Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 383
Filtrar
1.
Sci Rep ; 11(1): 14286, 2021 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-34253766

RESUMEN

We examined the effect of total and afferent renal denervation (RDN) on hypertension and the renin-angiotensin system (RAS) in a rodent model of juvenile-onset polycystic kidney disease (PKD). Lewis Polycystic Kidney (LPK) and control rats received total, afferent or sham RDN by periaxonal application of phenol, capsaicin or normal saline, respectively, and were monitored for 4-weeks. Afferent RDN did not affect systolic blood pressure (SBP) determined by radiotelemetry in either strain (n = 19) while total RDN significantly reduced SBP in Lewis rats 4-weeks post-denervation (total vs. sham, 122 ± 1 vs. 130 ± 2 mmHg, P = 0.002, n = 25). Plasma and kidney renin content determined by radioimmunoassay were significantly lower in LPK vs. Lewis (plasma: 278.2 ± 6.7 vs. 376.5 ± 11.9 ng Ang I/ml/h; kidney: 260.1 ± 6.3 vs. 753.2 ± 37.9 ng Ang I/mg/h, P < 0.001, n = 26). These parameters were not affected by RDN. Intrarenal mRNA expression levels of renin, angiotensinogen, angiotensin-converting enzyme (ACE)2, and angiotensin II receptor type 1a were significantly lower, whereas ACE1 expression was significantly higher in the LPK vs. Lewis (all P < 0.05, n = 26). This pattern of intrarenal RAS expression was not changed by RDN. In conclusion, RDN does not affect hypertension or the RAS in the LPK model and indicates RDN might not be a suitable antihypertensive strategy for individuals with juvenile-onset PKD.


Asunto(s)
Desnervación , Hipertensión/complicaciones , Riñón/inervación , Riñón/fisiopatología , Enfermedades Renales Poliquísticas/fisiopatología , Sistema Renina-Angiotensina , Animales , Antihipertensivos/farmacología , Presión Sanguínea/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Riñón/cirugía , Masculino , ARN Mensajero/metabolismo , Radioinmunoensayo , Ratas , Ratas Endogámicas Lew , Renina/metabolismo
2.
Clin Exp Nephrol ; 25(8): 902-910, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-33881641

RESUMEN

BACKGROUND: Disease-specific trajectories of renal function in advanced chronic kidney disease (CKD) are not well defined. Here, we compared these trajectories in the estimated glomerular filtration rate (eGFR) by CKD stages. METHODS: Patients with multiple eGFR measurements during the 5-year preregistration period of the REACH-J study were enrolled. Mean annual eGFR declines were calculated from linear mixed effect models with the adjustment variables of baseline CKD stage, age, sex and the current CKD stage and the level of proteinuria (CKDA1-3). RESULTS: Among 1,969 eligible patients with CKDG3b-5, the adjusted eGFR decline (ml/min/1.73 m2/year) was significantly faster in diabetic kidney disease (DKD) patients and polycystic kidney disease (PKD) patients than in patients with other kidney diseases (DKD, - 2.96 ± 0.13; PKD, - 2.82 ± 0.17; and others, - 1.95 ± 0.05, p < 0.01). The declines were faster with higher CKD stages. In DKD patients, the eGFR decline was significantly faster in CKDG5 than CKDG4 (- 4.10 ± 0.18 vs - 2.76 ± 0.20, p < 0.01), while these declines in PKD patients were similar. The eGFR declines in PKD patients were significantly faster than DKD patients in CKDG4 (- 2.92 ± 0.23 vs - 2.76 ± 0.20, p < 0.01) and in CKDA2 (- 3.36 ± 0.35 vs - 1.40 ± 0.26, p < 0.01). CONCLUSION: Our study revealed the disease-specific annual eGFR declines by CKD stages and the level of proteinuria. Comparing to the other kidney diseases, the declines in PKD patients were getting faster from early stages of CKD. These results suggest the importance of CKD managements in PKD patients from the early stages.


Asunto(s)
Tasa de Filtración Glomerular , Fallo Renal Crónico/fisiopatología , Anciano , Anciano de 80 o más Años , Nefropatías Diabéticas/complicaciones , Nefropatías Diabéticas/fisiopatología , Femenino , Humanos , Fallo Renal Crónico/etiología , Masculino , Persona de Mediana Edad , Enfermedades Renales Poliquísticas/complicaciones , Enfermedades Renales Poliquísticas/fisiopatología , Estudios Prospectivos
3.
JCI Insight ; 6(10)2021 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-33886508

RESUMEN

Flow-activated Na+ and HCO3- transport in kidney proximal tubules (PT) underlies relatively constant fractional reabsorption during changes in glomerular filtration rate (GFR) or glomerulotubular balance (GTB). In view of hypothesized connections of epithelial cilia to flow sensing, we examined flow-activated transport in 3 polycystic kidney disease-related mouse models based on inducible conditional KO of Pkd1, Pkd2, and Kif3a. PTs were harvested from mice after gene inactivation but prior to cyst formation, and flow-mediated PT transport was measured. We confirm that higher flow increased both Na+ and HCO3- absorption in control mice, and we observed that this flow effect was preserved in PTs of Pkd1-/- and Kif3a-/-mice. However, flow activation was absent in Pkd2+/- and Pkd2-/- PT. In heterozygous (Pkd2+/-) mice, a dopamine receptor 1 (DA1) antagonist (SCH23390) restored transport flow sensitivity. When given chronically, this same antagonist reduced renal cyst formation in Pkd2-/-, as evidenced by reduced kidney weight, BUN, and the cystic index, when compared with untreated mice. In contrast, SCH23390 did not prevent cyst formation in Pkd1-/- mice. These results indicate that Pkd2 is necessary for normal GTB and that restoration of flow-activated transport by DA1 antagonist can slow renal cyst formation in Pkd2-/- mice.


Asunto(s)
Túbulos Renales Proximales/fisiología , Enfermedades Renales Poliquísticas , Animales , Benzazepinas/farmacología , Modelos Animales de Enfermedad , Femenino , Riñón/metabolismo , Masculino , Ratones , Ratones Noqueados , Enfermedades Renales Poliquísticas/genética , Enfermedades Renales Poliquísticas/metabolismo , Enfermedades Renales Poliquísticas/patología , Enfermedades Renales Poliquísticas/fisiopatología , Receptores de Dopamina D1/antagonistas & inhibidores , Receptores de Dopamina D1/metabolismo , Canales Catiónicos TRPP/genética , Canales Catiónicos TRPP/metabolismo
4.
Am J Kidney Dis ; 77(3): 410-419, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33039432

RESUMEN

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


Asunto(s)
Síndrome de Bardet-Biedl/genética , Cilios/metabolismo , Ciliopatías/genética , Enfermedades Renales Poliquísticas/genética , Anomalías Múltiples/genética , Anomalías Múltiples/metabolismo , Anomalías Múltiples/fisiopatología , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras del Transporte Vesicular/genética , Síndrome de Bardet-Biedl/metabolismo , Síndrome de Bardet-Biedl/fisiopatología , Cerebelo/anomalías , Cerebelo/metabolismo , Cerebelo/fisiopatología , Chaperoninas/genética , Cilios/fisiología , Trastornos de la Motilidad Ciliar/genética , Trastornos de la Motilidad Ciliar/metabolismo , Trastornos de la Motilidad Ciliar/fisiopatología , Ciliopatías/metabolismo , Ciliopatías/fisiopatología , Proteínas del Citoesqueleto/genética , Encefalocele/genética , Encefalocele/metabolismo , Encefalocele/fisiopatología , Anomalías del Ojo/genética , Anomalías del Ojo/metabolismo , Anomalías del Ojo/fisiopatología , Humanos , Enfermedades Renales Quísticas/genética , Enfermedades Renales Quísticas/metabolismo , Enfermedades Renales Quísticas/fisiopatología , Amaurosis Congénita de Leber/genética , Amaurosis Congénita de Leber/metabolismo , Amaurosis Congénita de Leber/fisiopatología , Proteínas de la Membrana/genética , Proteínas Asociadas a Microtúbulos/genética , Atrofias Ópticas Hereditarias/genética , Atrofias Ópticas Hereditarias/metabolismo , Atrofias Ópticas Hereditarias/fisiopatología , Enfermedades Renales Poliquísticas/metabolismo , Enfermedades Renales Poliquísticas/fisiopatología , Proteínas/genética , Retina/anomalías , Retina/metabolismo , Retina/fisiopatología , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/metabolismo , Retinitis Pigmentosa/fisiopatología , Canales Catiónicos TRPP/genética
5.
Adv Drug Deliv Rev ; 161-162: 176-189, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32866560

RESUMEN

Polycystic kidney disease (PKD) is characterized by progressive cyst growth and is a leading cause of renal failure worldwide. Currently, there are limited therapeutic options available to PKD patients, and only one drug, tolvaptan, has been FDA-approved to slow cyst progression. Similar to other small molecule drugs, however, tolvaptan is costly, only moderately effective, and causes adverse events leading to high patient dropout rates. Peptides may mitigate many drawbacks of small molecule drugs, as they can be highly tissue-specific, biocompatible, and economically scaled-up. Peptides can function as targeting ligands that direct therapies to diseased renal tissue, or be potent as therapeutic agents themselves. This review discusses various aberrant signaling pathways in PKD and renal receptors that can be potential targets of peptide-mediated strategies. Additionally, peptides utilized in other kidney applications, but may prove useful in the context of PKD, are highlighted. Insights into novel peptide-based solutions that have potential to improve clinical management of PKD are provided.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Riñón/fisiología , Péptidos/administración & dosificación , Enfermedades Renales Poliquísticas/tratamiento farmacológico , Enfermedades Renales Poliquísticas/fisiopatología , Animales , Progresión de la Enfermedad , Humanos , Transducción de Señal
7.
Nat Rev Nephrol ; 16(9): 489-508, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32704047

RESUMEN

Autophagy is a conserved lysosomal pathway for the degradation of cytoplasmic components. Basal autophagy in kidney cells is essential for the maintenance of kidney homeostasis, structure and function. Under stress conditions, autophagy is altered as part of the adaptive response of kidney cells, in a process that is tightly regulated by signalling pathways that can modulate the cellular autophagic flux - mammalian target of rapamycin, AMP-activated protein kinase and sirtuins are key regulators of autophagy. Dysregulated autophagy contributes to the pathogenesis of acute kidney injury, to incomplete kidney repair after acute kidney injury and to chronic kidney disease of varied aetiologies, including diabetic kidney disease, focal segmental glomerulosclerosis and polycystic kidney disease. Autophagy also has a role in kidney ageing. However, questions remain about whether autophagy has a protective or a pathological role in kidney fibrosis, and about the precise mechanisms and signalling pathways underlying the autophagy response in different types of kidney cells and across the spectrum of kidney diseases. Further research is needed to gain insights into the regulation of autophagy in the kidneys and to enable the discovery of pathway-specific and kidney-selective therapies for kidney diseases and anti-ageing strategies.


Asunto(s)
Lesión Renal Aguda/fisiopatología , Envejecimiento/fisiología , Autofagia/fisiología , Insuficiencia Renal Crónica/fisiopatología , Proteínas Quinasas Activadas por AMP/metabolismo , Lesión Renal Aguda/metabolismo , Envejecimiento/metabolismo , Nefropatías Diabéticas/metabolismo , Nefropatías Diabéticas/fisiopatología , Fibrosis , Glomeruloesclerosis Focal y Segmentaria/metabolismo , Glomeruloesclerosis Focal y Segmentaria/fisiopatología , Homeostasis , Humanos , Glomérulos Renales , Túbulos Renales Proximales , Mitofagia/fisiología , Enfermedades Renales Poliquísticas/metabolismo , Enfermedades Renales Poliquísticas/fisiopatología , Insuficiencia Renal Crónica/metabolismo , Transducción de Señal , Sirtuinas/metabolismo , Serina-Treonina Quinasas TOR/metabolismo
8.
J Am Soc Nephrol ; 31(6): 1178-1190, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32295825

RESUMEN

The kidney contains a network of lymphatic vessels that clear fluid, small molecules, and cells from the renal interstitium. Through modulating immune responses and via crosstalk with surrounding renal cells, lymphatic vessels have been implicated in the progression and maintenance of kidney disease. In this Review, we provide an overview of the development, structure, and function of lymphatic vessels in the healthy adult kidney. We then highlight the contributions of lymphatic vessels to multiple forms of renal pathology, emphasizing CKD, transplant rejection, and polycystic kidney disease and discuss strategies to target renal lymphatics using genetic and pharmacologic approaches. Overall, we argue the case for lymphatics playing a fundamental role in renal physiology and pathology and treatments modulating these vessels having therapeutic potential across the spectrum of kidney disease.


Asunto(s)
Enfermedades Renales/etiología , Vasos Linfáticos/fisiología , Inmunidad Adaptativa , Rechazo de Injerto , Humanos , Enfermedades Renales/fisiopatología , Trasplante de Riñón/efectos adversos , Linfa/fisiología , Linfangiogénesis , Vasos Linfáticos/anatomía & histología , Vasos Linfáticos/citología , Enfermedades Renales Poliquísticas/fisiopatología , Factor A de Crecimiento Endotelial Vascular/fisiología
9.
Expert Opin Ther Targets ; 24(6): 589-599, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32250187

RESUMEN

Introduction: Polycystic liver disease (PLD) is a rare disease defined by the growth of hepatic cysts and occurs either isolated or as an extrarenal manifestation of polycystic kidney disease. While surgery has been the mainstay in treatment of symptomatic PLD, recently discovered regulatory mechanisms affecting hepatic cystogenesis provide potential new therapies to reduce hepatic cyst burden.Areas covered: This review summarizes intracellular pathways and therapeutic targets involved in hepatic cystogenesis. While drugs that target cAMP, mTOR and bile acids were evaluated in clinical trials, investigation in autophagy, Wnt and miRNA signaling pathways are still in the pre-clinical phase. Recent epidemiological data present female hormones as a promising therapeutic target. Additionally, therapeutic advances in renal cystogenesis are reviewed for their potential application in treatment of hepatic cysts.Expert opinion: Further elucidation of the pathophysiology of hepatic cystogenesis is needed to provide additional targets and improve the efficacy of current treatments. The most promising therapeutic target in PLD is the female hormone pathway, given the increased severity in women and the harmful effects of exogenous estrogens. In addition, combining current pharmaceutical and surgical therapies can lead to improved outcomes. Lastly, the rarity of PLD creates the need to share expertise internationally.


Asunto(s)
Quistes/tratamiento farmacológico , Hepatopatías/tratamiento farmacológico , Terapia Molecular Dirigida , Enfermedades Renales Poliquísticas/tratamiento farmacológico , Animales , Autofagia/efectos de los fármacos , Quistes/fisiopatología , Femenino , Humanos , Hepatopatías/fisiopatología , MicroARNs/metabolismo , Enfermedades Renales Poliquísticas/fisiopatología , Transducción de Señal/efectos de los fármacos , Vía de Señalización Wnt/efectos de los fármacos
10.
Am J Physiol Renal Physiol ; 318(3): F557-F564, 2020 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-31928223

RESUMEN

Klotho interacts with various membrane proteins such as receptors for transforming growth factor-ß (TGF-ß) and insulin-like growth factor (IGF). Renal expression of klotho is diminished in polycystic kidney disease (PKD). In the present study, the effects of klotho supplementation on PKD were assessed. Recombinant human klotho protein (10 µg·kg-1·day-1) or a vehicle was administered daily by subcutaneous injection to 6-wk-old mice with PKD (DBA/2-pcy). Blood pressure was measured using tail-cuff methods. After 2 mo, mice were killed, and the kidneys were harvested for analysis. Exogenous klotho protein supplementation reduced kidney weight, cystic area, systolic blood pressure, renal angiotensin II levels, and 8-epi-PGF2α excretion (P < 0.05). Klotho protein supplementation enhanced glomerular filtration rate, renal expression of superoxide dismutase, and klotho itself (P < 0.05). Klotho supplementation attenuated renal expressions of TGF-ß and collagen type I and diminished renal abundance of Twist, phosphorylated Akt, and mammalian target of rapamycin (P < 0.05). Pathological examination revealed that klotho decreased the fibrosis index and nuclear staining of Smad in PKD kidneys (P < 0.05). Our data indicate that klotho protein supplementation ameliorates the renin-angiotensin system, reducing blood pressure in PKD mice. Furthermore, the present results implicate klotho supplementation in the suppression of Akt/mammalian target of rapamycin signaling, slowing cystic expansion. Finally, our findings suggest that klotho protein supplementation attenuated fibrosis at least partly by inhibiting epithelial mesenchymal transition in PKD.


Asunto(s)
Presión Sanguínea/efectos de los fármacos , Glucuronidasa/uso terapéutico , Enfermedades Renales Poliquísticas/tratamiento farmacológico , Enfermedades Renales Poliquísticas/genética , Animales , Células Cultivadas , Femenino , Glucuronidasa/administración & dosificación , Inyecciones Subcutáneas , Riñón/fisiología , Proteínas Klotho , Ratones , Miofibroblastos/efectos de los fármacos , Enfermedades Renales Poliquísticas/fisiopatología , Proteínas Recombinantes
11.
J Mol Cell Biol ; 12(8): 644-653, 2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31065693

RESUMEN

Polycystic kidney disease (PKD) is a prevalent genetic disorder, characterized by the formation of kidney cysts that progressively lead to kidney failure. The currently available drug tolvaptan is not well tolerated by all patients and there remains a strong need for alternative treatments. The signaling rewiring in PKD that drives cyst formation is highly complex and not fully understood. As a consequence, the effects of drugs are sometimes difficult to predict. We previously established a high throughput microscopy phenotypic screening method for quantitative assessment of renal cyst growth. Here, we applied this 3D cyst growth phenotypic assay and screened 2320 small drug-like molecules, including approved drugs. We identified 81 active molecules that inhibit cyst growth. Multi-parametric phenotypic profiling of the effects on 3D cultured cysts discriminated molecules that showed preferred pharmacological effects above genuine toxicological properties. Celastrol, a triterpenoid from Tripterygium Wilfordii, was identified as a potent inhibitor of cyst growth in vitro. In an in vivo iKspCre-Pkd1lox,lox mouse model for PKD, celastrol inhibited the growth of renal cysts and maintained kidney function.


Asunto(s)
Evaluación Preclínica de Medicamentos , Triterpenos Pentacíclicos/uso terapéutico , Enfermedades Renales Poliquísticas/tratamiento farmacológico , Animales , Quistes/patología , Quistes/fisiopatología , Pruebas de Función Renal , Ratones , Triterpenos Pentacíclicos/farmacología , Fenotipo , Enfermedades Renales Poliquísticas/patología , Enfermedades Renales Poliquísticas/fisiopatología , Transducción de Señal , Bibliotecas de Moléculas Pequeñas/análisis , Bibliotecas de Moléculas Pequeñas/farmacología , Bibliotecas de Moléculas Pequeñas/uso terapéutico
12.
Hum Pathol ; 97: 1-7, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31857138

RESUMEN

Nonfunctioning kidneys secondary to various etiologies display different histopathological features. Studies focused on incidence and types of renal neoplasms using the new World Health Organization and International Society of Urological Pathology classification system in various types of nonfunctioning kidneys are very limited. We identified 311 nephrectomies of nonfunctioning kidneys and categorized them into 5 categories: acquired cystic kidney disease (ACKD, n = 61); end-stage renal disease, nonspecific (ESRD, n = 63); adult polycystic kidney disease (APKD, n = 49); failed transplant kidney (FTK, n = 96); and those caused by obstructive conditions in the kidney (OCK, n = 42). ACKD (70%) and ESRD (43%) had higher cancer incidences than the other 3 groups (APKD = 2%, FTK = 0%, and OCK = 5%). Besides clear cell renal cell carcinoma (RCC) and papillary RCC, clear cell papillary RCC had a much higher incidence within ACKD patients (13/61) compared to other groups. ACKD-associated RCC was only identified in ACKD patients. ACKD patients had significantly longer dialysis duration compared to ESRD, APKD, and FTK. Although they had similar risk for clear cell RCC and papillary RCC, ACKD patients had a much higher risk for ACKD-associated RCC and clear cell papillary RCC than ESRD patients. Although most RCCs arising in these nonfunctioning kidneys were early pT1 stage, 6 ACKD patients and 3 ESRD patients had higher-stage diseases, which can be fatal if not treated appropriately. Therefore, precise clinicopathological classification of these nonfunctioning kidneys is important for predicting kidney cancer risk. These results indicate the need for active monitoring of the patients with high-risk nonfunctioning kidney diseases and appropriate surgical treatment when necessary.


Asunto(s)
Carcinoma de Células Renales/patología , Enfermedades Renales Quísticas/patología , Fallo Renal Crónico/patología , Neoplasias Renales/patología , Enfermedades Renales Poliquísticas/patología , Adulto , Anciano , Anciano de 80 o más Años , Carcinoma de Células Renales/epidemiología , Carcinoma de Células Renales/fisiopatología , Carcinoma de Células Renales/cirugía , Femenino , Humanos , Incidencia , Enfermedades Renales Quísticas/epidemiología , Enfermedades Renales Quísticas/fisiopatología , Enfermedades Renales Quísticas/terapia , Fallo Renal Crónico/epidemiología , Fallo Renal Crónico/fisiopatología , Fallo Renal Crónico/terapia , Neoplasias Renales/epidemiología , Neoplasias Renales/fisiopatología , Neoplasias Renales/cirugía , Trasplante de Riñón , Masculino , Persona de Mediana Edad , Nefrectomía , Enfermedades Renales Poliquísticas/epidemiología , Enfermedades Renales Poliquísticas/fisiopatología , Enfermedades Renales Poliquísticas/terapia , Pronóstico , Diálisis Renal , Medición de Riesgo , Factores de Riesgo , Insuficiencia del Tratamiento , Adulto Joven
13.
Adv Exp Med Biol ; 1165: 81-100, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31399962

RESUMEN

Polycystic kidney disease (PKD) is a common genetic disorder characterized by formations of numerous cysts in kidneys and most caused by PKD1 or PKD2 mutations in autosomal dominant polycystic kidney disease (ADPKD). The interstitial inflammation and fibrosis is one of the major pathological changes in polycystic kidney tissues with an accumulation of inflammatory cells, chemokines, and cytokines. The immune response is observed across different stages and occurs prior to or coincident with cyst formation in ADPKD. Evidence for inflammation as an important contributor to cyst growth and fibrosis includes increased interstitial macrophages, upregulated expressions of pro-inflammatory cytokines, activated complement system, and activated pathways including NF-κB and JAK-STAT signaling in polycystic kidney tissues. Inflammatory cells are responsible for overproduction of several pro-fibrotic growth factors which promote renal fibrosis in ADPKD. These growth factors trigger epithelial mesenchymal transition and myofibroblast/fibrocyte activation, which stimulate the expansion of extracellular matrix (ECM) including collagen I, III, IV, V, and fibronectin, leading to renal fibrosis and reduced renal function. Besides, there are imbalanced ECM turnover regulators which lead to the increased ECM production and inadequate degradation in polycystic kidney tissues. Several fibrosis associated signaling pathways, such as TGFß-SMAD, Wnt, and periostin-integrin-linked kinase are also activated in polycystic kidney tissues. Although the effective anti-fibrotic treatments are limited at the present time, slowing the cyst expansion and fibrosis development is very important for prolonging life span and improving the palliative care of ADPKD patients. The inhibition of pro-fibrotic cytokines involved in fibrosis might be a new therapeutic strategy for ADPKD in the future.


Asunto(s)
Riñón/patología , Enfermedades Renales Poliquísticas/fisiopatología , Proteínas del Sistema Complemento , Citocinas , Matriz Extracelular , Fibrosis , Humanos , Péptidos y Proteínas de Señalización Intercelular , Macrófagos , Riñón Poliquístico Autosómico Dominante , Transducción de Señal
14.
Am J Physiol Renal Physiol ; 317(2): F343-F360, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31091126

RESUMEN

Polycystic kidney disease (PKD) is characterized by slowly expanding renal cysts that damage the kidney, typically resulting in renal failure by the fifth decade. The most common cause of death in these patients, however, is cardiovascular disease. Expanding cysts in PKD induce chronic kidney injury that is accompanied by immune cell infiltration, including macrophages, which we and others have shown can promote disease progression in PKD mouse models. Here, we show that monocyte chemoattractant protein-1 [MCP-1/chemokine (C-C motif) ligand 2 (CCL2)] is responsible for the majority of monocyte chemoattractant activity produced by renal PKD cells from both mice and humans. To test whether the absence of MCP-1 lowers renal macrophage concentration and slows disease progression, we generated genetic knockout (KO) of MCP-1 in a mouse model of PKD [congenital polycystic kidney (cpk) mice]. Cpk mice are born with rapidly expanding renal cysts, accompanied by a decline in kidney function and death by postnatal day 21. Here, we report that KO of MCP-1 in these mice increased survival, with some mice living past 3 mo. Surprisingly, however, there was no significant difference in renal macrophage concentration, nor was there improvement in cystic disease or kidney function. Examination of mice revealed cardiac hypertrophy in cpk mice, and measurement of cardiac electrical activity via ECG revealed repolarization abnormalities. MCP-1 KO did not affect the number of cardiac macrophages, nor did it alleviate the cardiac aberrancies. However, MCP-1 KO did prevent the development of pulmonary edema, which occurred in cpk mice, and promoted decreased resting heart rate and increased heart rate variability in both cpk and noncystic mice. These data suggest that in this mouse model of PKD, MCP-1 altered cardiac/pulmonary function and promoted death outside of its role as a macrophage chemoattractant.


Asunto(s)
Arritmias Cardíacas/metabolismo , Cardiomegalia/metabolismo , Quimiocina CCL2/metabolismo , Riñón/metabolismo , Pulmón/metabolismo , Miocardio/metabolismo , Enfermedades Renales Poliquísticas/metabolismo , Edema Pulmonar/metabolismo , Animales , Arritmias Cardíacas/patología , Arritmias Cardíacas/fisiopatología , Cardiomegalia/patología , Cardiomegalia/fisiopatología , Células Cultivadas , Quimiocina CCL2/deficiencia , Quimiocina CCL2/genética , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Fibrosis , Humanos , Mediadores de Inflamación/metabolismo , Riñón/patología , Riñón/fisiopatología , Pulmón/patología , Pulmón/fisiopatología , Macrófagos/metabolismo , Macrófagos/patología , Proteínas de la Membrana/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Miocardio/patología , Enfermedades Renales Poliquísticas/patología , Enfermedades Renales Poliquísticas/fisiopatología , Edema Pulmonar/patología , Edema Pulmonar/fisiopatología , Edema Pulmonar/prevención & control , Factores de Tiempo
15.
Adv Exp Med Biol ; 1132: 99-112, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31037629

RESUMEN

Periostin is a matricellular protein that is expressed in several tissues during embryonic development; however, its expression in adults is mostly restricted to collagen-rich connective tissues. Periostin is expressed only briefly during kidney development, but it is not normally detected in the adult kidney. Recent evidence has revealed that periostin is aberrantly expressed in several forms of chronic kidney disease (CKD), and that its expression correlates with the degree of interstitial fibrosis and the decline in renal function. Polycystic kidney disease (PKD), a genetic disorder, is characterized by the formation of numerous fluid-filled cysts in the kidneys. Periostin is secreted by the cyst epithelial cells and accumulates within the extracellular matrix adjacent to the cysts. In PKD mice, periostin overexpression accelerates cyst growth and contributes to structural changes in the kidneys, including interstitial fibrosis. Recent evidence suggests that periostin is a tissue repair molecule; however, its role in repair following acute kidney injury has not been investigated. It is thought that persistent expression of this protein in CKD contributes importantly to tubulointerstitial fibrosis and the progressive decline in renal function. Future studies to define the diverse actions of periostin during kidney injury may lead to effective therapies to slow PKD progression and possibly prevent the development of CKD. This chapter reviews the current literature on the expression of periostin in PKD and other forms of CKD, mechanisms for periostin stimulated cyst growth, its potential role in extracellular matrix production and renal fibrosis, and the evidence for periostin as a novel biomarker for kidney disease.


Asunto(s)
Moléculas de Adhesión Celular/fisiología , Riñón/fisiología , Insuficiencia Renal Crónica/fisiopatología , Animales , Matriz Extracelular , Fibrosis , Humanos , Riñón/patología , Ratones , Enfermedades Renales Poliquísticas/fisiopatología
16.
Am J Nephrol ; 49(3): 233-240, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30820006

RESUMEN

BACKGROUND: Genetic characteristics of polycystic kidney disease (PKD) patients without apparent family history were reported to be different from those with a positive family history. However, the clinical course of PKD patients with no apparent family history is not well documented in the literature. METHODS: We evaluated the relationship between genotype and the clinical course of 62 PKD patients with no apparent family history. RESULTS: The annual decline of renal function was faster in the patients with PKD1/PKD2 mutation (PKD1 truncating [-3.08; 95% CI -5.30 to -0.87, p = 0.007], PKD1 nontruncating [-2.10; -3.82 to -0.38, p = 0.02], and PKD2 [-2.31; -4.40 to -0.23, p = 0.03]) than in the other patients without PKD1/PKD2 mutation. Similar results were obtained after adjustment for gender, age, estimated glomerular filtration rate (eGFR), height-adjusted total kidney volume (TKV), and mean arterial pressure (MAP). There was no significant difference in the annual decline of renal function among the different PKD1/PKD2 groups, but Kaplan-Meier analysis showed that progression to eGFR < 15 mL/min/1.73 m2 was significantly faster in PKD1 truncating group (p = 0.05). The annual rate of TKV increase was larger in the patients with PKD1/PKD2 mutation (PKD1 truncating [4.63; 95% CI 0.62-8.64, p = 0.03], PKD1 nontruncating [3.79; 0.55-7.03, p = 0.02], and PKD2 [2.11; -1.90 to 6.12, p = 0.29]) than in the other patients without PKD1/PKD2 mutation. Similar results were obtained after adjustment for gender, age, eGFR, and MAP. CONCLUSION: Detection of PKD1/PKD2 mutation, especially PKD1 truncating, is useful for predicting the renal outcome and rate of TKV increase in PKD patients with no apparent family history.


Asunto(s)
Fallo Renal Crónico/epidemiología , Riñón/fisiopatología , Enfermedades Renales Poliquísticas/genética , Canales Catiónicos TRPP/genética , Adulto , Anciano , Análisis Mutacional de ADN , Progresión de la Enfermedad , Estudios de Factibilidad , Femenino , Pruebas Genéticas , Genotipo , Tasa de Filtración Glomerular/genética , Humanos , Fallo Renal Crónico/fisiopatología , Fallo Renal Crónico/terapia , Masculino , Anamnesis , Persona de Mediana Edad , Mutación , Enfermedades Renales Poliquísticas/fisiopatología , Enfermedades Renales Poliquísticas/terapia , Valor Predictivo de las Pruebas , Terapia de Reemplazo Renal/estadística & datos numéricos , Estudios Retrospectivos , Medición de Riesgo/métodos
17.
Pediatr Nephrol ; 34(6): 1065-1075, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30666461

RESUMEN

BACKGROUND: HNF1B gene mutations are an important cause of bilateral (cystic) dysplasia in children, complicated by chronic renal insufficiency. The clinical variability, the absence of genotype-phenotype correlations, and limited long-term data render counseling of affected families difficult. METHODS: Longitudinal data of 62 children probands with genetically proven HNF1B nephropathy was obtained in a multicenter approach. Genetic family cascade screening was performed in 30/62 cases. RESULTS: Eighty-seven percent of patients had bilateral dysplasia, 74% visible bilateral, and 16% unilateral renal cysts at the end of observation. Cyst development was non-progressive in 72% with a mean glomerular filtration rate (GFR) loss of - 0.33 ml/min/1.73m2 per year (± 8.9). In patients with an increase in cyst number, the annual GFR reduction was - 2.8 ml/min/1.73m2 (± 13.2), in the total cohort - 1.0 ml/min/1.73m2 (±10.3). A subset of HNF1B patients differs from this group and develops end stage renal disease (ESRD) at very early ages < 2 years. Hyperuricemia (37%) was a frequent finding at young age (median 1 year), whereas hypomagnesemia (24%), elevated liver enzymes (21%), and hyperglycemia (8%) showed an increased incidence in the teenaged child. Genetic analysis revealed no genotype-phenotype correlations but a significant parent-of-origin effect with a preponderance of 81% of maternal inheritance in dominant cases. CONCLUSIONS: In most children, HNF1B nephropathy has a non-progressive course of cyst development and a slow-progressive course of kidney function. A subgroup of patients developed ESRD at very young age < 2 years requiring special medical attention. The parent-of-origin effect suggests an influence of epigenetic modifiers in HNF1B disease.


Asunto(s)
Factor Nuclear 1-beta del Hepatocito/genética , Enfermedades Renales Poliquísticas/genética , Enfermedades Renales Poliquísticas/patología , Enfermedades Renales Poliquísticas/fisiopatología , Adolescente , Edad de Inicio , Niño , Preescolar , Progresión de la Enfermedad , Femenino , Estudios de Asociación Genética , Alemania , Humanos , Lactante , Recién Nacido , Fallo Renal Crónico/genética , Masculino , Fenotipo , Sistema de Registros
18.
J Dev Orig Health Dis ; 10(2): 154-163, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30274564

RESUMEN

Maternal insufficiency during fetal development can have long-lasting effects on the offspring, most notably on nephron endowment. In polycystic kidney disease (PKD), variability in severity of disease is observed and maternal environment may be a modifying factor. In this study, we first established that in a rodent model of PKD, the Lewis polycystic kidney (LPK) rat's nephron numbers are 25% lower compared with wildtype animals. We then investigated the effects of prenatal and postnatal maternal environment on phenotype and nephron number. LPK pups born from and raised by homozygous LPK dams (control) were compared with LPK pups cross-fostered onto heterozygous LPK dams to improve postnatal environment; with LPK pups born from and raised by heterozygous LPK dams to improve both prenatal and postnatal environment and with LPK pups born from and raised by Wistar Kyoto-LPK heterozygous dams to improve both prenatal and postnatal environment on a different genetic background. Improvement in both prenatal and postnatal environment improved postnatal growth, renal function and reduced blood pressure, most notably in animals with different genetic background. Animals with improved postnatal environment only showed improved growth and blood pressure, but to a lesser extent. All intervention groups showed increased nephron number compared with control LPK. In summary, prenatal and postnatal environment had significant effect in delaying progression and reducing severity of PKD, including nephron endowment.


Asunto(s)
Desarrollo Fetal/genética , Hipertensión/fisiopatología , Quinasas Relacionadas con NIMA/genética , Nefronas/fisiopatología , Enfermedades Renales Poliquísticas/genética , Animales , Animales Recién Nacidos/fisiología , Presión Sanguínea/fisiología , Modelos Animales de Enfermedad , Femenino , Heterocigoto , Homocigoto , Hipertensión/etiología , Lactancia/fisiología , Masculino , Ratones Transgénicos , Mutación , Nefronas/crecimiento & desarrollo , Nefronas/patología , Enfermedades Renales Poliquísticas/complicaciones , Enfermedades Renales Poliquísticas/fisiopatología , Embarazo , Ratas , Ratas Endogámicas Lew , Índice de Severidad de la Enfermedad
19.
J Physiol ; 597(2): 499-519, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30242840

RESUMEN

KEY POINTS: Significant and selective up-regulation of the Na+ /H+ exchanger NHA2 (SLC9B2) was observed in cysts of patients with autosomal dominant polycystic kidney disease. Using the MDCK cell model of cystogenesis, it was found that NHA2 increases cyst size. Silencing or pharmacological inhibition of NHA2 inhibits cyst formation in vitro. Polycystin-1 represses NHA2 expression via Ca2+ /NFAT signalling whereas the dominant negative membrane-anchored C-terminal fragment (PC1-MAT) increased NHA2 levels. Drugs (caffeine, theophylline) and hormones (vasopressin, aldosterone) known to exacerbate cysts elicit NHA2 expression. Taken together, the findings reveal NHA2 as a potential new player in salt and water homeostasis in the kidney and in the pathogenesis of polycystic kidney disease. ABSTRACT: Autosomal dominant polycystic kidney disease (ADPKD) is caused by mutations in PKD1 and PKD2 encoding polycystin-1 (PC1) and polycystin-2 (PC2), respectively. The molecular pathways linking polycystins to cyst development in ADPKD are still unclear. Intracystic fluid secretion via ion transporters and channels plays a crucial role in cyst expansion in ADPKD. Unexpectedly, we observed significant and selective up-regulation of NHA2, a member of the SLC9B family of Na+ /H+ exchangers, that correlated with cyst size and disease severity in ADPKD patients. Using three-dimensional cultures of MDCK cells to model cystogenesis in vitro, we showed that ectopic expression of NHA2 is causal to increased cyst size. Induction of PC1 in MDCK cells inhibited NHA2 expression with concordant inhibition of Ca2+ influx through store-dependent and -independent pathways, whereas reciprocal activation of Ca2+ influx by the dominant negative membrane-anchored C-terminal tail fragment of PC1 elevated NHA2. We showed that NHA2 is a target of Ca2+ /NFAT signalling and is transcriptionally induced by methylxanthine drugs such as caffeine and theophylline, which are contraindicated in ADPKD patients. Finally, we observed robust induction of NHA2 by vasopressin, which is physiologically consistent with increased levels of circulating vasopressin and up-regulation of vasopressin V2 receptors in ADPKD. Our findings have mechanistic implications on the emerging use of vasopressin V2 receptor antagonists such as tolvaptan as safe and effective therapy for polycystic kidney disease and reveal a potential new regulator of transepithelial salt and water transport in the kidney.


Asunto(s)
Antiportadores/genética , Enfermedades Renales Poliquísticas , Animales , Antiportadores/metabolismo , Técnicas de Cultivo de Célula , Quistes , Perros , Células HEK293 , Homeostasis , Humanos , Riñón/metabolismo , Riñón/fisiopatología , Células de Riñón Canino Madin Darby , Modelos Biológicos , Enfermedades Renales Poliquísticas/genética , Enfermedades Renales Poliquísticas/metabolismo , Enfermedades Renales Poliquísticas/fisiopatología
20.
Clin Exp Nephrol ; 23(4): 455-464, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30426292

RESUMEN

BACKGROUND: Tolvaptan is an effective treatment for polycystic kidney disease (PKD), but also causes unfortunate polyuria. Hydrochlorothiazide (HCTZ) has been shown to reduce urine volume in nephrogenic diabetes insipidus, raising the possibility that HCTZ could also be effective in reducing tolvaptan-induced polyuria. In this study, we examined the combined administration of HCTZ and tolvaptan. METHODS: Male PCK rats were divided into four groups of normal chow (Cont), normal chow plus tolvaptan, gavage HCTZ treatment, and tolvaptan + HCTZ. Biochemical examinations of the plasma and urine were performed as well as histological and molecular (mRNA and protein expression) analyses. RESULTS: Groups treated with tolvaptan had significantly higher 24 h urine excretion, which was significantly reduced in the tolvaptan + HCTZ group after 2 weeks. Cyst size, pERK protein expression, and Cyclin D1 mRNA expression were all significantly reduced in both the tolvaptan and tolvaptan + HCTZ groups, indicating that HCTZ did not affect the beneficial functions of tolvaptan. Notably, aquaporin 2 redistribution from the apical to intracellular domains was observed in tolvaptan-treated rats and was partially reversed in the tolvaptan + HCTZ group. The renal glomerular filtration rate was reduced in the tolvaptan + HCTZ group. Significantly lowered mRNA expression of neuronal nitric oxide synthase, prostaglandin E synthase 2 and renin were also found in the medulla, but not in the cortex. CONCLUSION: HCTZ reduces tolvaptan-induced polyuria without altering its beneficial effects on PKD. This novel therapeutic combination could potentially lead to better PKD treatments and improved quality of life for the affected patients.


Asunto(s)
Antagonistas de los Receptores de Hormonas Antidiuréticas/uso terapéutico , Diuréticos/uso terapéutico , Hidroclorotiazida/uso terapéutico , Enfermedades Renales Poliquísticas/tratamiento farmacológico , Poliuria/tratamiento farmacológico , Tolvaptán/uso terapéutico , Animales , Antagonistas de los Receptores de Hormonas Antidiuréticas/efectos adversos , Acuaporina 2/metabolismo , Presión Sanguínea/efectos de los fármacos , Quimioterapia Combinada , Expresión Génica/efectos de los fármacos , Tasa de Filtración Glomerular , Masculino , Óxido Nítrico Sintasa de Tipo I/genética , Enfermedades Renales Poliquísticas/fisiopatología , Poliuria/inducido químicamente , Prostaglandina-E Sintasas/genética , ARN Mensajero/metabolismo , Ratas , Renina/genética , Tolvaptán/efectos adversos , Orina
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA