Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 101
Filtrar
Más filtros

Medicinas Complementárias
Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 22(23)2021 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-34884638

RESUMEN

Diagnosis and cure for rare diseases represent a great challenge for the scientific community who often comes up against the complexity and heterogeneity of clinical picture associated to a high cost and time-consuming drug development processes. Here we show a drug repurposing strategy applied to nephropathic cystinosis, a rare inherited disorder belonging to the lysosomal storage diseases. This approach consists in combining mechanism-based and cell-based screenings, coupled with an affordable computational analysis, which could result very useful to predict therapeutic responses at both molecular and system levels. Then, we identified potential drugs and metabolic pathways relevant for the pathophysiology of nephropathic cystinosis by comparing gene-expression signature of drugs that share common mechanisms of action or that involve similar pathways with the disease gene-expression signature achieved with RNA-seq.


Asunto(s)
Sistemas de Transporte de Aminoácidos Neutros/genética , Cistinosis/tratamiento farmacológico , Cistinosis/genética , Reposicionamiento de Medicamentos , Enfermedades Renales/tratamiento farmacológico , Enfermedades Renales/genética , Enfermedades Raras/tratamiento farmacológico , Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Sistemas de Transporte de Aminoácidos Neutros/efectos de la radiación , Células Cultivadas , Biología Computacional/métodos , Cistinosis/metabolismo , Evaluación Preclínica de Medicamentos/métodos , Humanos , Enfermedades Renales/metabolismo , Túbulos Renales Proximales/efectos de los fármacos , Túbulos Renales Proximales/metabolismo , Túbulos Renales Proximales/patología , Redes y Vías Metabólicas , Enfermedades Raras/genética , Enfermedades Raras/metabolismo , Transcriptoma
2.
Biomed Pharmacother ; 142: 111965, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34385105

RESUMEN

Cisplatin (CDDP) may induce apoptosis of renal tubular epithelial cells (RTEC) and cause CDDP-induced acute kidney injury (CAKI) during cancer treatment, but yet lack of preventive measures and effective treatment. As a new Chinese herbal preparation, Panax notoginseng saponins (PNS) has been found to mitigate CDDP-induced CAKI through elevating the expression of HIF-1α in the rat model, according to the data from our previous works. However, the underlying link between HIF-1α and apoptosis has not been well elucidated. The current study as a follow-up work, was aimed to reveal if PNS improves CAKI through HIF-1α-dependent apoptosis. A stably HIF-1α-knockdown human proximal tubular epithelial cell (HK-2) line was established by transfecting a HIF-1α-siRNA into HK-2 cells. Cell viability, mitochondrial function, cell apoptosis ratio and the expression of apoptosis-associated proteins (Cyt C, Bcl2, Bax, caspases 3) were determined. In order to elucidate the underlying mechanism, the expression of HIF-1α and BNIP3 were assessed. Our results showed that treatment of PNS rescued the cell viability of CDDP-injured HK-2 or HIF-1α-knockdown HK-2 cells, and increased the expression levels of ATP and MMP in HK-2 or HIF-1α-knockdown HK-2 cells which were reduced by CDDP. Moreover, PNS treatment decreased the CDDP or CDDP plus HIF-1α-knockdown-induced elevation of apoptosis and apoptosis-associated protein expressions. These findings demonstrate that PNS reduces CAKI through increasing HIF-1α to inhibit mitochondrial apoptosis pathway. Hence, we suggest PNS as a protective and therapeutic new drug for CDDP treatment of cancers, which might have significant meaning of further research and application potential.


Asunto(s)
Lesión Renal Aguda/prevención & control , Cisplatino/toxicidad , Panax notoginseng/química , Saponinas/farmacología , Lesión Renal Aguda/inducido químicamente , Animales , Antineoplásicos/toxicidad , Apoptosis/efectos de los fármacos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Células Epiteliales/patología , Técnicas de Silenciamiento del Gen , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Túbulos Renales Proximales/citología , Túbulos Renales Proximales/efectos de los fármacos , Túbulos Renales Proximales/patología , Proteínas de la Membrana , Mitocondrias/efectos de los fármacos , Mitocondrias/patología , Proteínas Proto-Oncogénicas , Ratas , Saponinas/aislamiento & purificación
3.
Oxid Med Cell Longev ; 2021: 5511881, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33859775

RESUMEN

Diabetic nephropathy is a microvascular complication induced by diabetes, and methylglyoxal (MGO) is a reactive carbonyl species causing oxidative stress that contributes to the induction of inflammatory response in kidney cells. Cudrania tricuspidata (CT), cultivated in Northeast Asia, has been used as traditional medicine for treating various diseases, including neuritis, liver damage, and cancer. In this study, we determined whether a CT root extract (CTRE) can prevent MGO-induced reactive oxygen species (ROS) production and inflammation and assessed underlying mechanisms using a kidney epithelial cell line, HK-2. We observed that CTRE inhibited MGO-induced ROS production. Additionally, CTRE ameliorated the activation of MGO-induced inflammatory signaling pathways such as p38 mitogen-activated protein kinase (MAPK), extracellular signal-regulated kinase (ERK), and c-JUN N-terminal kinase (JNK). Consistent with these results, expressions of p-nuclear factor-kappa B (NFκB) and inflammatory cytokines, tumor necrosis factor-α, interleukin- (IL-) 1ß, and IL-6, were decreased when compared with MGO-only exposed HK-2 cells. CTRE alleviated the MGO-induced decrease in nuclear factor (erythroid-derived 2)-like 2 (Nrf2) and antioxidant enzyme mRNA expressions. MGO induced the expression of NADPH oxidase 4 (NOX4); CTRE pretreatment inhibited this induction. Further studies revealed that the NOX4 expression was inhibited owing to the suppression of MGO-induced protein kinase C (PKC) activation following CTRE treatment. Collectively, our data suggest that CTRE attenuates MGO-induced inflammation and oxidative stress via inhibition of PKC activation and NOX4 expression, as well as upregulating the Nrf2-antioxidant enzyme pathway in HK-2 cells.


Asunto(s)
Inflamación/prevención & control , Riñón/efectos de los fármacos , Moraceae/química , Estrés Oxidativo/efectos de los fármacos , Extractos Vegetales/farmacología , Piruvaldehído/farmacología , Transducción de Señal/efectos de los fármacos , Línea Celular , Humanos , Inflamación/inducido químicamente , Inflamación/metabolismo , Inflamación/patología , Riñón/metabolismo , Riñón/patología , Túbulos Renales Proximales/efectos de los fármacos , Túbulos Renales Proximales/metabolismo , Túbulos Renales Proximales/patología , NADPH Oxidasa 4/metabolismo , Extractos Vegetales/química , Raíces de Plantas/química , Proteína Quinasa C/metabolismo , Especies Reactivas de Oxígeno/metabolismo
4.
Biol Pharm Bull ; 43(11): 1693-1698, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33132314

RESUMEN

Cisplatin is a widely used chemotherapy for solid tumors; however, its benefits are limited by serious nephrotoxicity, particularly in proximal tubular cells. The present study investigated the renoprotective effect and mechanisms of germacrone, a bioactive terpenoid compound found in Curcuma species on cisplatin-induced toxicity of renal cells. Germacrone (50 and 100 µM) attenuated apoptosis of human renal proximal tubular cells, RPTEC/TERT1 following treatment with 50 µM cisplatin and for 48 h. Co-treating RPTEC/TERT1 cells with cisplatin and germacrone significantly reduced cellular platinum content compared with cisplatin treatment alone. The effect of germacrone on organic cation transporter 2 (OCT2) which is a transporter responsible for cisplatin uptake was determined. Germacrone showed an inhibitory effect on OCT2-mediated methyl-4-phenylpyridinium acetate (3H-MPP+) uptake with IC50 of 15 µM with less effect on OCT1. The germacrone's protective effect on cisplatin-induced cytotoxicity was not observed in cancer cells; cisplatin's anti-cancer activity was preserved. In conclusion, germacrone prevents cisplatin-induced toxicity in renal proximal tubular cells via inhibition OCT2 transport function and reducing cisplatin accumulation. Thus germacrone may be a good candidate agent used for reducing cisplatin-induced nephrotoxicity.


Asunto(s)
Lesión Renal Aguda/prevención & control , Cisplatino/efectos adversos , Túbulos Renales Proximales/efectos de los fármacos , Transportador 2 de Cátion Orgánico/antagonistas & inhibidores , Sesquiterpenos de Germacrano/farmacología , Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/patología , Animales , Células CHO , Cricetulus , Evaluación Preclínica de Medicamentos , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Hep G2 , Humanos , Concentración 50 Inhibidora , Túbulos Renales Proximales/citología , Túbulos Renales Proximales/patología , Factor 1 de Transcripción de Unión a Octámeros/metabolismo , Transportador 2 de Cátion Orgánico/metabolismo , Sesquiterpenos de Germacrano/uso terapéutico
5.
J Am Soc Nephrol ; 31(7): 1522-1537, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32503896

RESUMEN

BACKGROUND: Mutations in the gene that encodes the lysosomal cystine transporter cystinosin cause the lysosomal storage disease cystinosis. Defective cystine transport leads to intralysosomal accumulation and crystallization of cystine. The most severe phenotype, nephropathic cystinosis, manifests during the first months of life, as renal Fanconi syndrome. The cystine-depleting agent cysteamine significantly delays symptoms, but it cannot prevent progression to ESKD and does not treat Fanconi syndrome. This suggests the involvement of pathways in nephropathic cystinosis that are unrelated to lysosomal cystine accumulation. Recent data indicate that one such potential pathway, lysosome-mediated degradation of autophagy cargoes, is compromised in cystinosis. METHODS: To identify drugs that reduce levels of the autophagy-related protein p62/SQSTM1 in cystinotic proximal tubular epithelial cells, we performed a high-throughput screening on the basis of an in-cell ELISA assay. We then tested a promising candidate in cells derived from patients with, and mouse models of, cystinosis, and in preclinical studies in cystinotic zebrafish. RESULTS: Of 46 compounds identified as reducing p62/SQSTM1 levels in cystinotic cells, we selected luteolin on the basis of its efficacy, safety profile, and similarity to genistein, which we previously showed to ameliorate other lysosomal abnormalities of cystinotic cells. Our data show that luteolin improves the autophagy-lysosome degradative pathway, is a powerful antioxidant, and has antiapoptotic properties. Moreover, luteolin stimulates endocytosis and improves the expression of the endocytic receptor megalin. CONCLUSIONS: Our data show that luteolin improves defective pathways of cystinosis and has a good safety profile, and thus has potential as a treatment for nephropathic cystinosis and other renal lysosomal storage diseases.


Asunto(s)
Antioxidantes/farmacología , Cistinosis/tratamiento farmacológico , Evaluación Preclínica de Medicamentos/métodos , Luteolina/farmacología , ARN Mensajero/metabolismo , Sistemas de Transporte de Aminoácidos Neutros/genética , Animales , Antioxidantes/efectos adversos , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Células Cultivadas , Cistinosis/metabolismo , Modelos Animales de Enfermedad , Endocitosis/efectos de los fármacos , Humanos , Túbulos Renales Proximales/patología , Proteína 2 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Luteolina/efectos adversos , Lisosomas/efectos de los fármacos , Ratones , Estrés Oxidativo/efectos de los fármacos , Fenotipo , Proteína Sequestosoma-1/genética , Proteína Sequestosoma-1/metabolismo , Pez Cebra
6.
Biochim Biophys Acta Mol Basis Dis ; 1866(7): 165792, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32251763

RESUMEN

Renal ischemia-reperfusion is a major cause of acute kidney injury, a disease currently without effective treatments. Irisin was initially identified as an important factor produced by muscles to mediate the health benefits of exercise, and recent work has further suggested its protective effect against lung and liver injury. However, the role of Irisin in kidney diseases, including renal ischemia-reperfusion injury (IRI), remains unknown. In the present study, we found that the Irisin precursor, fibronectin type III domain-containing protein 5 (Fndc5), was induced in renal tubules in a mouse model of renal IRI and in cultured mouse renal proximal tubular cells subjected ATP depletion injury. Functionally, silencing Fndc5 in cultured proximal tubular cells increased the sensitivity to ATP depletion-induced apoptosis, whereas both Fndc5 overexpression and supplementation of recombinant Irisin alleviated ATP depletion-induced apoptosis. In vivo, administration of recombinant Irisin dramatically attenuated kidney dysfunction, tissue damage, tubular cell apoptosis, and inflammation during renal IRI in mice. Mechanistically, Irisin suppressed the activation of p53 in renal IRI, a critical factor in tubular cell death. Together, these results indicate that Irisin is induced in renal IRI as a protective mechanism for renal tubular cells, suggesting the therapeutic potential of recombinant Irisin in renal IRI and related kidney diseases.


Asunto(s)
Lesión Renal Aguda/genética , Fibronectinas/genética , Daño por Reperfusión/genética , Proteína p53 Supresora de Tumor/genética , Lesión Renal Aguda/tratamiento farmacológico , Lesión Renal Aguda/patología , Adenosina Trifosfato/metabolismo , Animales , Apoptosis/efectos de los fármacos , Modelos Animales de Enfermedad , Fibronectinas/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Túbulos Renales Proximales/efectos de los fármacos , Túbulos Renales Proximales/patología , Ratones , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología , Daño por Reperfusión/tratamiento farmacológico , Daño por Reperfusión/patología , Proteína p53 Supresora de Tumor/antagonistas & inhibidores
7.
Mater Sci Eng C Mater Biol Appl ; 110: 110732, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32204043

RESUMEN

Astragalus polysaccharide (APS) was modified using the Na2SeO3/HNO3 method to obtain selenized APS (Se-APS) with a selenium content of 1.75 mg/g. The structure and physicochemical properties of APS and Se-APS were investigated through transmission electron microscopy-energy dispersive spectroscopy mapping, fourier transform infrared spectroscopy, nuclear magnetic resonance, nano-zetasizer analysis, atomic force microscopy, and scanning electron microscopy. APS and Se-APS did not exhibit toxic effects on human kidney proximal tubular epithelial (HK-2) cells and were able to remove hydroxyl and DPPH radicals, alleviate the damage caused by calcium oxalate (CaOx) monohydrate (COM) crystals to HK-2 cells, reduce intracellular reactive oxygen species levels, and restore cell viability and morphology. Both APS and Se-APS could inhibit COM growth, induce calcium oxalate dihydrate formation, and increase the absolute zeta potential of the crystals to inhibit crystal aggregation. However, the ability of Se-APS to regulate CaOx crystals and protect the cells from COM-induced damage was better than that of APS. These results suggested that Se-APS might be a candidate drug for the treatment and prevention of kidney stones.


Asunto(s)
Planta del Astrágalo/química , Células Epiteliales , Cálculos Renales , Túbulos Renales Proximales , Polisacáridos , Selenio , Oxalato de Calcio/metabolismo , Línea Celular , Células Epiteliales/metabolismo , Células Epiteliales/patología , Humanos , Cálculos Renales/tratamiento farmacológico , Cálculos Renales/metabolismo , Cálculos Renales/patología , Túbulos Renales Proximales/metabolismo , Túbulos Renales Proximales/patología , Polisacáridos/química , Polisacáridos/farmacología , Selenio/química , Selenio/farmacología
8.
Biomolecules ; 9(12)2019 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-31861234

RESUMEN

Polyacetylenic compounds isolated from Panax species are comprised of non-polar C17 compounds, exhibiting anti-inflammatory, antitumor, and antifungal activities. Panaxynol represents the major component of the essential oils of ginseng. We investigated whether panaxynol isolated from Panax vietnamensis (Vietnamese ginseng, VG) could prevent cisplatin-induced renal damage induced in vitro and in vivo. Cisplatin-induced apoptotic cell death was observed by staining with annexin V conjugated with Alexa Fluor 488, and western blotting evaluated the molecular mechanism. Panaxynol at concentrations above 0.25 µM prevented cisplatin-induced LLC-PK1 porcine renal proximal tubular cell death. LLC-PK1 cells treated with cisplatin demonstrated an increase in apoptotic cell death, whereas pretreatment with 2 and 4 µM panaxynol decreased this effect. Cisplatin demonstrated a marked increase in the phosphorylation of c-Jun N-terminal kinase (JNK), P38, and cleaved caspase-3. However, pretreatment with 2 and 4 µM panaxynol reversed the upregulated phosphorylation of JNK, P38, and the expression of cleaved caspase-3. We confirmed that the protective effect of panaxynol isolated from P. vietnamensis in LLC-PK1 cells was at least partially mediated by reducing the cisplatin-induced apoptotic damage. In the animal study, panaxynol treatment ameliorated body weight loss and blood renal function markers and downregulated the mRNA expression of inflammatory mediators.


Asunto(s)
Lesión Renal Aguda/tratamiento farmacológico , Cisplatino/farmacología , Diinos/farmacología , Alcoholes Grasos/farmacología , Túbulos Renales Proximales/efectos de los fármacos , Panax/química , Sustancias Protectoras/farmacología , Lesión Renal Aguda/inducido químicamente , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Nitrógeno de la Urea Sanguínea , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Creatinina/sangre , Diinos/química , Diinos/aislamiento & purificación , Alcoholes Grasos/química , Alcoholes Grasos/aislamiento & purificación , Túbulos Renales Proximales/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Sustancias Protectoras/química , Sustancias Protectoras/aislamiento & purificación , Porcinos
9.
Int J Mol Sci ; 20(19)2019 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-31547201

RESUMEN

Although the kidneys comprise a critical target of uranium exposure, the dynamics of renal uranium distribution have remained obscure. Uranium is considered to function physiologically in the form of uranyl ions that have high affinity for phosphate groups. The present study applied microbeam-based elemental analysis to precisely determine the distribution of phosphorus and uranium in the kidneys of male Wistar rats exposed to uranium. One day after a single subcutaneous injection of uranyl acetate (2 mg/kg), areas of concentrated phosphorus were scattered in the S3 segments of the proximal tubule of the kidneys, whereas the S3 segments in control rats and in rats given a lower dose of uranium (0.5 mg/kg) contained phosphorus without concentrated phosphorus. Areas with concentrated phosphorus contained uranium 4- to 14-fold more than the mean uranium concentration (126-472 vs. 33.1 ± 4.6 µg/g). The chemical form of uranium in the concentrated phosphorus examined by XAFS was uranium (VI), suggesting that the interaction of uranyl ions with the phosphate groups of biomolecules could be involved in the formation of uranium concentration in the proximal tubules of kidneys in rats exposed to uranium.


Asunto(s)
Túbulos Renales Proximales/metabolismo , Compuestos Organometálicos , Fósforo/metabolismo , Uranio/metabolismo , Animales , Túbulos Renales Proximales/patología , Masculino , Compuestos Organometálicos/farmacocinética , Compuestos Organometálicos/farmacología , Ratas , Ratas Wistar
10.
Life Sci ; 234: 116755, 2019 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-31415769

RESUMEN

AIMS: Vitamin D and its receptor, vitamin D receptor (VDR), have renoprotection effect against diabetic nephropathy (DN). But the exact mechanism has not been fully elucidated. Epoxyeicosatrienoic acids (EETs) are cytochrome P450 (CYP) epoxygenase-derived metabolites of arachidonic acid, protecting against diabetes and DN. Herein, we hypothesized that activation of VDR attenuated high glucose-induced cellular injury in renal tubular epithelial cells partially through up-regulating CYP2J5 expression. MAIN METHODS: Streptozotocin (STZ) was injected to induce diabetic in wild type and Vdr-/- mice. The effects of VDR knockout and an activator of VDR, paricalcitol, on the renal injury were detected. In vitro, a murine kidney proximal tubule epithelial cell line BU.MPT induced by high glucose were treated with or without paricalcitol (30 mM) for 12 h or 24 h. KEY FINDINGS: The expression of CYP2J5 was significantly decreased both in wild type and Vdr-/- diabetic mice induced by STZ. The STZ-induced kidney architecture damage and apoptosis rate in Vdr-/- mice were more severe. In vitro, high glucose treatment strongly reduced the CYP2J5 expression and the synthesis of 14,15-EET in BU.MPT cells. Supplement of 14,15-EET significantly reduced the lactate dehydrogenase (LDH) release induced by high glucose in BU.MPT cells. Furthermore, treatment with paricalcitol attenuated cellular injury and restored the expression of CYP2J5 reduced by high glucose in BU.MPT cells. SIGNIFICANCE: We conclude that activation of VDR attenuates high glucose-induced cellular injury partially dependent on CYP2J5 in murine renal tubule epithelial cells and paricalcitol may represent a potential therapy for DN.


Asunto(s)
Sistema Enzimático del Citocromo P-450/genética , Diabetes Mellitus Experimental/complicaciones , Nefropatías Diabéticas/tratamiento farmacológico , Nefropatías Diabéticas/etiología , Ergocalciferoles/farmacología , Túbulos Renales Proximales/efectos de los fármacos , Receptores de Calcitriol/agonistas , Animales , Línea Celular , Citocromo P-450 CYP2J2 , Sistema Enzimático del Citocromo P-450/metabolismo , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patología , Nefropatías Diabéticas/genética , Nefropatías Diabéticas/patología , Ergocalciferoles/uso terapéutico , Eliminación de Gen , Regulación de la Expresión Génica/efectos de los fármacos , Glucosa/metabolismo , Túbulos Renales Proximales/metabolismo , Túbulos Renales Proximales/patología , Masculino , Ratones , Ratones Noqueados , Receptores de Calcitriol/genética , Receptores de Calcitriol/metabolismo
11.
Physiol Rep ; 7(14): e14172, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31325249

RESUMEN

Treatment modalities for kidney disease caused by long-term exposure to heavy metals, such as cadmium (Cd), are limited. Often, chronic, long-term environmental exposure to heavy metal is not recognized in the early stages; therefore, chelation therapy is not an effective option. Extracellular vesicles (EVs) derived from stem cells have been demonstrated to reduce disease pathology in both acute and chronic kidney disease models. To test the ability of EVs derived from human bone marrow mesenchymal stem cells (hBM-MSCs) to treat Cd damage, we generated a Cd-exposed medaka model. This model develops heavy metal-induced cell damage in various organs and tissues, and shows decreased overall survival. Intravenous injection of highly purified EVs from hBM-MSCs repaired the damage to apical and basolateral membranes and mitochondria of kidney proximal tubules, glomerular podocytes, bone deformation, and improved survival. Our system also serves as a model with which to study age- and sex-dependent cell injuries of organs caused by various agents and diseases. The beneficial effects of EVs on the tissue repair process, as shown in our novel Cd-exposed medaka model, may open new broad avenues for interventional strategies.


Asunto(s)
Intoxicación por Cadmio/terapia , Vesículas Extracelulares/trasplante , Trasplante de Células Madre Mesenquimatosas/métodos , Animales , Células de la Médula Ósea/metabolismo , Intoxicación por Cadmio/metabolismo , Células Cultivadas , Vesículas Extracelulares/metabolismo , Humanos , Túbulos Renales Proximales/metabolismo , Túbulos Renales Proximales/patología , Células Madre Mesenquimatosas/metabolismo , Mitocondrias/metabolismo , Mitocondrias/patología , Oryzias , Podocitos/metabolismo , Podocitos/patología
12.
Clin Nephrol ; 91(3): 180-186, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30614440

RESUMEN

AIMS: To investigate the clinicopathological features and outcomes of adefovir dipivoxil (ADV)-related renal impairment in Chinese patients. MATERIALS AND METHODS: Clinical, pathological, and follow-up data from 15 patients with ADV-related renal impairment were studied. Proximal renal tubular dysfunction (PRTD) was defined by the presence of at least two of the following four abnormalities: hypophosphatemia, hypouricemia, nondiabetic glucosuria, and proteinuria. RESULTS: All patients were treated for 3 - 15 (mean 6.7) years with daily ADV of 10 mg. Renal impairment manifested as PRTD (12, 80%), elevated serum creatinine (12, 80%), and hematuria (2, 13.3%). Mild to moderate tubulointerstitial injury primarily affecting the proximal tubules by light microscopy, and enlarged, dysmorphic mitochondria with loss and disorientation of cristae by electron microscope were identified in all of our cases. Four patients had pathological evidence of IgA nephropathy. The phosphorus, serum uric acid, and creatinine levels were normalized after ADV cessation in 66.7% (8/12) of affected patients, 27.3% (3/11) of affected patients, and 25% (3/12) of affected patients, respectively; proteinuria was eliminated in 7 of 13 affected patients (53.8%); and glucosuria and hematuria both disappeared in all affected patients. These abnormalities had hardly any recovery, and even aggravated with new-onset glucosuria, new-onset hematuria in 3 patients who replaced ADV with tenofovir. CONCLUSION: Nephrotoxicity developed in patients undergoing long-term ADV treatment and was partially reversible after drug cessation. Tubulointerstitial lesions and heteromorphic mitochondria were the predominant pathological changes. Patients with ADV-induced renal impairment should replace ADV with other antiviral agents other than tenofovir.
.


Asunto(s)
Adenina/análogos & derivados , Antivirales/efectos adversos , Organofosfonatos/efectos adversos , Insuficiencia Renal/inducido químicamente , Insuficiencia Renal/patología , Adenina/efectos adversos , Adulto , Creatinina/sangre , Femenino , Glucosuria/inducido químicamente , Hematuria/inducido químicamente , Hepatitis B Crónica/tratamiento farmacológico , Humanos , Hipofosfatemia/inducido químicamente , Túbulos Renales Proximales/patología , Túbulos Renales Proximales/fisiopatología , Masculino , Persona de Mediana Edad , Mitocondrias/patología , Fósforo/sangre , Proteinuria/inducido químicamente , Insuficiencia Renal/fisiopatología , Ácido Úrico/sangre
13.
Free Radic Biol Med ; 131: 376-381, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30578920

RESUMEN

Chronic kidney disease (CKD) is a worldwide public health problem with an estimated prevalence of 8.2%. This study reports glutathione deficiency, excess oxidative stress, and altered vitamin D metabolism in the kidney of mice fed a high-fat diet (HFD). The levels of GCLC and GCLM gene expression were significantly downregulated and the protein carbonylation level, a hallmark of oxidative damage, was significantly increased in the kidney of HFD-fed mice. While the levels of VD-regulatory genes 1-alpha-hydroxylase (CYP27B1), VDR, and RXRα were significantly downregulated in the kidney of mice fed a HFD, those of 24-hydroxylase (CYP24A1) were significantly elevated. In vitro, GSH deficiency per se causes excess oxidative damage (protein carbonylation), and significantly decreases the levels of VD-regulatory genes (CYP27B1, VDR, and RXRα), but increases levels of CYP24A1 in human renal proximal tubule epithelial cells (RPTEC), similar to findings in the kidney of HFD-fed diabetic mice. L-cysteine supplementation restores GSH and prevents oxidative damage in RPTEC. These studies suggest a potential role of GSH precursor in reducing excess oxidative stress and renal injury that commonly accompanies obesity/diabetes.


Asunto(s)
25-Hidroxivitamina D3 1-alfa-Hidroxilasa/genética , Diabetes Mellitus Experimental/enzimología , Glutatión/deficiencia , Receptores de Calcitriol/genética , Insuficiencia Renal Crónica/enzimología , Vitamina D3 24-Hidroxilasa/genética , 25-Hidroxivitamina D3 1-alfa-Hidroxilasa/metabolismo , Animales , Cisteína/farmacología , Diabetes Mellitus Experimental/etiología , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patología , Dieta Alta en Grasa/efectos adversos , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/patología , Regulación de la Expresión Génica , Glutamato-Cisteína Ligasa/genética , Glutamato-Cisteína Ligasa/metabolismo , Humanos , Túbulos Renales Proximales/metabolismo , Túbulos Renales Proximales/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Estrés Oxidativo , Cultivo Primario de Células , Carbonilación Proteica , Receptores de Calcitriol/metabolismo , Insuficiencia Renal Crónica/etiología , Insuficiencia Renal Crónica/genética , Insuficiencia Renal Crónica/patología , Receptor alfa X Retinoide/genética , Receptor alfa X Retinoide/metabolismo , Transducción de Señal , Vitamina D3 24-Hidroxilasa/metabolismo
14.
J Cell Biochem ; 120(3): 3861-3873, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30304552

RESUMEN

BACKGROUND: Smilax glabra Roxb, a traditional Chinese herb, has been widely used in folk medicine. The current study was performed to investigate the protective effect of S. glabra Roxb extract, pure total flavonoids from Smilax glabra Roxb (PTFS), on renal interstitial fibrosis (RIF) and its underlying mechanism. METHODS: First, a surgical model of unilateral ureteral obstruction was established in rats to induce RIF. Then, rats were grouped and treated with PTFS at different concentration. Second, HK-2 cells underwent an epithelial-mesenchymal transition (EMT) by the addition of transforming growth factor-ß1 (TGF-ß1). Additionally, HK-2 cells after inducing for EMT were transfected with microRNA-21 (miR-21) mimic or inhibitor. These HK-2 cells were grouped and treated with PTFS at different concentration. Finally, real-time polymerase chain reaction and Western blot analysis were performed to detect the expression of possible signaling factor involved in RIF in renal tissues or HK-2 cells after PTFS treatment. RESULTS: In vivo and in vitro experiments indicated that PTFS treatment could decrease the expression of α-smooth muscle actin (α-SMA; mesenchymal marker) and increase the expression of E-cadherin (epithelial marker) in both messenger RNA and protein level. Moreover, PTFS also attenuated the expression of TGF-ß1/Smad signaling in both renal tissues and HK-2 cells that underwent EMT. Overexpression or inhibition of miR-21 in HK-2 cells activated or blocked the PI3K/Akt signaling via targeting phosphatase and tension homolog (PTEN), and then promoted or suppressed the progress of TGF-ß1-induced EMT by regulating the expression of α-SMA and E-cadherin. Furthermore, PTFS treatment inhibited TGF-ß1-induced EMT progress by blocking miR-21/PTEN/PI3K/Akt signaling. CONCLUSION: PTFS has strong anti-EMT and antifibrosis effects both in vitro and in vivo. The mechanism underlying these effects may be related to inhibition of TGF-ß1/Smad, and their downstream miR-21/PTEN signaling, leading to blocks of EMT process during RIF.


Asunto(s)
Transición Epitelial-Mesenquimal/efectos de los fármacos , Flavonoides/farmacología , MicroARNs/genética , Fosfohidrolasa PTEN/genética , Sustancias Protectoras/farmacología , Smilax/química , Obstrucción Ureteral/tratamiento farmacológico , Actinas/genética , Actinas/metabolismo , Animales , Antagomirs/genética , Antagomirs/metabolismo , Cadherinas/genética , Cadherinas/metabolismo , Línea Celular Transformada , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/patología , Transición Epitelial-Mesenquimal/genética , Fibrosis/prevención & control , Flavonoides/aislamiento & purificación , Regulación de la Expresión Génica , Humanos , Túbulos Renales Proximales/efectos de los fármacos , Túbulos Renales Proximales/metabolismo , Túbulos Renales Proximales/patología , Masculino , MicroARNs/agonistas , MicroARNs/antagonistas & inhibidores , MicroARNs/metabolismo , Oligorribonucleótidos/genética , Oligorribonucleótidos/metabolismo , Fosfohidrolasa PTEN/metabolismo , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Sustancias Protectoras/aislamiento & purificación , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal , Proteínas Smad/genética , Proteínas Smad/metabolismo , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo , Obstrucción Ureteral/genética , Obstrucción Ureteral/metabolismo , Obstrucción Ureteral/patología
15.
Intern Med ; 57(24): 3603-3610, 2018 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-30101934

RESUMEN

We present a case of Dent disease caused by a novel intronic mutation, 1348-1G>A, of the chloride voltage-gated channel 5 (CLCN5) gene. Cultured proximal tubule cells obtained from the patient showed impaired acidification of the endosome and/or lysosome, indicating that the 1348-1G>A mutation was indeed the cause of Dent disease. Although the prevalence of osteomalacia in Dent disease is low in Japan, several factors-including poor medication adherence-caused severe osteomalacia in the current case. Oral supplementation with calcium and native/active vitamin D therapy, with careful attention to medication adherence, led to the improvement of the patient's bone status.


Asunto(s)
Canales de Cloruro/genética , Enfermedad de Dent/genética , Osteomalacia/genética , Mutación Puntual , Adulto , Calcio de la Dieta/uso terapéutico , Enfermedad de Dent/complicaciones , Enfermedad de Dent/patología , Suplementos Dietéticos , Humanos , Intrones , Japón , Túbulos Renales Proximales/patología , Masculino , Cumplimiento de la Medicación , Osteomalacia/tratamiento farmacológico , Osteomalacia/etiología , Osteomalacia/patología , Vitamina D/uso terapéutico , Vitaminas/uso terapéutico
16.
J Biochem Mol Toxicol ; 32(9): e22189, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29992668

RESUMEN

Acute renal failure is one of the most frequent effects observed after taking medicine. Such situations have been tardily discovered, given that existing methods for assessing toxicity are not predictive. In this light, the present work evaluated the effects of gentamicin, a form of nephrotoxic drug, on HK-2 and HEK-293 cells. By using MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) and flow cytometry, both cells demonstrated that cytotoxicity occurs in a dose-dependent manner through the processes of apoptosis and cell necrosis. Gene expression analysis showed a relative increase of expression for genes related to cell processes and classic biomarkers, such as TP53, CASP3, CASP8, CASP9, ICAM-1, EXOC3, KIM-1, and CST3. A decrease in expression for genes BCL2L1 and EGF was observed. This study, therefore, indicates that, when the methods are used together, gene expression analysis is able to evaluate the nephrotoxic potential of a substance.


Asunto(s)
Antibacterianos/efectos adversos , Apoptosis/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Gentamicinas/efectos adversos , Riñón/efectos de los fármacos , Inhibidores de la Síntesis de la Proteína/efectos adversos , Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/patología , Alternativas al Uso de Animales , Biomarcadores Farmacológicos/metabolismo , Línea Celular Transformada , Supervivencia Celular/efectos de los fármacos , Cistatina C/agonistas , Cistatina C/genética , Cistatina C/metabolismo , Evaluación Preclínica de Medicamentos/métodos , Factor de Crecimiento Epidérmico/antagonistas & inhibidores , Factor de Crecimiento Epidérmico/genética , Factor de Crecimiento Epidérmico/metabolismo , Citometría de Flujo , Perfilación de la Expresión Génica , Receptor Celular 1 del Virus de la Hepatitis A/agonistas , Receptor Celular 1 del Virus de la Hepatitis A/genética , Receptor Celular 1 del Virus de la Hepatitis A/metabolismo , Humanos , Concentración 50 Inhibidora , Interleucina-18/antagonistas & inhibidores , Interleucina-18/genética , Interleucina-18/metabolismo , Riñón/metabolismo , Riñón/patología , Túbulos Renales Proximales/efectos de los fármacos , Túbulos Renales Proximales/metabolismo , Túbulos Renales Proximales/patología , Necrosis
17.
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
18.
BMJ Case Rep ; 20182018 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-29735496

RESUMEN

A 64-year-old woman had fragility fractures which caused her to have gross deformities and confined her to bed. These were initially ascribed to vitamin D deficiency. However, despite correction of the deficiency, she did not improve. A review of previous records already showed glucosuria in the absence of diabetes, but this finding was overlooked. Eight years into the disease, it was realised that the glucosuria despite normal blood sugar could also mean that the patient was losing other substances needed for proper bone formation. Further investigations showed hypophosphataemia, renal phosphate wasting, hypokalaemia, mild metabolic acidosis, alkaline urine pH, hypouricaemia and aminoaciduria, all compatible with a proximal renal tubular defect (Fanconi syndrome). The fragility fractures were due to poor bone mineralisation because of hypophosphataemia induced by the inability of the kidneys to conserve phosphorus.


Asunto(s)
Síndrome de Fanconi/complicaciones , Fracturas Óseas/etiología , Glucosuria/etiología , Hipofosfatemia/etiología , Túbulos Renales Proximales/anomalías , Absorciometría de Fotón/métodos , Diagnóstico Diferencial , Síndrome de Fanconi/tratamiento farmacológico , Síndrome de Fanconi/patología , Síndrome de Fanconi/orina , Femenino , Fracturas Óseas/diagnóstico por imagen , Fracturas Óseas/tratamiento farmacológico , Fracturas Óseas/cirugía , Humanos , Hipopotasemia/etiología , Hipopotasemia/metabolismo , Hipofosfatemia/metabolismo , Túbulos Renales Proximales/metabolismo , Túbulos Renales Proximales/patología , Persona de Mediana Edad , Fósforo/administración & dosificación , Fósforo/uso terapéutico , Resultado del Tratamiento
19.
J Biochem Mol Toxicol ; 32(3): e22038, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29327458

RESUMEN

This study investigates the exposure of lead-induced reactive oxygen species (ROS) generation, DNA damage, and apoptosis and also evaluates the therapeutic intervention using antioxidants in human renal proximal tubular cells (HK-2 cells). Following treatment of HK-2 cells with an increasing concentration of lead nitrate (0-50 µM) for 24 h, the intracellular ROS level increased whereas the GSH level decreased significantly in a dose-dependent manner. Comet assay results revealed that lead nitrate showed the ability to increase the levels of DNA strand breaks in HK-2 cells. Lead exposure also induced apoptosis through caspase-3 activation at 30 µg/mL. Pretreatment with N-acetylcysteine (NAC) and tannic acid showed a significant ameliorating effect on lead-induced ROS, DNA damage, and apoptosis. In conclusion, lead induces ROS, which may exacerbate the DNA damage and apoptosis via caspase-3 activation. Additionally, supplementation of antioxidants such as NAC and tannic acid may be used as salvage therapy for lead-induced DNA damage and apoptosis in an exposed person.


Asunto(s)
Acetilcisteína/farmacología , Apoptosis/efectos de los fármacos , Daño del ADN , Células Epiteliales/metabolismo , Túbulos Renales Proximales/metabolismo , Plomo/toxicidad , Taninos/farmacología , Células Epiteliales/patología , Humanos , Túbulos Renales Proximales/patología
20.
Am J Physiol Renal Physiol ; 314(5): F702-F714, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28515173

RESUMEN

Ferroptosis is an iron-dependent form of regulated nonapoptotic cell death, which contributes to damage in models of acute kidney injury (AKI). Heme oxygenase-1 (HO-1) is a cytoprotective enzyme induced in response to cellular stress, and is protective against AKI because of its antiapoptotic and anti-inflammatory properties. However, the role of HO-1 in regulating ferroptosis is unclear. The purpose of this study was to elucidate the role of HO-1 in regulating ferroptotic cell death in renal proximal tubule cells (PTCs). Immortalized PTCs obtained from HO-1+/+ and HO-1-/- mice were treated with erastin or RSL3, ferroptosis inducers, in the presence or absence of antioxidants, an iron source, or an iron chelator. Cells were assessed for changes in morphology and metabolic activity as an indicator of cell viability. Treatment of HO-1+/+ PTCs with erastin resulted in a time- and dose-dependent increase in HO-1 gene expression and protein levels compared with vehicle-treated controls. HO-1-/- cells showed increased dose-dependent erastin- or RSL3-induced cell death in comparison to HO-1+/+ PTCs. Iron supplementation with ferric ammonium citrate in erastin-treated cells decreased cell viability further in HO-1-/- PTCs compared with HO-1+/+ cells. Cotreatment with ferrostatin-1 (ferroptosis inhibitor), deferoxamine (iron chelator), or N-acetyl-l-cysteine (glutathione replenisher) significantly increased cell viability and attenuated erastin-induced ferroptosis in both HO-1+/+ and HO-1-/- PTCs. These results demonstrate an important antiferroptotic role of HO-1 in renal epithelial cells.


Asunto(s)
Lesión Renal Aguda/enzimología , Hemo-Oxigenasa 1/metabolismo , Túbulos Renales Proximales/enzimología , Proteínas de la Membrana/metabolismo , Acetilcisteína/farmacología , Lesión Renal Aguda/genética , Lesión Renal Aguda/patología , Lesión Renal Aguda/prevención & control , Animales , Antioxidantes/farmacología , Carbolinas/toxicidad , Muerte Celular , Línea Celular , Ciclohexilaminas/farmacología , Deferoxamina/farmacología , Relación Dosis-Respuesta a Droga , Compuestos Férricos/toxicidad , Glutatión/metabolismo , Hemo-Oxigenasa 1/deficiencia , Hemo-Oxigenasa 1/genética , Quelantes del Hierro/farmacología , Túbulos Renales Proximales/efectos de los fármacos , Túbulos Renales Proximales/patología , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Ratones Noqueados , Fenilendiaminas/farmacología , Piperazinas/toxicidad , Compuestos de Amonio Cuaternario/toxicidad , Transducción de Señal , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA