Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Toxicol Mech Methods ; 34(1): 20-31, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37621060

RESUMEN

Nephrotoxicity induced by aristolochic acid I (AAI) is related to redox stress and apoptosis. Apurinic/apyrimidine endonuclease 1 (APE1) has antioxidant and anti-apoptotic effects. This study investigated the potential role of APE1 in AAI-induced nephrotoxicity. Renal injury was successfully induced in C57BL/6J mice by intraperitoneal injection of AAI every other day for 28 days. Expressions of APE1, nuclear factor erythroid 2-related factor 2 (Nrf2), and heme oxygenase 1 (HO-1) in renal tissues of the model mice was inhibited, accompanied by oxidative damage and apoptosis. Similar results were obtained in vitro in human proximal tubular (HK-2) cells damaged by AAI. In the presence of a low concentration of the APE1 inhibitor E3330, expression of Nrf2 and HO-1 proteins in HK-2 cells was decreased and AAI-induced apoptosis was aggravated. Overexpression of APE1 in HK-2 cells promoted the expression of Nrf2 and HO-1, and alleviated apoptosis and renal injury induced by AAI. The collective findings demonstrate that AAI can inhibit the induction of oxidative stress and apoptosis by the APE1/Nrf2/HO-1 axis, leading to AAI renal injury. Targeting APE1 may be an effective therapeutic strategy to treat AA nephrotoxicity.


Asunto(s)
Ácidos Aristolóquicos , Factor 2 Relacionado con NF-E2 , Ratones , Humanos , Animales , Factor 2 Relacionado con NF-E2/metabolismo , Hemo-Oxigenasa 1/genética , Hemo-Oxigenasa 1/metabolismo , Ratones Endogámicos C57BL , Estrés Oxidativo , Apoptosis , Ácidos Aristolóquicos/toxicidad
2.
Artículo en Inglés | MEDLINE | ID: mdl-38777778

RESUMEN

BACKGROUND: Aristolochic acid nephropathy (AAN) is a rapidly progressive interstitial nephropathy caused by Aristolochic acid (AA). AAN is associated with the development of nephropathy and urothelial carcinoma. It is estimated that more than 100 million people worldwide are at risk of developing AAN. However, the underlying mechanisms driving renal deterioration in AAN remain poorly understood, and the treatment options are limited. METHODS: We obtained GSE27168 and GSE136276 series matrix data from the Gene Expression Omnibus (GEO) related to AAN. Using the R Studio environment, we applied the limma package and WGCNA package to identify co-differently expressed genes (co-DEGs). By GO/KEGG/GSVA analysis, we revealed common biological pathways. Subsequently, co-DEGs were subjected to the String database to construct a protein-protein interaction (PPI) network. The MCC algorithms implemented in the Cytohubba plugin were employed to identify hub genes. The hub genes were cross-referenced with the transcription factor (TF) database to identify hub TFs. Immune infiltration analysis was performed to identify key immune cell groups by utilizing CIBERSORT. The expressions of AAN-associated hub TFs were verified in vivo and in vitro. Finally, siRNA intervention was performed on the two TFs to verify their regulatory effect in AAN. RESULTS: Our analysis identified 88 co-DEGs through the "limma" and "WGCNA" R packages. A PPI network comprising 53 nodes and 34 edges was constructed with a confidence level >0.4. ATF3 and c-JUN were identified as hub TFs potentially linked to AAN. Additionally, expressions of ATF3 and c-JUN positively correlated with monocytes, basophils, and vessels, and negatively correlated with eosinophils and endothelial cells. We observed a significant increase in protein and mRNA levels of these two hub TFs. Furthermore, it was found that siRNA intervention targeting ATF3, but not c-JUN, alleviated cell damage induced by AA. The knockdown of ATF3 protects against oxidative stress and inflammation in the AAN cell model. CONCLUSION: This study provides novel insights into the role of ATF3 in AAN. The comprehensive analysis sheds light on the molecular mechanisms and identifies potential biomarkers and drug targets for AAN treatment.


Asunto(s)
Ácidos Aristolóquicos , Enfermedades Renales , Factores de Transcripción , Ácidos Aristolóquicos/toxicidad , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Enfermedades Renales/inducido químicamente , Enfermedades Renales/genética , Animales , Ratones , Humanos , Factor de Transcripción Activador 3/genética , Factor de Transcripción Activador 3/metabolismo , Mapas de Interacción de Proteínas
3.
Molecules ; 29(1)2023 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-38202664

RESUMEN

Aristolochic acids (AAs) are a toxic substance present in certain natural plants. Direct human exposure to these plants containing AAs leads to a severe and irreversible condition known as aristolochic acid nephropathy (AAN). Additionally, AAs accumulation in the food chain through environmental mediators can trigger Balkan endemic nephropathy (BEN), an environmental variant of AAN. This paper presents a concise overview of the oncogenic pathways associated with AAs and explores the various routes of environmental exposure to AAs. The detection and removal of AAs in natural plants, drugs, and environmental and biological samples were classified and summarized, and the advantages and disadvantages of the various methods were analyzed. It is hoped that this review can provide effective insights into the detection and removal of AAs in the future.


Asunto(s)
Ácidos Aristolóquicos , Nefropatía de los Balcanes , Plantas Medicinales , Humanos , Ácidos Aristolóquicos/toxicidad , Nefropatía de los Balcanes/inducido químicamente , Exposición a Riesgos Ambientales
4.
Pharm Biol ; 61(1): 23-29, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36524761

RESUMEN

CONTEXT: Salvianolic acid B (SAB) can alleviate renal fibrosis and improve the renal function. OBJECTIVE: To investigate the effect of SAB on renal tubulointerstitial fibrosis and explore its underlying mechanisms. MATERIALS AND METHODS: Male C57 mice were subjected to unilateral ureteric obstruction (UUO) and aristolochic acid nephropathy (AAN) for renal fibrosis indication. Vehicle or SAB (10 mg/kg/d, i.p.) were given consecutively for 2 weeks in UUO mice while 4 weeks in AAN mice. The serum creatinine (Scr) and blood urine nitrogen (BUN) were measured. Masson's trichrome staining and the fibrotic markers (FN and α-SMA) were used to evaluate renal fibrosis. NRK-49F cells exposed to 2.5 ng/mL TGF-ß were treated with SAB in the presence or absence of 20 µM 3-DZNep, an inhibitor of EZH2. The protein expression of EZH2, H3k27me3 and PTEN/Akt signaling pathway in renal tissue and NRK-49F cells were measured by Western blots. RESULTS: SAB significantly improved the levels of Scr by 24.3% and BUN by 35.7% in AAN mice. SAB reduced renal interstitial collagen deposition by 34.7% in UUO mice and 72.8% in AAN mice. Both in vivo and in vitro studies demonstrated that SAB suppressed the expression of FN and α-SMA, increased PTEN and decreased the phosphorylation of Akt, which were correlated with the down-regulation of EZH2 and H3k27me3. The inhibition of EZH2 attenuated the anti-fibrotic effects of SAB in NRK-49Fs. CONCLUSION: SAB might have therapeutic potential on renal fibrosis of CKD through inhibiting EZH2, which encourages further clinical trials.


Asunto(s)
Enfermedades Renales , Animales , Masculino , Ratones , Fibrosis/tratamiento farmacológico , Fibrosis/patología , Histonas/metabolismo , Riñón/efectos de los fármacos , Riñón/patología , Enfermedades Renales/tratamiento farmacológico , Enfermedades Renales/prevención & control , Enfermedades Renales/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Obstrucción Ureteral/complicaciones , Obstrucción Ureteral/tratamiento farmacológico , Benzofuranos/farmacología , Benzofuranos/uso terapéutico , Depsidos/farmacología , Depsidos/uso terapéutico , Proteína Potenciadora del Homólogo Zeste 2/antagonistas & inhibidores , Proteína Potenciadora del Homólogo Zeste 2/efectos de los fármacos , Fosfohidrolasa PTEN/metabolismo
5.
Cell Biol Toxicol ; 38(4): 629-648, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-34677723

RESUMEN

Aristolochic acid nephropathy (AAN) is a progressive kidney disease caused by some herbal medicines, but treatment remains ineffective. We previously found that leucine-rich α-2-glycoprotein 1 (LRG1), which regulates cellular processes, plays an important role in a kidney injury model. However, the underlying mechanism by which LRG1 regulates AAN is still unknown. In this study, we established an AAN model in vivo, a coculture system of macrophages and TECs, and a macrophage/TEC conditioned media culture model in vitro. We found that macrophage infiltration promoted injury, oxidative stress, and apoptosis in TECs. Furthermore, the role of macrophages in AAN was dependent on macrophage-derived extracellular vesicles (EVs). Importantly, we found that macrophage-derived, LRG1-enriched EVs induced TEC injury and apoptosis via a TGFßR1-dependent process. This study may help design a better therapeutic strategy to treat AAN patients.


Asunto(s)
Vesículas Extracelulares , Enfermedades Renales , Animales , Ácidos Aristolóquicos , Modelos Animales de Enfermedad , Glicoproteínas , Humanos , Enfermedades Renales/inducido químicamente , Macrófagos
6.
FASEB J ; 34(9): 12599-12614, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32706145

RESUMEN

Renal tubulointerstitial fibrosis (TIF) is a common pathological feature of aristolochic acid (AA) nephropathy (AAN). G2/M arrest of proximal tubular cells (PTCs) is implicated in renal fibrosis of AAN, but the upstream regulatory molecule remains unknown. Hypoxia inducible factor-1α (HIF-1α) promotes renal fibrosis in kidney disease, but the role of HIF-1α in AAN is unclear. Evidence shows that HIF-1α and p21, a known inducer of cellular G2/M arrest, are closely related to each other. To investigate the role of HIF-1α in renal fibrosis of AAN and its effects on p21 expression and PTCs G2/M arrest, mice with HIF-1α gene knockout PTCs (PT-HIF-1α-KO) were generated, and AAN was induced by AA. In vitro tests were conducted on the human PTCs line HK-2 and primary mouse PTCs. HIF-1α and p21 expression, fibrogenesis, and G2/M arrest of PTCs were determined. Results showed that HIF-1α was upregulated in the kidneys of wild-type (WT) AAN mice, accompanied by p21 upregulation, PTCs G2/M arrest and renal fibrosis, and these alterations were reversed in PT-HIF-1α-KO AAN mice. Similar results were observed in HK-2 cells and were further confirmed in primary PTCs from PT-HIF-1α-KO and WT mice. Inhibiting p21 in HK-2 cells and primary PTCs did not change the expression of HIF-1α, but G2/M arrest and fibrogenesis were reduced. These data indicate that HIF-1α plays a key role in renal fibrosis in AAN by inducing PTCs G2/M arrest modulated through p21. HIF-1α may serve as a potential therapeutic target for AAN.


Asunto(s)
Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Células Epiteliales/citología , Subunidad alfa del Factor 1 Inducible por Hipoxia/fisiología , Túbulos Renales Proximales , Nefritis Intersticial/metabolismo , Animales , Ácidos Aristolóquicos , Línea Celular , Fibrosis/inducido químicamente , Puntos de Control de la Fase G2 del Ciclo Celular , Humanos , Túbulos Renales Proximales/citología , Túbulos Renales Proximales/patología , Ratones , Ratones Noqueados
7.
Int J Mol Sci ; 22(19)2021 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-34638820

RESUMEN

The plant extract aristolochic acid (AA), containing aristolochic acids I (AAI) and II (AAII) as major components, causes aristolochic acid nephropathy (AAN) and Balkan endemic nephropathy (BEN), unique renal diseases associated with upper urothelial cancer. Recently (Chemical Research in Toxicology 33(11), 2804-2818, 2020), we showed that the in vivo metabolism of AAI and AAII in Wistar rats is influenced by their co-exposure (i.e., AAI/AAII mixture). Using the same rat model, we investigated how exposure to the AAI/AAII mixture can influence AAI and AAII DNA adduct formation (i.e., AA-mediated genotoxicity). Using 32P-postlabelling, we found that AA-DNA adduct formation was increased in the livers and kidneys of rats treated with AAI/AAII mixture compared to rats treated with AAI or AAII alone. Measuring the activity of enzymes involved in AA metabolism, we showed that enhanced AA-DNA adduct formation might be caused partially by both decreased AAI detoxification as a result of hepatic CYP2C11 inhibition during treatment with AAI/AAII mixture and by hepatic or renal NQO1 induction, the key enzyme predominantly activating AA to DNA adducts. Moreover, our results indicate that AAII might act as an inhibitor of AAI detoxification in vivo. Consequently, higher amounts of AAI might remain in liver and kidney tissues, which can be reductively activated, resulting in enhanced AAI DNA adduct formation. Collectively, these results indicate that AAII present in the plant extract AA enhances the genotoxic properties of AAI (i.e., AAI DNA adduct formation). As patients suffering from AAN and BEN are always exposed to the plant extract (i.e., AAI/AAII mixture), our findings are crucial to better understanding host factors critical for AAN- and BEN-associated urothelial malignancy.


Asunto(s)
Ácidos Aristolóquicos/toxicidad , Carcinogénesis , Carcinógenos/toxicidad , Aductos de ADN/metabolismo , ADN de Neoplasias/metabolismo , Animales , Carcinogénesis/inducido químicamente , Carcinogénesis/metabolismo , Masculino , Ratas , Ratas Wistar
8.
Biomed Chromatogr ; 32(3)2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29088495

RESUMEN

Aristolochic acid (AA) causes interstitial renal fibrosis, called aristolochic acid nephropathy (AAN). There is no specific indicator for diagnosing AAN, so this study aimed to investigate the biomarkers for AAN using a proteomics method. The C3H/He female mice were given ad libitum AA-distilled water (0.5 mg/kg/day) and distilled water for 56 days in the AA and normal groups, respectively. The AA-induced proteins in the kidney were investigated using a proteomics study, including fluorogenic derivatization with 7-chloro-N-[2-(dimethylamino)ethyl]-2,1,3-benzoxadiazole-4-sulfonamide, followed by high-performance liquid chromatography analysis and liquid chromatography tandem mass spectrometry with a MASCOT database searching system. There were two altered proteins, thrombospondin type 1 (TSP1) and G protein-coupled receptor 87 (GPR87), in the kidney of AA-group mice on day 56. GPR87, a tumorigenesis-related protein, is reported for the first time in the current study. The renal interstitial fibrosis was certainly induced in the AA-group mice under histological examination. Based on the results of histological examination and the proteomics study, this model might be applied to AAN studies in the future. TSP1 might be a novel biomarker for AAN, and the further role of GPR87 leading to AA-induced tumorigenesis should be researched in future studies.


Asunto(s)
Ácidos Aristolóquicos/efectos adversos , Enfermedades Renales/inducido químicamente , Enfermedades Renales/metabolismo , Proteoma/análisis , Proteoma/efectos de los fármacos , Animales , Cromatografía Líquida de Alta Presión/métodos , Femenino , Riñón/química , Riñón/efectos de los fármacos , Riñón/metabolismo , Ratones , Ratones Endogámicos C3H , Proteínas , Proteómica , Receptores del Ácido Lisofosfatídico/análisis , Espectrometría de Masas en Tándem/métodos , Trombospondina 1/orina
9.
Apoptosis ; 22(6): 769-776, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28386751

RESUMEN

Aristolochic acid nephropathy remains a leading cause of chronic kidney disease (CKD), however few treatment strategies exist. Emerging evidence has shown that H2 relaxin (RLX) possesses powerful antifibrosis and anti-apoptotic properties, therefore we aimed to investigate whether H2 relaxin can be employed to reduce AA-induced cell apoptosis. Human proximal tubular epithelial (HK-2) cells exposed to AA-I were treated with or without administration of H2 RLX. Cell viability was examined using the WST-8 assay. Apoptotic morphologic alterations were observed using the Hoechst 33342 staining method. Apoptosis was detected using flow cytometry. The expression of caspase 3, caspase 8, caspase 9, ERK1/2, Bax, Bcl-2, and Akt proteins was determined by Western blot. Co-treatment with RLX reversed the increased apoptosis observed in the AA-I only treated group. RLX restored expression of phosphorylated Akt which found to be decreased in the AA-I only treated cells. RLX co-treatment led to a decrease in the Bax/Bcl-2 ratio as well as the cleaved form of caspase-3 compared to the AA-I only treated cells. This anti-apoptotic effect of RLX was attenuated by co-administration of the Akt inhibitor LY294002. The present study demonstrated H2 RLX can decrease AA-I induced apoptosis through activation of the PI3K/Akt signaling pathway.


Asunto(s)
Apoptosis/efectos de los fármacos , Ácidos Aristolóquicos/toxicidad , Células Epiteliales/metabolismo , Células Epiteliales/patología , Túbulos Renales/patología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Relaxina/farmacología , Caspasas/metabolismo , Línea Celular , Forma de la Célula/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Fosforilación/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Proteína X Asociada a bcl-2/metabolismo
10.
Arch Toxicol ; 91(4): 1957-1975, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27557898

RESUMEN

Exposure to aristolochic acid (AA) causes aristolochic acid nephropathy (AAN) and Balkan endemic nephropathy (BEN). Conflicting results have been found for the role of human sulfotransferase 1A1 (SULT1A1) contributing to the metabolic activation of aristolochic acid I (AAI) in vitro. We evaluated the role of human SULT1A1 in AA bioactivation in vivo after treatment of transgenic mice carrying a functional human SULT1A1-SULT1A2 gene cluster (i.e. hSULT1A1/2 mice) and Sult1a1(-/-) mice with AAI and aristolochic acid II (AAII). Both compounds formed characteristic DNA adducts in the intact mouse and in cytosolic incubations in vitro. However, we did not find differences in AAI-/AAII-DNA adduct levels between hSULT1A1/2 and wild-type (WT) mice in all tissues analysed including kidney and liver despite strong enhancement of sulfotransferase activity in both kidney and liver of hSULT1A1/2 mice relative to WT, kidney and liver being major organs involved in AA metabolism. In contrast, DNA adduct formation was strongly increased in hSULT1A1/2 mice compared to WT after treatment with 3-nitrobenzanthrone (3-NBA), another carcinogenic aromatic nitro compound where human SULT1A1/2 is known to contribute to genotoxicity. We found no differences in AAI-/AAII-DNA adduct formation in Sult1a1(-/-) and WT mice in vivo. Using renal and hepatic cytosolic fractions of hSULT1A1/2, Sult1a1(-/-) and WT mice, we investigated AAI-DNA adduct formation in vitro but failed to find a contribution of human SULT1A1/2 or murine Sult1a1 to AAI bioactivation. Our results indicate that sulfo-conjugation catalysed by human SULT1A1 does not play a role in the activation pathways of AAI and AAII in vivo, but is important in 3-NBA bioactivation.


Asunto(s)
Ácidos Aristolóquicos/toxicidad , Arilsulfotransferasa/genética , Benzo(a)Antracenos/toxicidad , Aductos de ADN/efectos de los fármacos , Animales , Carcinógenos/toxicidad , Citosol/efectos de los fármacos , Citosol/metabolismo , Aductos de ADN/genética , Humanos , Riñón/efectos de los fármacos , Riñón/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Familia de Multigenes
11.
Arch Toxicol ; 90(11): 2595-2615, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27538407

RESUMEN

Balkan endemic nephropathy (BEN) is a unique, chronic renal disease frequently associated with upper urothelial cancer (UUC). It only affects residents of specific farming villages located along tributaries of the Danube River in Bosnia-Herzegovina, Croatia, Macedonia, Serbia, Bulgaria, and Romania where it is estimated that ~100,000 individuals are at risk of BEN, while ~25,000 have the disease. This review summarises current findings on the aetiology of BEN. Over the last 50 years, several hypotheses on the cause of BEN have been formulated, including mycotoxins, heavy metals, viruses, and trace-element insufficiencies. However, recent molecular epidemiological studies provide a strong case that chronic dietary exposure to aristolochic acid (AA) a principal component of Aristolochia clematitis which grows as a weed in the wheat fields of the endemic regions is the cause of BEN and associated UUC. One of the still enigmatic features of BEN that need to be resolved is why the prevalence of BEN is only 3-7 %. This suggests that individual genetic susceptibilities to AA exist in humans. In fact dietary ingestion of AA along with individual genetic susceptibility provides a scenario that plausibly can explain all the peculiarities of BEN such as geographical distribution and high risk of urothelial cancer. For the countries harbouring BEN implementing public health measures to avoid AA exposure is of the utmost importance because this seems to be the best way to eradicate this once mysterious disease to which the residents of BEN villages have been completely and utterly at mercy for so long.


Asunto(s)
Ácidos Aristolóquicos/toxicidad , Nefropatía de los Balcanes/inducido químicamente , Carcinógenos Ambientales/toxicidad , Enfermedades Endémicas , Medicina Basada en la Evidencia , Contaminación de Alimentos , Riñón/efectos de los fármacos , Animales , Aristolochia/química , Aristolochia/crecimiento & desarrollo , Aristolochia/toxicidad , Ácidos Aristolóquicos/análisis , Nefropatía de los Balcanes/epidemiología , Nefropatía de los Balcanes/fisiopatología , Nefropatía de los Balcanes/prevención & control , Carcinógenos Ambientales/análisis , Factores de Confusión Epidemiológicos , Productos Agrícolas/crecimiento & desarrollo , Dieta/efectos adversos , Resistencia a Medicamentos , Europa Oriental/epidemiología , Harina/efectos adversos , Harina/análisis , Contaminación de Alimentos/prevención & control , Humanos , Riñón/fisiopatología , Malezas/química , Malezas/crecimiento & desarrollo , Malezas/toxicidad , Prevalencia , Riesgo , Semillas/crecimiento & desarrollo , Triticum/crecimiento & desarrollo , Neoplasias Urológicas/inducido químicamente , Neoplasias Urológicas/epidemiología , Neoplasias Urológicas/fisiopatología , Neoplasias Urológicas/prevención & control
12.
Int J Mol Sci ; 17(2): 213, 2016 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-26861298

RESUMEN

Aristolochic acid I (AAI) is a plant drug found in Aristolochia species that causes aristolochic acid nephropathy, Balkan endemic nephropathy and their associated urothelial malignancies. AAI is activated via nitroreduction producing genotoxic N-hydroxyaristolactam, which forms DNA adducts. The major enzymes responsible for the reductive bioactivation of AAI are NAD(P)H: quinone oxidoreductase and cytochromes P450 (CYP) 1A1 and 1A2. Using site-directed mutagenesis we investigated the possible mechanisms of CYP1A1/1A2/1B1-catalyzed AAI nitroreduction. Molecular modelling predicted that the hydroxyl groups of serine122/threonine124 (Ser122/Thr124) amino acids in the CYP1A1/1A2-AAI binary complexes located near to the nitro group of AAI, are mechanistically important as they provide the proton required for the stepwise reduction reaction. In contrast, the closely related CYP1B1 with no hydroxyl group containing residues in its active site is ineffective in catalyzing AAI nitroreduction. In order to construct an experimental model, mutant forms of CYP1A1 and 1A2 were prepared, where Ser122 and Thr124 were replaced by Ala (CYP1A1-S122A) and Val (CYP1A2-T124V), respectively. Similarly, a CYP1B1 mutant was prepared in which Ala133 was replaced by Ser (CYP1B1-A133S). Site-directed mutagenesis was performed using a quickchange approach. Wild and mutated forms of these enzymes were heterologously expressed in Escherichia coli and isolated enzymes characterized using UV-vis spectroscopy to verify correct protein folding. Their catalytic activity was confirmed with CYP1A1, 1A2 and 1B1 marker substrates. Using (32)P-postlabelling we determined the efficiency of wild-type and mutant forms of CYP1A1, 1A2, and 1B1 reconstituted with NADPH:CYP oxidoreductase to bioactivate AAI to reactive intermediates forming covalent DNA adducts. The S122A and T124V mutations in CYP1A1 and 1A2, respectively, abolished the efficiency of CYP1A1 and 1A2 enzymes to generate AAI-DNA adducts. In contrast, the formation of AAI-DNA adducts was catalyzed by CYP1B1 with the A133S mutation. Our experimental model confirms the importance of the hydroxyl group possessing amino acids in the active center of CYP1A1 and 1A2 for AAI nitroreduction.


Asunto(s)
Ácidos Aristolóquicos/metabolismo , Hidrocarburo de Aril Hidroxilasas/genética , Hidrocarburo de Aril Hidroxilasas/metabolismo , Dominio Catalítico/genética , Mutación , Catálisis , Citocromo P-450 CYP1A1 , Citocromo P-450 CYP1A2 , Citocromo P-450 CYP1B1 , Aductos de ADN/metabolismo , Humanos , Mutagénesis Sitio-Dirigida , Oxidación-Reducción , Proteínas Recombinantes , Especificidad por Sustrato
13.
Molecules ; 21(10)2016 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-27763560

RESUMEN

Aristolochic acid nephropathy (AAN) is a progressive kidney disease caused by a Chinese herb containing aristolochic acid. Excessive death of renal tubular epithelial cells (RTECs) characterized the acute phase of AAN. Therapies for acute AAN were limited, such as steroids and angiotensin-receptor blockers (ARBs)/angiotensin-converting enzyme inhibitors (ACEIs). It was interesting that, in acute AAN, female patients showed relative slower progression to renal failure than males. In a previous study, female hormone 17ß-estradiol (E2) was found to attenuate renal ischemia-reperfusion injury. Thus, the aim of this study was to investigate the potential protective role of E2 in acute AAN. Compared with male C57BL/6 mice of acute AAN, lower serum creatinine (SCr) and less renal injury, together with RTEC apoptosis in females, were found. Treatment with E2 in male AAN mice reduced SCr levels and attenuated renal tubular injury and RTEC apoptosis. In the mice kidney tissue and human renal proximal tubule cells (HK-2 cells), E2 both attenuated AA-induced cell apoptosis and downregulated the expression of phosphor-p53 (Ser15), p53, and cleaved-caspase-3. This study highlights that E2 exhibited protective effects on the renal injury of acute AAN in male mice by reducing RTEC apoptosis, which might be related to inhibiting the p53 signaling pathway.


Asunto(s)
Ácidos Aristolóquicos/toxicidad , Células Epiteliales/efectos de los fármacos , Estradiol/administración & dosificación , Enfermedades Renales/prevención & control , Túbulos Renales/citología , Animales , Apoptosis/efectos de los fármacos , Caspasa 3/metabolismo , Línea Celular , Creatinina/sangre , Modelos Animales de Enfermedad , Estradiol/farmacología , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Enfermedades Renales/inducido químicamente , Enfermedades Renales/metabolismo , Túbulos Renales/efectos de los fármacos , Masculino , Ratones , Fosforilación/efectos de los fármacos , Proteína p53 Supresora de Tumor/metabolismo
14.
Nephrol Dial Transplant ; 30(11): 1893-6, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25964302

RESUMEN

BACKGROUND: Aristolochia clematitis (AC), a herbaceous plant that belongs to the family of Aristolochiaceae, is today considered as being responsible for Balkan endemic nephropathy (BEN). Very scarce information is available in the medical literature about the presence of AC outside Balkan area. This article reports on the finding of AC in Northwest Italy and the results of a questionnaire delivered to locals on their knowledge about AC. METHODS: AC was found in an uncultivated piece of land of a hilly area of Northwest Italy. It was identified by matching it with images available in the literature and Internet. The questionnaire, which was delivered with a set of 12 photographs and a bunch of true AC, contained 15 questions aimed at collecting information on the knowledge of the respondents about the existence, name, distribution and possible uses of AC. RESULTS: A total of 23 locals, mostly farmers, were interviewed. Among them, 22 (95.6%) had already seen AC, mostly in uncultivated areas; 4 (18%) had a name for it; 21 (95.4%) considered it as a weed and denied any personal use of it; 18 (81.8%) stated that breeding animals disliked AC and no one was aware that AC might damage kidneys. CONCLUSIONS: This study demonstrates that AC can be found outside the Balkan region and that people know it but today do not make any use of it. Other studies carried out by nephrologists in other geographic areas could expand our knowledge about AC outside the basin of BEN.


Asunto(s)
Aristolochia/fisiología , Ácidos Aristolóquicos/efectos adversos , Nefropatía de los Balcanes/inducido químicamente , Nefropatía de los Balcanes/epidemiología , Etnobotánica , Adulto , Anciano , Anciano de 80 o más Años , Aristolochia/química , Femenino , Humanos , Italia/epidemiología , Masculino , Persona de Mediana Edad , Nefrología , Médicos , Encuestas y Cuestionarios
15.
Transpl Int ; 28(2): 199-205, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25377421

RESUMEN

Intravesical instillation of bacillus Calmette-Guerin (BCG) is the treatment of choice for non-muscle-invasive bladder cancer (NMIBC) of high grade and/or carcinoma in situ. This study evaluated the feasibility, efficacy, and tolerance of BCG instillations in eight kidney recipients for end-stage aristolochic acid nephropathy (AAN), a condition at high risk of urothelial carcinoma, and diagnosed for NMIBC. Five of them had relapsed after mitomycin C treatment. Tolerance to BCG was evaluated clinically and regular follow-up with fluorescence cystoscopy was performed along with renal graft function monitoring. Immunosuppression doses were adjusted and prophylactic anti-tuberculous treatment given to reduce risks of graft rejection and infection. After a mean follow-up period of 50 months, seven of the eight patients are free of relapse and kidney graft function remained unchanged. Tolerance was good, except for one episode of fever and one early discontinuation because of subjective discomfort. No systemic tuberculous infection was observed. This is the first clinical observation of successful BCG therapy for NMIBC in patients given transplant for end-stage AAN. Under standardized conditions, immunotherapy based on intravesical BCG is feasible, effective, and well tolerated in renal transplantation.


Asunto(s)
Ácidos Aristolóquicos/toxicidad , Vacuna BCG/uso terapéutico , Fallo Renal Crónico/cirugía , Trasplante de Riñón , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Administración Intravesical , Adulto , Anciano , Femenino , Estudios de Seguimiento , Humanos , Fallo Renal Crónico/inducido químicamente , Persona de Mediana Edad , Invasividad Neoplásica , Neoplasias de la Vejiga Urinaria/patología
16.
Arch Toxicol ; 89(11): 2141-58, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25209566

RESUMEN

Exposure to the plant nephrotoxin and carcinogen aristolochic acid (AA) leads to the development of AA nephropathy, Balkan endemic nephropathy (BEN) and upper urothelial carcinoma (UUC) in humans. Beside AA, exposure to ochratoxin A (OTA) was linked to BEN. Although OTA was rejected as a factor for BEN/UUC, there is still no information whether the development of AA-induced BEN/UUC is influenced by OTA exposure. Therefore, we studied the influence of OTA on the genotoxicity of AA (AA-DNA adduct formation) in vivo. AA-DNA adducts were formed in liver and kidney of rats treated with AA or AA combined with OTA, but no OTA-related DNA adducts were detectable in rats treated with OTA alone or OTA combined with AA. Compared to rats treated with AA alone, AA-DNA adduct levels were 5.4- and 1.6-fold higher in liver and kidney, respectively, of rats treated with AA combined with OTA. Although AA and OTA induced NAD(P)H: quinone oxidoreductase (NQO1) activating AA to DNA adducts, their combined treatment did not lead to either higher NQO1 enzyme activity or higher AA-DNA adduct levels in ex vivo incubations. Oxidation of AA I (8-methoxy-6-nitrophenanthro[3,4-d]-1,3-dioxole-5-carboxylic acid) to its detoxification metabolite, 8-hydroxyaristolochic acid, was lower in microsomes from rats treated with AA and OTA, and this was paralleled by lower activities of cytochromes P450 1A1/2 and/or 2C11 in these microsomes. Our results indicate that a decrease in AA detoxification after combined exposure to AA and OTA leads to an increase in AA-DNA adduct formation in liver and kidney of rats.


Asunto(s)
Ácidos Aristolóquicos/toxicidad , Carcinógenos/toxicidad , Aductos de ADN/efectos de los fármacos , Ocratoxinas/farmacología , Animales , Ácidos Aristolóquicos/metabolismo , Carcinógenos/metabolismo , Inactivación Metabólica/efectos de los fármacos , Riñón/efectos de los fármacos , Riñón/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , NAD(P)H Deshidrogenasa (Quinona)/metabolismo , Oxidación-Reducción , Ratas , Ratas Wistar
17.
Toxicol Mech Methods ; 25(8): 645-52, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26108275

RESUMEN

Aristolochic acid I (AAI) affects TGF-ß1/Smad signaling, which causes AA nephropathy (AAN), but the mechanisms are not fully understood. We aimed to clarify whether Arkadia and UCH37 participate in TGF-ß1/Smad signaling via Smad7, and the regulatory mechanisms of Smad7. One side, mice and cultured mouse renal tubular epithelial cells (RTECs) were treated with various AAI doses and concentrations, respectively; on the other side, RTECs were transfected with small interfering RNA (siRNA) expression vectors against Arkadia and UCH37 and then treated with 10 µg/ml AAI. And then detect the mRNA and protein levels of Smad7, UCH37, Arkadia and any other relative factors by RT-PCR and Western blotting. In kidney tissues and RTECs, the mRNA and protein levels of Smad7 decreased with increasing AAI doses concentrations by real-time PCR and Western blotting, whereas those of Arkadia, UCH37, Smad2, Smad3 and TßRI increased. Cells transfected with the Arkadia siRNA expression vector showed reduced mRNA and protein levels of vimentin, α-SMA, Smad2, Smad3 and TßRI after AAI treatment, while those of CK18 and Smad7 increased compared with those of untransfected RTECs. Conversely, cells transfected with the UCH37 siRNA expression vector showed the opposite effect on analyzed signaling molecules after AAI treatment. Arkadia and UCH37 participate in TGF-ß1/Smad signaling-mediated renal fibrosis, and Smad7 blocks TGF-ß1 signaling by inhibiting Smad2/Smad3 phosphorylation and enhancing the degradation of TßRI.


Asunto(s)
Ácidos Aristolóquicos/toxicidad , Carcinógenos/toxicidad , Túbulos Renales/efectos de los fármacos , Proteína smad7/antagonistas & inhibidores , Ubiquitina Tiolesterasa/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación/efectos de los fármacos , Animales , Células Cultivadas , Relación Dosis-Respuesta a Droga , Regulación de la Expresión Génica/efectos de los fármacos , Túbulos Renales/citología , Túbulos Renales/inmunología , Túbulos Renales/metabolismo , Ratones , Nefritis/inducido químicamente , Nefritis/inmunología , Nefritis/metabolismo , Fosforilación/efectos de los fármacos , Procesamiento Proteico-Postraduccional , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Interferencia de ARN , ARN Mensajero/metabolismo , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/agonistas , Receptores de Factores de Crecimiento Transformadores beta/antagonistas & inhibidores , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Transducción de Señal/efectos de los fármacos , Proteína smad7/genética , Proteína smad7/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Ubiquitina Tiolesterasa/antagonistas & inhibidores , Ubiquitina Tiolesterasa/química , Ubiquitina Tiolesterasa/genética , Ubiquitina-Proteína Ligasas/antagonistas & inhibidores , Ubiquitina-Proteína Ligasas/química , Ubiquitina-Proteína Ligasas/genética
18.
Nephrol Dial Transplant ; 29(11): 2020-7, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24166461

RESUMEN

Currently used diagnostic criteria in different endemic (Balkan) nephropathy (EN) centers involve different combinations of parameters, various cut-off values and many of them are not in agreement with proposed international guidelines. Leaders of EN centers began to address these problems at scientific meetings, and this paper is the outgrowth of those discussions. The main aim is to provide recommendations for clinical work on current knowledge and expertise. This document is developed for use by general physicians, nephrologists, urologist, public health experts and epidemiologist, and it is hoped that it will be adopted by responsible institutions in countries harboring EN. National medical providers should cover costs of screening and diagnostic procedures and treatment of EN patients with or without upper urothelial cancers.


Asunto(s)
Nefropatía de los Balcanes , Consenso , Manejo de la Enfermedad , Tamizaje Masivo/métodos , Nefropatía de los Balcanes/clasificación , Nefropatía de los Balcanes/diagnóstico , Nefropatía de los Balcanes/terapia , Humanos
19.
Toxicol Mech Methods ; 24(6): 377-84, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24796935

RESUMEN

Aristolochic acid nephropathy (AAN) is mainly caused by aristolochic acid I (AAI), but the actual mechanism is still uncertain. The current study explored the correlation among the expression of Smad7, p300, histone deacetylase-1 (HDAC1) and the development of AAN using transmission electron microscopy (TEM), RT-PCR, and western blotting in the AAN mouse model and in the AAN cell model. TEM revealed that the renal tubular epithelial cells from the AAI-treated mice presented organelle damages and nuclear deformation. We found that a certain dose of AAI caused renal fibrosis and induced renal tubular epithelial cells to differentiate into myofibroblasts. There was a gradual increase in the expression of HDAC1 mRNA and protein observed using RT-PCR and western blotting in the AAN cell model compared with the control group. Gradual decrease in the expression of Smad7 and p300 mRNA and protein was revealed in the AAN mouse and cell models compared with the control group. These results suggest that AAI dose dependently contributed to the development of AAN, and HDAC1 and p300 participate in the modulation of TGF-ß/Smad pathway-mediated renal interstitial fibrosis.


Asunto(s)
Ácidos Aristolóquicos/toxicidad , Proteína p300 Asociada a E1A/metabolismo , Histona Desacetilasa 1/metabolismo , Enfermedades Renales/inducido químicamente , Actinas/genética , Actinas/metabolismo , Animales , Ácidos Aristolóquicos/administración & dosificación , Relación Dosis-Respuesta a Droga , Proteína p300 Asociada a E1A/genética , Regulación de la Expresión Génica/efectos de los fármacos , Histona Desacetilasa 1/genética , Riñón/efectos de los fármacos , Riñón/ultraestructura , Enfermedades Renales/patología , Ratones , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Smad/genética , Proteínas Smad/metabolismo , Organismos Libres de Patógenos Específicos , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo
20.
Br J Pharmacol ; 181(9): 1452-1473, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38073114

RESUMEN

BACKGROUND AND PURPOSE: Aristolochic acid nephropathy (AAN) is a progressive kidney disease caused by using herbal medicines. Currently, no therapies are available to treat or prevent aristolochic acid nephropathy. Histone deacetylase (HDAC) plays a crucial role in the development and progression of renal disease. We tested whether HDAC inhibitors could prevent aristolochic acid nephropathy and determined the underlying mechanism. EXPERIMENTAL APPROACH: HDACs expression in the aristolochic acid nephropathy model was examined. The activation of PANoptosis of mouse kidney and renal tubular epithelial cell were assessed after exposure to HDAC1 and HDAC2 blockade. Kidney-specific knock-in of proline-serine-threonine-phosphatase-interacting protein 2 (PSTPIP2) mice were used to investigate whether PSTPIP2 affected the production of PANoptosome. KEY RESULTS: Aristolochic acid upregulated the expression of HDAC1 and HDAC2 in the kidneys. Notably, the HDAC1 and HDAC2 specific inhibitor, romidepsin (FK228, depsipeptide), suppressed aristolochic acid-induced kidney injury, epithelial cell pyroptosis, apoptosis and necroptosis (PANoptosis). Moreover, romidepsin upregulated PSTPIP2 in renal tubular epithelial cells, which was enhanced by aristolochic acid treatment. Conditional knock-in of PSTPIP2 in the kidney protected against aristolochic acid nephropathy. In contrast, the knockdown of PSTPIP2 expression in PSTPIP2-knock-in mice restored kidney damage and PANoptosis. PSTPIP2 function was determined in vitro using PSTPIP2 knockdown or overexpression in mouse renal tubular epithelial cells (mTECs). Additionally, PSTPIP2 was found to regulate caspase 8 in aristolochic acid nephropathy. CONCLUSION AND IMPLICATIONS: HDAC-mediated silencing of PSTPIP2 may contribute to aristolochic acid nephropathy. Hence, HDAC1 and HDAC2 specific inhibitors or PSTPIP2 could be valuable therapeutic agents for preventing aristolochic acid nephropathy.


Asunto(s)
Ácidos Aristolóquicos , Enfermedades Renales , Ratones , Animales , Piroptosis , Histona Desacetilasas/metabolismo , Necroptosis , Enfermedades Renales/metabolismo , Ácidos Aristolóquicos/toxicidad , Apoptosis , Histona Desacetilasa 2/genética , Histona Desacetilasa 2/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA