Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 96
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Artículo en Inglés | MEDLINE | ID: mdl-38877850

RESUMEN

Hereditary angioedema (HAE) is a rare hereditary disorder characterized by episodic swelling and life-threatening airway obstruction caused by laryngeal angioedema. In most HAE patients, reduced level of serum C1-Inhibitor (type-I-HAE) or presence of aberrant C1-Inhibitor (type-II-HAE) result in the lost of regulation of the complementary system and contact activation system with downstream over-activation of bradykinin - the chief mediator leading to angioedema. Type-III HAE (HAE-nl-C1INH) is rare without deficient or dysfunction of C1-Inhibitor, often with genetic aberrant related to the contact activation system. The prevalence of HAE in the population is estimated at 1 in 50,000 individuals, often with early onset, but due to the heterogeneity of the disease, there is frequently a significant delay in diagnosis. Recently, better awareness by physicians, more access to diagnostic tools, better management and prophylaxis has decreased morbidity and mortality. A focus in HAE patient care shift from management of attacks with on-demand medication, to use of prophylaxis to reduce attacks has improved the overall quality of life of patients with HAE. One area in HAE research that has not been emphasized is the long-term consequence of C1-INH deficiency in HAE patients, other than the typical manifestations of HAE, as evidence have emerged linking this disorder with increased risk of cardiovascular diseases, auto-immune disorders, and malignancy. This review aims to gather the current knowledge and evidence of potential consequence of C1-Inhibitor deficiency in HAE aside from angioedema with emphasis in the improvement of long-term care and overall quality of life for HAE patients.

2.
Kidney Int ; 94(3): 524-535, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30045814

RESUMEN

Multiple large clinical trials have shown that sodium-glucose cotransporter (SGLT) 2 inhibitors reduce the risk of renal events. However, the mechanism responsible for this outcome remains unknown. Here we investigated the effects of the SGLT2 inhibitor luseogliflozin on the development of renal fibrosis after renal ischemia/reperfusion injury in non-diabetic mice. Luseogliflozin significantly suppressed development of renal fibrosis, prevented peritubular capillary congestion/hemorrhage, attenuated CD31-positive cell loss, suppressed hypoxia, and increased vascular endothelial growth factor (VEGF)-A expression in the kidney after ischemia/reperfusion injury. Luseogliflozin failed to induce the above-mentioned protection in animals co-treated with sunitinib, a VEGF receptor inhibitor. Additionally, luseogliflozin reduced glucose uptake and increased VEGF-A expression in the kidneys of glucose transporter 2 (GLUT2)-downregulated mice following ischemia/reperfusion and in GLUT2-knock-down cells compared with those in normal controls. Withdrawal of glucose from cultured medium, to halt glucose uptake, remarkably increased VEGF-A expression and reversed the luseogliflozin-induced increase in VEGF-A expression in the proximal tubular cells. Thus, luseogliflozin prevented endothelial rarefaction and subsequent renal fibrosis after renal ischemia/reperfusion injury through a VEGF-dependent pathway induced by the dysfunction of proximal tubular glucose uptake in tubules with injury-induced GLUT2 downregulation.


Asunto(s)
Lesión Renal Aguda/tratamiento farmacológico , Túbulos Renales Proximales/patología , Inhibidores del Cotransportador de Sodio-Glucosa 2/farmacología , Transportador 2 de Sodio-Glucosa/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Lesión Renal Aguda/etiología , Lesión Renal Aguda/patología , Inhibidores de la Angiogénesis/farmacología , Animales , Glucemia/metabolismo , Capilares/efectos de los fármacos , Capilares/metabolismo , Capilares/patología , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Fibrosis , Técnicas de Silenciamiento del Gen , Transportador de Glucosa de Tipo 2/genética , Transportador de Glucosa de Tipo 2/metabolismo , Humanos , Túbulos Renales Proximales/irrigación sanguínea , Túbulos Renales Proximales/efectos de los fármacos , Túbulos Renales Proximales/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Daño por Reperfusión/complicaciones , Daño por Reperfusión/patología , Inhibidores del Cotransportador de Sodio-Glucosa 2/uso terapéutico , Sorbitol/análogos & derivados , Sorbitol/farmacología , Sorbitol/uso terapéutico , Sunitinib/farmacología , Resultado del Tratamiento
3.
J Pharmacol Sci ; 137(2): 220-223, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29983235

RESUMEN

We aimed to examine the effects of a sodium glucose co-transporter 2 (SGLT2) inhibitor on systemic and intrarenal renin-angiotensin system (RAS) in subtotally nephrectomized non-diabetic rats, a model of chronic kidney disease (CKD). Oral administration of the selective SGLT2 inhibitor, TA-1887 (10 mg/kg/day), for 10 weeks induced glycosuria. However, plasma renin activity, plasma angiotensinogen levels, kidney angiotensin II contents and renal injury were not significantly affected by TA-1887. These data indicate that chronic treatment with an SGLT2 inhibitor does not activate the systemic and intrarenal RAS in subjects with non-diabetic CKD.


Asunto(s)
Glucósidos/uso terapéutico , Indoles/uso terapéutico , Riñón/metabolismo , Insuficiencia Renal Crónica/metabolismo , Sistema Renina-Angiotensina/efectos de los fármacos , Inhibidores del Cotransportador de Sodio-Glucosa 2 , Administración Oral , Angiotensina II/metabolismo , Angiotensinógeno/sangre , Animales , Modelos Animales de Enfermedad , Glucosuria/inducido químicamente , Masculino , Nefrectomía , Ratas Sprague-Dawley , Renina/sangre , Transportador 2 de Sodio-Glucosa
4.
Exp Cell Res ; 358(2): 343-351, 2017 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-28689812

RESUMEN

Hypoxia predisposes renal fibrosis. This study was conducted to identify novel approaches to ameliorate the pathogenic effect of hypoxia. Using human proximal tubular epithelial cells we showed that a pan-phosphodiesterase (PDE) inhibitor, 3-isobutyl-1-methylxanthine (IBMX) dose and time dependently downregulated hypoxia-inducible factor 1α (HIF-1α) mRNA expression, which was further augmented by addition of a transcriptional inhibitor, actinomycin D. IBMX also increased the cellular cyclic adenosine monophosphate (cAMP) level. Luciferase assay showed that blocking of protein kinase A (PKA) using H89 reduced, while 8-Br-cAMP agonized the repression of HIF-1α promoter activity in hypoxic condition. Deletion of cAMP response element binding sites from the HIF-1α promoter abrogated the effect of IBMX. Western blot and immunofluorescent study confirmed that the CoCl2 induced increased HIF-1α protein in whole cell lysate and in nucleus was reduced by the IBMX. Through this process, IBMX attenuated both CoCl2 and hypoxia induced mRNA expressions of two pro-fibrogenic factors, platelet-derived growth factor B and lysyl oxidase. Moreover, IBMX reduced production of a mesenchymal transformation factor, ß-catenin; as well as protected against hypoxia induced cell-death. Taken together, our study showed novel evidence that the PDE inhibitor IBMX can downregulate the transcription of HIF-1α, and thus may attenuate hypoxia induced renal fibrosis.


Asunto(s)
1-Metil-3-Isobutilxantina/farmacología , Células Epiteliales/efectos de los fármacos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Hipoxia de la Célula/efectos de los fármacos , Células Cultivadas , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Células Epiteliales/metabolismo , Humanos , Proteínas Proto-Oncogénicas c-sis/metabolismo , Transducción de Señal/efectos de los fármacos , Xantinas/farmacología , beta Catenina/metabolismo
5.
J Pharmacol Sci ; 2017 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-29110957

RESUMEN

The kidney expresses protease-activated receptor-1 (PAR-1). PAR-1 is known as a thrombin receptor, but its role in kidney injury is not well understood. In this study, we examined the contribution of PAR-1 to kidney glomerular injury and the effects of its inhibition on development of nephropathy. Mice were divided into 3 groups: control, doxorubicin + vehicle (15 mg/kg doxorubicin and saline) and doxorubicin + Q94 (doxorubicin at 15 mg/kg and the PAR-1 antagonist Q94 at 5 mg/kg/d) groups. Where indicated, doxorubicin was administered intravenously and PAR-1 antagonist or saline vehicle by subcutaneous osmotic mini-pump. PAR-1 expression was increased in glomeruli of mice treated with doxorubicin. Q94 treatment significantly suppressed the increased albuminuria in these nephropathic mice. Pathological analysis showed that Q94 treatment significantly attenuated periodic acid-Schiff and desmin staining, indicators of podocyte injury, and also decreased glomerular levels of podocin and nephrin. Furthermore, thrombin increased intracellular calcium levels in podocytes. This increase was suppressed by Q94 and Rox4560, a transient receptor potential cation channel (TRPC)3/6 antagonist. In addition, both Q94 and Rox4560 suppressed the doxorubicin-induced increase in activities of caspase-9 and caspase-3 in podocytes. These data suggested that PAR-1 contributes to development of podocyte and glomerular injury and that PAR-1 antagonists have therapeutic potential.

6.
Clin Exp Pharmacol Physiol ; 44(4): 522-525, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28063156

RESUMEN

Metabolic syndrome is often associated with disruption of circadian rhythm of systemic haemodynamics and cardiovascular disease. Experiments were conducted to investigate the effects of luseogliflozin, a selective SGLT2 inhibitor, on circadian rhythm of sympathetic nervous function and locomotor activity (LA) in metabolic syndrome rats. The difference in the low frequency component of systolic blood pressure between the dark and light period significantly increased in the luseogliflozin-treated SHRcp. LA also increased in the dark period compared with the light period following luseogliflozin treatment. These data suggest that circadian rhythm of sympathetic nervous function and LA is improved by luseogliflozin in metabolic syndrome rats, which may contribute to SGLT2 inhibitor-induced improvement of cardiovascular outcomes.


Asunto(s)
Ritmo Circadiano/efectos de los fármacos , Locomoción/efectos de los fármacos , Síndrome Metabólico/fisiopatología , Inhibidores del Cotransportador de Sodio-Glucosa 2 , Sorbitol/análogos & derivados , Sistema Nervioso Simpático/efectos de los fármacos , Sistema Nervioso Simpático/fisiopatología , Animales , Masculino , Ratas , Sorbitol/farmacología
7.
Int J Mol Sci ; 17(11)2016 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-27801805

RESUMEN

This study was performed to demonstrate urinary angiotensinogen as a potential prognostic marker of the albuminuria reduction effects of olmesartan in patients with metabolic syndrome. In 24 patients (eight women, 57.88 ± 2.00 years), 5-40 mg/day of olmesartan were given. Urinary concentrations of albumin and angiotensinogen (normalized by urinary concentrations of creatinine) and plasma renin activity were measured before and after the 12- and 24-week marks of olmesartan treatment. Olmesartan treatment increased plasma renin activity and decreased urinary albumin and urinary angiotensinogen significantly (p < 0.05). Based on the % change in urinary albumin, patients were divided into two groups, responders (<-50%) and non-responders (≥-50%), and a logistic analysis of urinary angiotensinogen before treatment showed the area under the curve as 0.694. When the cutoff value of urinary angiotensinogen before the treatment of 13.9 µg/g Cr was used, the maximum Youden index (0.500, specificity: 11/12 = 91.7% and sensitivity: 7/12 = 58.3%) was obtained. When all patients were re-divided into two groups, those with higher values of urinary angiotensinogen before the treatment (Group H, n = 16) and those with lower values, Group H showed significantly decreased urinary albumin (p < 0.05). Therefore, urinary angiotensinogen could be a prognostic marker of the albuminuria reduction effects of olmesartan in patients with metabolic syndrome.


Asunto(s)
Albuminuria/orina , Angiotensinógeno/orina , Síndrome Metabólico/tratamiento farmacológico , Síndrome Metabólico/orina , Adulto , Albuminuria/tratamiento farmacológico , Albuminuria/patología , Biomarcadores Farmacológicos/orina , Creatinina/orina , Femenino , Humanos , Imidazoles/administración & dosificación , Masculino , Síndrome Metabólico/patología , Persona de Mediana Edad , Pronóstico , Tetrazoles/administración & dosificación
8.
Biochem Biophys Res Commun ; 448(2): 175-81, 2014 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-24780397

RESUMEN

INTRODUCTION: Carnitine palmitoyltransferase II (CPT II) deficiency is an inherited disorder involving ß-oxidation of long-chain fatty acids (FAO), which leads to rhabdomyolysis and subsequent acute renal failure. The detailed mechanisms of disease pathogenesis remain unknown; however, the availability of relevant human cell types for investigation, such as skeletal muscle cells, is limited, and the development of novel disease models is required. METHODS: We generated human induced pluripotent stem cells (hiPSCs) from skin fibroblasts of a Japanese patient with CPT II deficiency. Mature myocytes were differentiated from the patient-derived hiPSCs by introducing myogenic differentiation 1 (MYOD1), the master transcriptional regulator of myocyte differentiation. Using an in vitro acylcarnitine profiling assay, we investigated the effects of a hypolipidemic drug, bezafibrate, and heat stress on mitochondrial FAO in CPT II-deficient myocytes and controls. RESULTS: CPT II-deficient myocytes accumulated more palmitoylcarnitine (C16) than did control myocytes. Heat stress, induced by incubation at 38°C, leads to a robust increase of C16 in CPT II-deficient myocytes, but not in controls. Bezafibrate reduced the amount of C16 in control and CPT II-deficient myocytes. DISCUSSION: In this study, we induced differentiation of CPT II-deficient hiPSCs into mature myocytes in a highly efficient and reproducible manner and recapitulated some aspects of the disease phenotypes of CPT II deficiency in the myocyte disease models. This approach addresses the challenges of modeling the abnormality of FAO in CPT II deficiency using iPSC technology and has the potential to revolutionize translational research in this field.


Asunto(s)
Carnitina O-Palmitoiltransferasa/deficiencia , Errores Innatos del Metabolismo/metabolismo , Errores Innatos del Metabolismo/patología , Células Musculares/patología , Células Madre Pluripotentes/patología , Bezafibrato/farmacología , Carnitina/análogos & derivados , Carnitina/metabolismo , Carnitina O-Palmitoiltransferasa/genética , Carnitina O-Palmitoiltransferasa/metabolismo , Diferenciación Celular , Células Cultivadas , Fibroblastos/patología , Regulación de la Expresión Génica , Humanos , Masculino , Células Musculares/efectos de los fármacos , Células Musculares/metabolismo , Palmitoilcarnitina/metabolismo , Células Madre Pluripotentes/metabolismo , Adulto Joven
9.
Int J Med Sci ; 11(3): 222-5, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24516344

RESUMEN

We examined the effects of overexpressed human chymase on survival and activity in lipopolysaccharide (LPS)-treated mice. Human chymase transgenic (Tg) and wild-type C57BL/6 (WT) mice were treated with LPS (0.03, 0.1 and 0.3 mg/day; intraperitoneal) for 2 weeks. Treatment with 0.03 mg LPS did not affect survival in either WT or Tg mice. WT mice were not affected by 0.1 mg/day of LPS, whereas 25% of Tg mice died. Survival of mice treated with 0.3 mg/day of LPS was 87.5% and 0% in WT and Tg, respectively. LPS-induced increases in chymase activity in the heart and skin were significantly greater in Tg than WT mice. These data suggest a possible contribution of human chymase activation to LPS-induced mortality.


Asunto(s)
Quimasas , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Miocardio/enzimología , Piel/enzimología , Animales , Quimasas/biosíntesis , Quimasas/genética , Humanos , Lipopolisacáridos/toxicidad , Ratones Transgénicos , Piel/efectos de los fármacos , Sobrevida
10.
Mol Genet Metab Rep ; 40: 101096, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38872960

RESUMEN

Citrin deficiency is a congenital secondary urea cycle disorder lacking useful disease models for effective treatment development. In this study, human induced pluripotent stem cells (iPSCs) were generated from two patients with citrin deficiency and differentiated into hepatocyte-like cells (HLCs). Citrin-deficient HLCs produced albumin and liver-specific markers but completely lacked citrin protein and expressed argininosuccinate synthase only weakly. In addition, ammonia concentrations in a medium cultured with citrin-deficient HLCs were higher than with control HLCs. Sodium pyruvate administration significantly reduced ammonia concentrations in the medium of citrin-deficient HLCs and slightly reduced ammonia in HLCs differentiated from control iPSCs, though this change was not significant. Our results suggest that sodium pyruvate may be an efficient treatment for patients with citrin deficiency. Citrin-deficient iPSCs are a pathological liver model for congenital urea cycle disorders to clarify pathogenesis and develop novel therapies.

11.
Circulation ; 125(11): 1402-13, 2012 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-22328542

RESUMEN

BACKGROUND: The presence of chronic kidney disease is a significant independent risk factor for poor prognosis in patients with chronic heart failure. However, the mechanisms and mediators underlying this interaction are poorly understood. In this study, we tested our hypothesis that chronic cardiac volume overload leads to de novo renal dysfunction by coactivating the sympathetic nervous system and renin-angiotensin system in the kidney. We also examined the therapeutic potential of renal denervation and renin-angiotensin system inhibition to suppress renal injury in chronic heart failure. METHODS AND RESULTS: Sprague-Dawley rats underwent aortic regurgitation and were treated for 6 months with vehicle, olmesartan (an angiotensin II receptor blocker), or hydralazine. At 6 months, albuminuria and glomerular podocyte injury were significantly increased in aortic regurgitation rats. These changes were associated with increased urinary angiotensinogen excretion, kidney angiotensin II and norepinephrine (NE) levels, and enhanced angiotensinogen and angiotensin type 1a receptor gene expression and oxidative stress in renal cortical tissues. Aortic regurgitation rats with renal denervation had decreased albuminuria and glomerular podocyte injury, which were associated with reduced kidney NE, angiotensinogen, angiotensin II, and oxidative stress. Renal denervation combined with olmesartan prevented podocyte injury and albuminuria induced by aortic regurgitation. CONCLUSIONS: In this chronic cardiac volume-overload animal model, activation of the sympathetic nervous system augments kidney renin-angiotensin system and oxidative stress, which act as crucial cardiorenal mediators. Renal denervation and olmesartan prevent the onset and progression of renal injury, providing new insight into the treatment of cardiorenal syndrome.


Asunto(s)
Albuminuria/prevención & control , Insuficiencia de la Válvula Aórtica/prevención & control , Riñón/inervación , Podocitos/patología , Simpatectomía , Albuminuria/complicaciones , Albuminuria/patología , Animales , Insuficiencia de la Válvula Aórtica/complicaciones , Insuficiencia de la Válvula Aórtica/patología , Línea Celular Transformada , Humanos , Riñón/patología , Riñón/fisiología , Masculino , Podocitos/fisiología , Ratas , Ratas Sprague-Dawley , Simpatectomía/métodos
12.
Arterioscler Thromb Vasc Biol ; 32(2): 257-63, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22173225

RESUMEN

OBJECTIVE: We previously showed that aldosterone induces insulin resistance in rat vascular smooth muscle cells (VSMCs). Because insulin-like growth factor-1 receptor (IGF1R) affects insulin signaling, we hypothesized that aldosterone induces vascular insulin resistance and remodeling via upregulation of IGF1R and its hybrid insulin/insulin-like growth factor-1 receptor. METHODS AND RESULTS: Hybrid receptor expression was measured by immunoprecipitation. Hypertrophy of VSMCs was evaluated by (3)H-labeled leucine incorporation. Aldosterone (10 nmol/L) significantly increased protein and mRNA expression of IGF1R and hybrid receptor in VSMCs but did not affect insulin receptor expression. Mineralocorticoid receptor blockade with eplerenone inhibited aldosterone-induced increases in IGF1R and hybrid receptor. Aldosterone augmented insulin (100 nmol/L)-induced extracellular signal-regulated kinase 1/2 phosphorylation. Insulin-induced leucine incorporation and α-smooth muscle actin expression were also augmented by aldosterone in VSMCs. These aldosterone-induced changes were significantly attenuated by eplerenone or picropodophyllin, an IGF1R inhibitor. Chronic infusion of aldosterone (0.75 µg/hour) increased blood pressure and aggravated glucose metabolism in rats. Expression of hybrid receptor, azan-positive area, and oxidative stress in aorta was increased in aldosterone-infused rats. Spironolactone and tempol prevented these aldosterone-induced changes. CONCLUSIONS: Aldosterone induces vascular remodeling through IGF1R- and hybrid receptor-dependent vascular insulin resistance. Mineralocorticoid receptor blockade may attenuate angiopathy in hypertensive patients with hyperinsulinemia.


Asunto(s)
Aldosterona/farmacología , Resistencia a la Insulina , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Receptor IGF Tipo 1/metabolismo , Receptor de Insulina/metabolismo , Animales , Aorta Torácica/efectos de los fármacos , Aorta Torácica/metabolismo , Aorta Torácica/patología , Presión Sanguínea/efectos de los fármacos , Células Cultivadas , Quimera/genética , Eplerenona , Glucosa/metabolismo , Hipertrofia , Insulina/farmacología , Masculino , Modelos Animales , Músculo Liso Vascular/patología , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Receptor IGF Tipo 1/genética , Receptor de Insulina/genética , Espironolactona/análogos & derivados , Espironolactona/farmacología , Regulación hacia Arriba/efectos de los fármacos
13.
J Pharmacol Sci ; 122(3): 176-83, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23800993

RESUMEN

A growing body of evidence suggests that podocyte apoptosis is a major cause of decreased podocyte number, which leads to albuminuria and glomerular injury. The aim of this study was to clarify the molecular mechanisms of angiotensin II (Ang II)-induced apoptosis in cultured mouse podocytes. We examined the effects of Ang II (100 nmol/L) on apoptosis, superoxide anions, and cytosol pH in podocytes. For intracellular pH measurements, image analysis was conducted using confocal laser microscopy after incubation with carboxy-seminaphthorhodafluor-1. Superoxide anions and intracellular pH were elevated with Ang II treatment. Apoptotic cell numbers, as measured by TUNEL staining and caspase 3 activity, were also augmented in the Ang II-treated group. Pre-treatment with olmesartan (100 nmol/L, an Ang II type 1-receptor blocker), apocynin (50 µmol/L, NADPH oxidase inhibitor), or 5-N,N hexamethylene amiloride [30 µmol/L, Na⁺/H⁺ exchanger type 1 (NHE-1) inhibitor] abolished Ang II-induced podocyte apoptosis, whereas NHE-1 mRNA and protein expression was not affected by Ang II treatment. Moreover, Ang II increased NHE-1 phosphorylation. These results suggest that superoxide production, NHE-1 activation, and intracellular alkalization were early features prior to apoptosis in Ang II-treated mouse podocytes, and may offer new insights into the mechanisms responsible for Ang II-induced podocyte injury.


Asunto(s)
Angiotensina II/efectos adversos , Apoptosis/efectos de los fármacos , Proteínas de Transporte de Catión/fisiología , Podocitos/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Intercambiadores de Sodio-Hidrógeno/fisiología , Animales , Caspasa 3/metabolismo , Proteínas de Transporte de Catión/metabolismo , Células Cultivadas , Citosol/metabolismo , Concentración de Iones de Hidrógeno , Ratones , NADPH Oxidasas/fisiología , Fosforilación/efectos de los fármacos , Podocitos/citología , Intercambiador 1 de Sodio-Hidrógeno , Intercambiadores de Sodio-Hidrógeno/metabolismo
14.
J Pharmacol Sci ; 121(2): 95-102, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23337436

RESUMEN

We examined the effects of angiotensin II AT1-receptor blockade with olmesartan on high fat (HF) diet-induced vascular oxidative stress and endothelial dysfunction in normal salt (NS) diet-fed Dahl salt-sensitive (DSS) rats. Treatment with NS + HF diet (32% crude fat, 0.3% NaCl) for 20 weeks significantly increased blood pressure in DSS rats. NS + HF diet-fed DSS rats also showed higher plasma levels of thiobarbituric acid-reactive substances, aortic superoxide production, and mRNA levels of p22(phox) and gp91(phox) in aortic tissues than NS diet-fed DSS rats. Furthermore, acetylcholine-induced vasorelaxation of aorta from NS + HF diet-fed DSS rats was significantly reduced. In NS + HF diet-fed DSS rats, treatment with olmesartan medoxomil (10 mg/kg per day, p.o.) and hydralazine (25 mg/kg per day, p.o.) similarly decreased blood pressure. However, in these animals, only olmesartan normalized plasma levels of thiobarbituric acid-reactive substances, vascular superoxide in aortic tissues, and acetylcholine-induced vasorelaxation. These data indicate that HF diet-induced hypertension is associated with vascular oxidative stress and endothelial dysfunction in NS diet-treated DSS rats. Inhibition of angiotensin II AT1 receptors may elicit beneficial effects on HF-induced hypertension and vascular injury in subjects that have genetically enhanced sodium-sensitive blood pressure.


Asunto(s)
Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Dieta Alta en Grasa/efectos adversos , Endotelio Vascular/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Receptor de Angiotensina Tipo 1/metabolismo , Animales , Antihipertensivos/farmacología , Aorta/efectos de los fármacos , Aorta/metabolismo , Presión Sanguínea/efectos de los fármacos , Presión Sanguínea/fisiología , Endotelio Vascular/metabolismo , Hidralazina/farmacología , Hipertensión/inducido químicamente , Hipertensión/metabolismo , Hipertensión/fisiopatología , Imidazoles/farmacología , Masculino , Glicoproteínas de Membrana/metabolismo , NADPH Oxidasa 2 , NADPH Oxidasas/biosíntesis , NADPH Oxidasas/metabolismo , Olmesartán Medoxomilo , Ratas , Ratas Endogámicas Dahl , Superóxidos/metabolismo , Tetrazoles/farmacología , Sustancias Reactivas al Ácido Tiobarbitúrico/metabolismo , Vasodilatación/efectos de los fármacos
15.
J Pharmacol Sci ; 122(2): 103-8, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23698111

RESUMEN

Angiotensin II (Ang II) infusion into rats elevates local angiotensin II levels through an AT1 receptor-dependent pathway in the kidney. We examined whether treatment with an angiotensin-converting enzyme (ACE) inhibitor, temocapril, or an AT1-receptor blocker, olmesartan, prevented elevation of Ang II levels in the kidney of angiotensin I (Ang I)-infused rats. Rats were infused with Ang I (100 ng/min) and treated with temocapril (30 mg/kg per day, n = 10) or olmesartan (10 mg/kg per day, n = 9) for 4 weeks. Ang I infusion significantly elevated blood pressure compared with vehicle-infused rats (n = 6). Treatment with temocapril or olmesartan suppressed Ang I-induced hypertension. Temocapril suppressed both plasma and renal ACE activity. Ang I infusion increased Ang II content in the kidney. Interestingly, temocapril failed to reduce the level of Ang II in the kidney, while olmesartan markedly suppressed an increase in renal Ang II levels. These results suggest a limitation of temocapril and a benefit of olmesartan to inhibit the renal renin-angiotensin system and suggest the possible existence of an ACE inhibitor-insensitive pathway that increases Ang II levels in rat kidney.


Asunto(s)
Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Angiotensina II/orina , Angiotensina I/farmacología , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Imidazoles/farmacología , Riñón/metabolismo , Tetrazoles/farmacología , Tiazepinas/farmacología , Angiotensina I/administración & dosificación , Bloqueadores del Receptor Tipo 1 de Angiotensina II/uso terapéutico , Inhibidores de la Enzima Convertidora de Angiotensina/uso terapéutico , Animales , Antihipertensivos/farmacología , Antihipertensivos/uso terapéutico , Presión Sanguínea/efectos de los fármacos , Hipertensión/tratamiento farmacológico , Hipertensión/etiología , Imidazoles/uso terapéutico , Masculino , Peptidil-Dipeptidasa A/metabolismo , Ratas , Ratas Sprague-Dawley , Sistema Renina-Angiotensina/efectos de los fármacos , Sistema Renina-Angiotensina/fisiología , Tetrazoles/uso terapéutico , Tiazepinas/uso terapéutico
16.
J Am Soc Nephrol ; 23(11): 1847-56, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22997258

RESUMEN

Patients and animals with renal injury exhibit increased urinary excretion of angiotensinogen. Although increased tubular synthesis of angiotensinogen contributes to the increased excretion, we do not know to what degree glomerular filtration of systemic angiotensinogen, especially through an abnormal glomerular filtration barrier, contributes to the increase in urinary levels. Here, we used multiphoton microscopy to visualize and quantify the glomerular permeability of angiotensinogen in the intact mouse and rat kidney. In healthy mice and Munich-Wistar-Frömter rats at the early stage of glomerulosclerosis, the glomerular sieving coefficient of systemically infused Atto565-labeled human angiotensinogen (Atto565-hAGT), which rodent renin cannot cleave, was only 25% of the glomerular sieving coefficient of albumin, and its urinary excretion was undetectable. In a more advanced phase of kidney disease, the glomerular permeability of Atto565-hAGT was slightly higher but still very low. Furthermore, unlike urinary albumin, the significantly higher urinary excretion of endogenous rat angiotensinogen did not correlate with either the Atto565-hAGT or Atto565-albumin glomerular sieving coefficients. These results strongly suggest that the vast majority of urinary angiotensinogen originates from the tubules rather than glomerular filtration.


Asunto(s)
Angiotensinógeno/metabolismo , Glomérulos Renales/metabolismo , Angiotensinógeno/administración & dosificación , Angiotensinógeno/orina , Animales , Femenino , Humanos , Túbulos Renales/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía de Fluorescencia por Excitación Multifotónica , Permeabilidad , Ratas , Ratas Endogámicas WF , Sistema Renina-Angiotensina/fisiología
17.
Sci Rep ; 13(1): 20010, 2023 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-37973990

RESUMEN

The mechanistic/mammalian target of rapamycin (mTOR) is involved in a wide range of cellular processes. However, the role of mTOR in podocytes remains unclear. In this study, we aimed to clarify the role of mTOR in podocyte differentiation from human induced pluripotent stem cells (hiPSCs) and to establish an efficient differentiation protocol for human podocytes. We generated podocytes from hiPSCs by modifying protocol. The expression of the podocyte-specific slit membrane components nephrin and podocin was measured using PCR, western blotting, flow cytometry, and immunostaining; and the role of mTOR was evaluated using inhibitors of the mTOR pathway. Nephrin and podocin were found to be expressed in cells differentiated from hiPSCs, and their expression was increased by mTOR inhibitor treatment. S6, a downstream component of the mTOR pathway, was also found to be involved in podocyte differentiation. we evaluated its permeability to albumin, urea, and electrolytes. The induced podocytes were permeable to the small molecules, but only poorly permeable to albumin. We have shown that the mTOR pathway is involved in podocyte differentiation. Our monolayer podocyte differential protocol, using an mTOR inhibitor, provides a novel in vitro model for studies of kidney physiology and pathology.


Asunto(s)
Células Madre Pluripotentes Inducidas , Podocitos , Humanos , Podocitos/metabolismo , Sirolimus/farmacología , Células Madre Pluripotentes Inducidas/metabolismo , Inhibidores mTOR , Riñón/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Diferenciación Celular , Albúminas/metabolismo
18.
Stem Cells Dev ; 32(21-22): 670-680, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37639359

RESUMEN

The parathyroid gland plays an essential role in mineral and bone metabolism. Cultivation of physiological human parathyroid cells has yet to be established and the method by which parathyroid cells differentiate from pluripotent stem cells remains uncertain. Therefore, it has been hard to clarify the mechanisms underlying the onset of parathyroid disorders, such as hyperparathyroidism. In this study, we developed a new method of parathyroid cell differentiation from human induced pluripotent stem (iPS) cells. Parathyroid cell differentiation occurred in accordance with embryologic development. Differentiated cells, which expressed the parathyroid hormone, adopted unique cell aggregation similar to the parathyroid gland. In addition, these differentiated cells were identified as calcium-sensing receptor (CaSR)/epithelial cell adhesion molecule (EpCAM) double-positive cells. Interestingly, stimulation with transforming growth factor-α (TGF-α), which is considered a causative molecule of parathyroid hyperplasia, increased the CaSR/EpCAM double-positive cells, but this effect was suppressed by erlotinib, which is an epidermal growth factor receptor (EGFR) inhibitor. These results suggest that TGF-α/EGFR signaling promotes parathyroid cell differentiation from iPS cells in a similar manner to parathyroid hyperplasia.


Asunto(s)
Células Madre Pluripotentes Inducidas , Glándulas Paratiroides , Humanos , Glándulas Paratiroides/metabolismo , Glándulas Paratiroides/patología , Células Madre Pluripotentes Inducidas/metabolismo , Hiperplasia/metabolismo , Hiperplasia/patología , Factor de Crecimiento Transformador alfa/farmacología , Factor de Crecimiento Transformador alfa/metabolismo , Molécula de Adhesión Celular Epitelial/metabolismo , Molécula de Adhesión Celular Epitelial/farmacología , Receptores ErbB/genética , Receptores ErbB/metabolismo , Diferenciación Celular , Receptores Sensibles al Calcio/genética , Receptores Sensibles al Calcio/metabolismo
19.
J Pharmacol Sci ; 119(4): 359-67, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22863666

RESUMEN

We recently demonstrated that cilnidipine, an L/N-type calcium channel blocker, elicits protective effects against glomerular podocyte injury, in particular, in obese hypertensive rats that express the N-type calcium channel (N-CC). Since the N-CC is known to be expressed in sympathetic nerve endings, we evaluated the reno-protective effects of cilnidipine in innervated and denervated spontaneously hypertensive rats (SHR). Male SHR were uninephrectomized and fed 4% high-salt diet (HS-UNX-SHR). Animals were divided into groups, as follows, and observed from 9 to 27 weeks of age: 1) vehicle (n = 14), 2) vehicle plus renal-denervation (n = 15), 3) cilnidipine (50 mg/kg per day, p.o.; n = 10), and 4) cilnidipine plus renal-denervation (n = 15). Renal denervation attenuated elevations in blood pressure, but failed to suppress urinary protein excretion and podocyte injury in HS-UNX-SHR. Cilnidipine in both innervated and denervated HS-UNX-SHR similarly induced significant antihypertensive effects, as well as suppressing the urinary protein excretion and podocyte injury, compared to vehicle-treated HS-UNX-SHR. These data indicate that renal nerves have a limited contribution to the cilnidipine-induced reno-protective effects in HS-UNX-SHR.


Asunto(s)
Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio Tipo N/fisiología , Dihidropiridinas/farmacología , Podocitos/efectos de los fármacos , Sustancias Protectoras/farmacología , Angiotensina II/metabolismo , Animales , Bloqueadores de los Canales de Calcio/uso terapéutico , Línea Celular , Desmina/metabolismo , Dihidropiridinas/uso terapéutico , Hipertensión/tratamiento farmacológico , Hipertensión/patología , Hipertensión/fisiopatología , Masculino , Ratones , Podocitos/patología , Podocitos/fisiología , Sustancias Protectoras/uso terapéutico , Proteinuria/tratamiento farmacológico , Proteinuria/patología , Proteinuria/fisiopatología , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Sistema Nervioso Simpático/efectos de los fármacos
20.
J Pharmacol Sci ; 119(2): 131-8, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22673148

RESUMEN

We hypothesized that aliskiren provides renoprotection in diabetic animals that did not receive sufficient renoprotection by AT1-receptor antagonist treatment. Type 2 diabetic KKAy mice were treated with group 1: vehicle or group 2: valsartan (15 mg/kg per day) from 12 to 16 weeks of age. The mice were subsequently divided into 4 groups and treated with the following combinations of drugs for another 6 weeks: 1: group 1 kept receiving vehicle, 2: group 2 continuously received 15 mg/kg per day of valsartan (Val-Val15), 3: group 2 received 50 mg/kg per day of valsartan (Val-Val50), 4: group 2 continuously received 15 mg/kg per day of valsartan with 25 mg/kg per day of aliskiren (Val-Val+Ali). Aliskiren exerted significant anti-albuminuric effects, whereas valsartan failed to ameliorate the albuminuria in the first four weeks. Surprisingly, the increasing dosage of valsartan in the Val-Val50 group showed non-significant tendencies to attenuate the albuminuria compared with vehicle infusion. Val-Val+Ali significantly suppressed the development of albuminuria and podocyte injury. Val-Val50 and Val-Val+Ali showed similar suppression of angiotensin II contents in the kidney of KKAy mice. In conclusion, the anti-albuminuric effect that was observed in the type 2 diabetic mice showing no anti-albuminuric effect by valsartan can be attributed to the add-on aliskiren.


Asunto(s)
Amidas/administración & dosificación , Bloqueadores del Receptor Tipo 1 de Angiotensina II/administración & dosificación , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Fumaratos/administración & dosificación , Riñón/efectos de los fármacos , Renina/antagonistas & inhibidores , Tetrazoles/administración & dosificación , Valina/análogos & derivados , Albuminuria/tratamiento farmacológico , Albuminuria/metabolismo , Angiotensina II/metabolismo , Animales , Glucemia/análisis , Presión Sanguínea/efectos de los fármacos , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Quimioterapia Combinada , Riñón/metabolismo , Riñón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , ARN Mensajero/metabolismo , Renina/genética , Valina/administración & dosificación , Valsartán
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA