 mRNAs. Cyclic AMP was measured using an enzyme immunoassay kit. RESULTS: R-568 had no effect on kidney weight, cyst volume, plasma BUN concentration or severity of the polycystic liver disease. A significant reduction in renal interstitial fibrosis was detected in PCK rats, but not in Pkd2(-/WS25) mice. R-568 administration, as anticipated, resulted in hypocalcemia and hyperphosphatemia, and significant increases in urine output, osmolar clearance, and urinary excretions of sodium, potassium and calcium. CONCLUSIONS: CaR activation had no detectable effect on cystogenesis in models of autosomal recessive or dominant polycystic kidney disease. The lack of protective effect could be due to the absence of CaR in the outer medullary and cortical collecting ducts, the reduction in extracellular calcium and the unaffected levels of renal cAMP and renal expression of cAMP-dependent genes. A possible beneficial effect on interstitial fibrosis deserves further study at more advanced stages of the disease.


Asunto(s)
Riñón Poliquístico Autosómico Dominante/genética , Riñón Poliquístico Autosómico Dominante/metabolismo , Riñón Poliquístico Autosómico Recesivo/genética , Riñón Poliquístico Autosómico Recesivo/metabolismo , Receptores Sensibles al Calcio/metabolismo , Compuestos de Anilina/farmacología , Animales , Acuaporina 2/genética , Calcio/agonistas , AMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Riñón/efectos de los fármacos , Riñón/metabolismo , Riñón/patología , Masculino , Ratones , Ratones Noqueados , Ratones Mutantes , Fenetilaminas , Riñón Poliquístico Autosómico Dominante/tratamiento farmacológico , Riñón Poliquístico Autosómico Dominante/patología , Riñón Poliquístico Autosómico Recesivo/tratamiento farmacológico , Riñón Poliquístico Autosómico Recesivo/patología , Propilaminas , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/genética , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Mutantes , Receptores Sensibles al Calcio/agonistas , Receptores de Vasopresinas/genética
20.
J Obstet Gynaecol Res ; 34(4 Pt 2): 599-602, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18840161

RESUMEN

Caroli's disease and Caroli's syndrome are rare congenital disorders characterized by non-obstructive cystic dilatation of the intrahepatic bile ducts. These disorders are often associated with autosomal recessive polycystic kidney disease. A young woman at 11 weeks of gestation was referred to our hospital for proper management of Caroli's disease during pregnancy. Magnetic resonance imaging and laboratory tests revealed Caroli's disease with chronic renal failure caused by polycystic kidney disease. She received diet control, erythropoietin and prophylactic oral antibiotics. Her pregnancy course was uneventful, and she gave birth at 37 weeks of gestation. Thereafter, her renal function gradually worsened. Hemodialysis was begun 5 years after parturition. Though the courses of pregnancies complicated by Caroli's disease or Caroli's syndrome are variable and can include life-threatening conditions, uneventful outcomes can be expected if careful management prevents biliary and renal infection.


Asunto(s)
Enfermedad de Caroli/complicaciones , Riñón Poliquístico Autosómico Recesivo/complicaciones , Complicaciones del Embarazo/etiología , Enfermedad de Caroli/dietoterapia , Enfermedad de Caroli/tratamiento farmacológico , Femenino , Humanos , Riñón Poliquístico Autosómico Recesivo/dietoterapia , Riñón Poliquístico Autosómico Recesivo/tratamiento farmacológico , Embarazo , Complicaciones del Embarazo/dietoterapia , Complicaciones del Embarazo/tratamiento farmacológico , Resultado del Embarazo , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...