Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 189
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Biochem Biophys Res Commun ; 709: 149807, 2024 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-38552554

RESUMEN

Minimal Change Disease (MCD), which is associated with podocyte injury, is the leading cause of nephrotic syndrome in children. A considerable number of patients experience relapses and require prolonged use of prednisone and immunosuppressants. Multi-drug resistance and frequent relapses can lead to disease progression to focal and segmental glomerulosclerosis (FSGS). To identify potential targets for therapy of podocyte injury, we examined microarray data of mRNAs in glomerular samples from both MCD patients and healthy donors, obtained from the GEO database. Differentially expressed genes (DEGs) were used to construct the protein-protein interactions (PPI) network through the application of the search tool for the retrieval of interacting genes (STRING) tool. The most connected genes in the network were ranked using cytoHubba. 16 hub genes were selected and validated by qRT-PCR. RAC2 was identified as a potential therapeutic target for further investigation. By downregulating RAC2, Adriamycin (ADR)-induced human podocytes (HPCs) injury was attenuated. EHT-1864, a small molecule inhibitor that targets the RAC (RAC1, RAC2, RAC3) family, proved to be more effective than RAC2 silencing in reducing HPCs injury. In conclusion, our research suggests that EHT-1864 may be a promising new molecular drug candidate for patients with MCD and FSGS.


Asunto(s)
Glomeruloesclerosis Focal y Segmentaria , Nefrosis Lipoidea , Podocitos , Humanos , Doxorrubicina/efectos adversos , Glomeruloesclerosis Focal y Segmentaria/inducido químicamente , Glomeruloesclerosis Focal y Segmentaria/tratamiento farmacológico , Glomeruloesclerosis Focal y Segmentaria/genética , Glomérulos Renales , Recurrencia
2.
Clin Sci (Lond) ; 138(13): 777-795, 2024 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-38860674

RESUMEN

Renal tubular injury is considered as the main pathological feature of acute kidney injury (AKI), and mitochondrial dysfunction in renal tubular cells is implicated in the pathogenesis of AKI. The estrogen-related receptor γ (ERRγ) is a member of orphan nuclear receptors which plays a regulatory role in mitochondrial biosynthesis, energy metabolism and many metabolic pathways. Online datasets showed a dominant expression of ERRγ in renal tubules, but the role of ERRγ in AKI is still unknown. In the present study, we investigated the role of ERRγ in the pathogenesis of AKI and the therapeutic efficacy of ERRγ agonist DY131 in several murine models of AKI. ERRγ expression was reduced in kidneys of AKI patients and AKI murine models along with a negative correlation to the severity of AKI. Consistently, silencing ERRγ in vitro enhanced cisplatin-induced tubular cells apoptosis, while ERRγ overexpression in vivo utilizing hydrodynamic-based tail vein plasmid delivery approach alleviated cisplatin-induced AKI. ERRγ agonist DY131 could enhance the transcriptional activity of ERRγ and ameliorate AKI in various murine models. Moreover, DY131 attenuated the mitochondrial dysfunction of renal tubular cells and metabolic disorders of kidneys in AKI, and promoted the expression of the mitochondrial transcriptional factor A (TFAM). Further investigation showed that TFAM could be a target gene of ERRγ and DY131 might ameliorate AKI by enhancing ERRγ-mediated TFAM expression protecting mitochondria. These findings highlighted the protective effect of DY131 on AKI, thus providing a promising therapeutic strategy for AKI.


Asunto(s)
Lesión Renal Aguda , Receptores de Estrógenos , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/genética , Animales , Receptores de Estrógenos/metabolismo , Humanos , Masculino , Ratones , Mitocondrias/metabolismo , Ratones Endogámicos C57BL , Enfermedades Metabólicas/metabolismo , Apoptosis , Modelos Animales de Enfermedad , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Cisplatino , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/genética
3.
J Formos Med Assoc ; 123(4): 496-500, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-37903693

RESUMEN

BACKGROUND: Multisystem inflammatory syndrome in children (MIS-C) is a novel disease associated with COVID-19. The COVID-19 epidemic peaked in May 2022 in Taiwan, and we encountered our first case of MIS-C in late May 2022. We aimed to present patients' clinical manifestations and identify risk factors for shock. METHODS: We included patients diagnosed with MIS-C at two medical centers from May 2022 to August 2022. We separated those patients into two groups according to whether they experienced shock. We collected demographic, clinical manifestation, and laboratory data of the patients and performed statistical analysis between the two groups. RESULTS: We enrolled 28 patients, including 13 (46 %) with shock and 15 (54 %) without shock. The median age was 6.4 years (IQR: 1.9-7.5). In single variable analysis, patients with shock tended to be older, had more neurological symptoms, more conjunctivitis and strawberry tongue, lower lymphocyte count, lower platelet counts, and higher C-reactive protein, higher procalcitonin, higher ferritin, and higher D-dimer levels than those without shock. The area under the ROC curve that used procalcitonin to be the risk factor of shock with MIS-C was 0.815 (95 % CI 0.644 to 0.987). The cutoff value obtained by ROC analysis of procalcitonin was 1.68 ng/mL. With this cutoff, the test characteristics of procalcitonin were as follows: sensitivity 77 %, specificity 93 %, positive predictive value 91 %, negative predictive value 82 %. Multivariable analysis revealed that procalcitonin was the only independent risk factor of shock with MIS-C on admission (OR, 26.00, 95 % CI, 1.01-668.89). CONCLUSIONS: MIS-C patients with high initial procalcitonin levels have higher risks of experiencing shock and may need ICU admission.


Asunto(s)
COVID-19 , COVID-19/complicaciones , Neumonía Viral , Síndrome de Respuesta Inflamatoria Sistémica , Niño , Humanos , Neumonía Viral/epidemiología , Polipéptido alfa Relacionado con Calcitonina , COVID-19/epidemiología , Proteína C-Reactiva/análisis , Estudios Retrospectivos
4.
Clin Genet ; 103(2): 179-189, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36284407

RESUMEN

The "toe syndactyly, telecanthus and anogenital and renal malformations" (STAR) syndrome is a rare X-linked dominant inherited kidney ciliopathy caused by CCNQ gene mutations. Here, we investigated the genotype and phenotype in the first two twin sisters with a novel tail extension CCNQ variant in Asia. Genetic variants of the pedigree were screened using whole-exome sequence analysis and validated by direct Sanger sequencing. The genetic function was investigated through cultured cells and zebrafish embryos transfected with mutant. The proband is suffered from end-stage renal disease, telecanthus, scoliosis, anal atresia, bilateral hydronephrosis pyeloureter dilation and hearing loss, while her twin sister had milder phenotypes. A novel heterozygous variant c.502_518delinsA (p.Val168SerfsTer173) in CCNQ gene was identified in the twins and their asymptomatic mosaic mother. The concurrent deletion of 17 bases and insertion of one base variant led to the loss of 5 amino acids, subsequently caused a 96 more amino acids tail extension delaying the appearance of stop codon. The loss-of-function variant of CCNQ not only led to the impaired expression of cyclin M but also increased the binding affinity of CDK10-cyclin M complex, which is different from the previous study. The research expanded the genotypic and phenotypic spectrum of STAR syndrome.


Asunto(s)
Sindactilia , Pez Cebra , Femenino , Animales , Humanos , Pez Cebra/genética , Riñón/anomalías , Mutación , Fenotipo , Sindactilia/genética , Ciclinas/genética , Linaje
5.
Clin Sci (Lond) ; 137(6): 435-452, 2023 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-36815438

RESUMEN

Cisplatin-induced nephrotoxicity is the main adverse effect of cisplatin-based chemotherapy and highly limits its clinical use. DMXAA, a flavonoid derivative, is a promising vascular disrupting agent and known as an agonist of STING. Although cGAS-STING activation has been demonstrated to mediate cisplatin-induced acute kidney injury (AKI), the role of DMXAA in this condition is unclear. Here, we defined an unexpected and critical role of DMXAA in improving renal function, ameliorating renal tubular injury and cell apoptosis, and suppressing inflammation in cisplatin-induced AKI. Moreover, we confirmed that DMXAA combated AKI in a STING-independent manner, as evidenced by its protective effect in STING global knockout mice subjected to cisplatin. Furthermore, we compared the role of DMXAA with another STING agonist SR717 in cisplatin-treated mice and found that DMXAA but not SR717 protected animals against AKI. To better evaluate the role of DMXAA, we performed transcriptome analyses and observed that both inflammatory and metabolic pathways were altered by DMXAA treatment. Due to the established role of metabolic disorders in AKI, which contributes to kidney injury and recovery, we also performed metabolomics using kidney tissues from cisplatin-induced AKI mice with or without DMXAA treatment. Strikingly, our results revealed that DMXAA improved the metabolic disorders in kidneys of AKI mice, especially regulated the tryptophan metabolism. Collectively, therapeutic administration of DMXAA ameliorates cisplatin-induced AKI independent of STING, suggesting a promising potential for preventing nephrotoxicity induced by cisplatin-based chemotherapy.


Asunto(s)
Lesión Renal Aguda , Xantonas , Ratones , Animales , Cisplatino/efectos adversos , Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/tratamiento farmacológico , Lesión Renal Aguda/prevención & control , Xantonas/metabolismo , Xantonas/farmacología , Xantonas/uso terapéutico , Riñón/metabolismo , Apoptosis , Ratones Endogámicos C57BL
6.
Ecotoxicol Environ Saf ; 268: 115690, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37976933

RESUMEN

The longitudinal associations of urinary concentrations of diphenyl phosphate (DPHP), bis(2-chloroethyl) phosphate (BCEP), and bis(1,3-dichloro-2-propyl) phosphate (BDCPP) with all-cause, cardiovascular, and cancer mortality in a population of adults aged 40 years and older are still unclear. A total of 3238 participants were included in this cohort study. Urinary BCEP levels were positively associated with all-cause mortality and cardiovascular mortality. Specifically, a logarithmic increase in BCEP concentration was related to a 26 % higher risk of all-cause mortality and a 32 % higher risk of cardiovascular mortality. No significant associations were observed for DPHP and BDCPP in relation to mortality. Doseresponse analysis confirmed the linear associations of BCEP with all-cause and cardiovascular mortality and the nonlinear inverted U-shaped association between DPHP exposure and all-cause mortality. Notably, the economic burden associated with BCEP exposure was estimated, and it was shown that concentrations in the third tertile of BCEP exposure incurred approximately 507 billion dollars of financial burden for all-cause mortality and approximately 717 billion dollars for cardiovascular mortality. These results highlight the importance of addressing exposure to BCEP and its potential health impacts on the population. More research is warranted to explore the underlying mechanisms and develop strategies for reducing exposure to this harmful chemical.


Asunto(s)
Enfermedades Cardiovasculares , Retardadores de Llama , Humanos , Adulto , Persona de Mediana Edad , Organofosfatos/toxicidad , Organofosfatos/orina , Retardadores de Llama/toxicidad , Retardadores de Llama/análisis , Estudios de Cohortes , Causas de Muerte , Fosfatos
7.
BMC Pediatr ; 22(1): 692, 2022 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-36460986

RESUMEN

BACKGROUND: Henoch-Schönlein purpura (HSP) with refractory gastrointestinal (GI) symptoms is always difficult to handle because of its resistance to supportive therapies and glucocorticoid. This study aimed to evaluate the efficacy of hemoperfusion (HP) and intravenous immunoglobulins (IVIG) therapies in this population. METHODS: Sixty-four HSP patients with refractory GI involvement (R-GI group) and 64 cases with mild GI symptoms (control group) were retrospectively analyzed in our center from March 2016 to October 2019. In R-GI group, 42 cases (subgroup A) were treated with IVIG and steroid, 13 cases (subgroup B) used HP and steroid, 9 cases (subgroup C) executed a combination of IVIG, HP and steroid. Demographic characteristics, clinical features, laboratory indexes and treatment outcomes were recorded. t-test, One-way ANOVA, Mann-Whitney U test, and multivariate logistic regression were used in comparing differences among subgroups and predicting independent risk factors. RESULTS: Compared with the control group, R-GI cases experienced higher risk of renal involvement (P = 0.000), more steroid exposure (P = 0.000), six times expenses (P = 0.000) and 2.3 times length of hospitalization (P = 0.000). The independent risk factors of R-GI group were elevated neutrophils (OR 1.250 [95% CI 1.130-1.383]) and the percentage of B lymphocytes (OR 1.100 [95% CI 1.026-1.179]) as well as decreased IgG (OR 0.847 [95% CI 0.732-0.98]). In R-GI group, increased age (OR 1.039 [95% CI 1.016-1.062]) and IgM (OR 5.994 [95% CI 1.403-27.611]) were verified to be risk factors of HSP nephritis. All three subgroups could alleviate the symptoms effectively. Compared with those in subgroup A, patients in subgroup B were elder (P = 0.004), had less relapse (P = 0.002), steroid exposure (P = 0.033) and expenses (P = 0.031), more significant decrease of WBC (P = 0.026) after treatment. CONCLUSION: The HSP with refractory GI involvement had much higher risk of medical burden and renal involvement. Both IVIG and HP therapies could ameliorate refractory GI symptoms efficiently. HP therapy tended to reduce the relapse, costs and steroid exposure in its audiences who were cooperated and with stable hemodynamics, while IVIG had better use in younger children.


Asunto(s)
Glomerulonefritis , Hemoperfusión , Vasculitis por IgA , Niño , Humanos , Vasculitis por IgA/complicaciones , Vasculitis por IgA/tratamiento farmacológico , Inmunoglobulinas Intravenosas/uso terapéutico , Estudios Retrospectivos
8.
Am J Physiol Renal Physiol ; 320(4): F608-F616, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33615891

RESUMEN

Stimulator of interferon genes (STING) is an important adaptor in cytosolic DNA-sensing pathways. A recent study found that the deletion of STING ameliorated cisplatin-induced acute kidney injury (AKI), suggesting that STING could serve as a potential target for AKI therapy. Up to now, a series of small-molecule STING inhibitors/antagonists have been identified. However, none of the research was performed to explore the role of human STING inhibitors in AKI. Here, we investigated the effect of a newly generated covalent antagonist, H151, which targets both human and murine STING, in cisplatin-induced AKI. We found that H151 treatment significantly ameliorated cisplatin-induced kidney injury as shown by the improvement of renal function, kidney morphology, and renal inflammation. In addition, tubular cell apoptosis and increased renal tubular injury marker neutrophil gelatinase-associated lipocalin induced by cisplatin were also effectively attenuated in H151-treated mice. Moreover, the mitochondrial injury caused by cisplatin was also reversed as evidenced by improved mitochondrial morphology, restored mitochondrial DNA content, and reversed mitochondrial gene expression. Finally, we observed enhanced mitochondrial DNA levels in the plasma of patients receiving platinum-based chemotherapy compared with healthy controls, which could potentially activate STING signaling. Taken together, these findings suggested that H151 could be a potential therapeutic agent for treating AKI possibly through inhibiting STING-mediated inflammation and mitochondrial injury.NEW & NOTEWORTHY Although various stimulator of interferon genes (STING) inhibitors have been identified, no research was performed to investigate the role of human STING inhibitors in AKI. Here, we evaluated the effect of H151 targeting both human and murine STING on cisplatin-induced AKI and observed a protection against renal injury possibly through ameliorating inflammation and mitochondrial dysfunction.


Asunto(s)
Lesión Renal Aguda/tratamiento farmacológico , Cisplatino/farmacología , Lipocalina 2/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Lesión Renal Aguda/metabolismo , Animales , Apoptosis/efectos de los fármacos , Inflamación/metabolismo , Riñón/efectos de los fármacos , Riñón/metabolismo , Lipocalina 2/metabolismo , Ratones , Mitocondrias/metabolismo , Nefritis/metabolismo , Transducción de Señal/efectos de los fármacos
9.
Kidney Int ; 99(4): 854-869, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33181155

RESUMEN

Emerging evidence has shown that mitochondrial dysfunction is closely related to the pathogenesis of podocytopathy, but the molecular mechanisms mediating mitochondrial dysfunction in podocytes remain unclear. Lon protease 1 is an important soluble protease localized in the mitochondrial matrix, although its exact role in podocyte injury has yet to be determined. Here we investigated the specific role of this protease in podocyte in glomerular injury and the progression of podocytopathy using podocyte-specific Lon protease 1 knockout mice, murine podocytes in culture and kidney biopsy samples from patients with focal segmental glomerular sclerosis and minimal change disease. Downregulated expression of Lon protease 1 was observed in glomeruli of kidney biopsy samples demonstrating a negative correlation with urinary protein levels and glomerular pathology of patients with focal segmental glomerular sclerosis and minimal change disease. Podocyte-specific deletion of Lon protease 1 caused severe proteinuria, impaired kidney function, severe kidney injury and even mortality in mice. Mechanistically, we found that continuous podocyte Lon protease 1 ablation induced mitochondrial homeostasis imbalance and dysfunction, which then led to podocyte injury and glomerular sclerosis. In vitro experiments implicated the kidney protective effect of Lon protease 1, which inhibited mitochondrial dysfunction and podocyte apoptosis. Thus, our findings suggest that the regulation of Lon protease 1 in podocytes may provide a novel therapeutic approach for the podocytopathy.


Asunto(s)
Glomeruloesclerosis Focal y Segmentaria , Podocitos , Proteasa La , Animales , Humanos , Glomérulos Renales , Ratones , Proteinuria/genética
10.
Clin Sci (Lond) ; 135(14): 1707-1726, 2021 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-34255035

RESUMEN

Acute kidney injury (AKI) is a known risk factor for the development of chronic kidney disease (CKD), with no satisfactory strategy to prevent the progression of AKI to CKD. Damage to the renal vascular system and subsequent hypoxia are common contributors to both AKI and CKD. Hypoxia-inducible factor (HIF) is reported to protect the kidney from acute ischemic damage and a novel HIF stabilizer, FG4592 (Roxadustat), has become available in the clinic as an anti-anemia drug. However, the role of FG4592 in the AKI-to-CKD transition remains elusive. In the present study, we investigated the role of FG4592 in the AKI-to-CKD transition induced by unilateral kidney ischemia-reperfusion (UIR). The results showed that FG4592, given to mice 3 days after UIR, markedly alleviated kidney fibrosis and enhanced renal vascular regeneration, possibly via activating the HIF-1α/vascular endothelial growth factor A (VEGFA)/VEGF receptor 1 (VEGFR1) signaling pathway and driving the expression of the endogenous antioxidant superoxide dismutase 2 (SOD2). In accordance with the improved renal vascular regeneration and redox balance, the metabolic disorders of the UIR mice kidneys were also attenuated by treatment with FG4592. However, the inflammatory response in the UIR kidneys was not affected significantly by FG4592. Importantly, in the kidneys of CKD patients, we also observed enhanced HIF-1α expression which was positively correlated with the renal levels of VEGFA and SOD2. Together, these findings demonstrated the therapeutic effect of the anti-anemia drug FG4592 in preventing the AKI-to-CKD transition related to ischemia and the redox imbalance.


Asunto(s)
Lesión Renal Aguda/tratamiento farmacológico , Antioxidantes/farmacología , Glicina/análogos & derivados , Isoquinolinas/farmacología , Regeneración/efectos de los fármacos , Insuficiencia Renal Crónica/tratamiento farmacológico , Lesión Renal Aguda/metabolismo , Animales , Antioxidantes/metabolismo , Modelos Animales de Enfermedad , Fibrosis/tratamiento farmacológico , Glicina/farmacología , Riñón/efectos de los fármacos , Riñón/metabolismo , Masculino , Ratones Endogámicos C57BL , Preparaciones Farmacéuticas/metabolismo , Insuficiencia Renal Crónica/metabolismo , Daño por Reperfusión/tratamiento farmacológico , Daño por Reperfusión/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
11.
Am J Physiol Renal Physiol ; 318(3): F576-F588, 2020 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-31961716

RESUMEN

Nephrotoxicity is a known clinical complication of cisplatin that limits the use of this potent antitumor drug. Cyclic nucleotide phosphodiesterases (PDEs) play complex roles in physiology and pathology. PDE4, which is a member of the PDE family, has four subtypes (PDE4A-PDE4D), and PDE4B plays an important role in inflammation. Thus, in the present study, we investigated the effect of PDE4/PDE4B inhibition on renal function and inflammation in a cisplatin nephrotoxicity model. In mice, cisplatin enhanced mRNA and protein expression of PDE4B in renal tubules. After treatment with the PDE4 inhibitor cilomilast, cisplatin-induced renal dysfunction, renal tubular injury, tubular cell apoptosis, and inflammation were all improved. Next, after silencing PDE4B in vivo, we observed a protective effect against cisplatin nephrotoxicity similar to that of the PDE4 inhibitor. In vitro, cisplatin-induced renal tubular cell death was strikingly ameliorated by the PDE4 inhibitor and PDE4B knockdown along with the blockade of the inflammatory response. Considering the known roles of some cell survival pathways in antagonizing insults, we examined levels of PDE4-associated proteins sirtuin 1, phosphatidylinositol 3-kinase, and phosphorylated AKT in cisplatin-treated renal tubular cells with or without cilomilast treatment. Strikingly, cisplatin treatment downregulated the expression of the above proteins, and this effect was largely abolished by the PDE4 inhibitor. Together, these findings indicate the beneficial role of PDE4/PDE4B inhibition in treating cisplatin nephrotoxicity, possibly through antagonizing inflammation and restoring cell survival signaling pathways.


Asunto(s)
Lesión Renal Aguda/inducido químicamente , Cisplatino/toxicidad , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/metabolismo , Ácidos Ciclohexanocarboxílicos/farmacología , Inflamación/tratamiento farmacológico , Nitrilos/farmacología , Inhibidores de Fosfodiesterasa 4/farmacología , Lesión Renal Aguda/tratamiento farmacológico , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Células Cultivadas , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/genética , Ácidos Ciclohexanocarboxílicos/uso terapéutico , Células Epiteliales/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Túbulos Renales/citología , Masculino , Ratones , Nitrilos/uso terapéutico , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo
12.
Am J Physiol Renal Physiol ; 318(4): F994-F1005, 2020 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-32068461

RESUMEN

Renal ischemia-reperfusion (IR) injury is one of the most common acute kidney injuries, but there is still a lack of effective treatment in the clinical setting. Trehalose (Tre), a natural disaccharide, has been demonstrated to protect against oxidative stress, inflammation, and apoptosis. However, whether it could protect against IR-induced renal injury needs to be investigated. In an in vivo experiment, C57BL/6J mice were pretreated with or without Tre (2 g/kg) through a daily single intraperitoneal injection from 3 days before renal IR surgery. Renal function, apoptosis, oxidative stress, and inflammation were analyzed to evaluate kidney injury. In an in vitro experiment, mouse proximal tubular cells were treated with or without Tre under a hypoxia/reoxygenation condition. Western blot analysis, autophagy flux detection, and apoptosis assay were performed to evaluate the level of autophagy and antiapoptotic effect of Tre. The in vivo results showed that the renal damage induced by IR was ameliorated by Tre treatment, as renal histology and renal function were improved and the enhanced protein levels of kidney injury molecule-1 and neutrophil gelatinase-associated lipocalin were blocked. Moreover, autophagy was activated by Tre pretreatment along with inhibition of the IR injury-induced apoptosis, oxidative stress, and inflammation. The in vitro results showed that Tre treatment activated autophagy and protected against hypoxia/reoxygenation-induced tubular cell apoptosis and oxidative stress. Our results demonstrated that Tre protects against IR-induced renal injury, possibly by enhancing autophagy and blocking oxidative stress, inflammation, and apoptosis, suggesting its potential use for the clinical treatment of renal IR injury.


Asunto(s)
Lesión Renal Aguda/prevención & control , Antiinflamatorios/farmacología , Antioxidantes/farmacología , Autofagia/efectos de los fármacos , Mediadores de Inflamación/metabolismo , Riñón/efectos de los fármacos , Nefritis/prevención & control , Estrés Oxidativo/efectos de los fármacos , Daño por Reperfusión/prevención & control , Trehalosa/farmacología , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/patología , Animales , Apoptosis/efectos de los fármacos , Biomarcadores/metabolismo , Células Cultivadas , Modelos Animales de Enfermedad , Riñón/metabolismo , Riñón/patología , Masculino , Ratones Endogámicos C57BL , Nefritis/metabolismo , Nefritis/patología , Infiltración Neutrófila/efectos de los fármacos , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Transducción de Señal
13.
Am J Physiol Renal Physiol ; 318(1): F96-F106, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31682173

RESUMEN

Cisplatin is one of the most effective antitumor agents, but its clinical use is highly limited by its severe side effects, especially nephrotoxicity. Recently, the active form of gasdermin D (GSDMD), termed GSDMD-N, was identified to mediate pyroptotic inflammatory cell death in several diseases. However, the role of the GSDMD-N fragment in cisplatin-induced acute kidney injury (AKI) remains unclear. In the present study, we found that pyroptosis was induced by cisplatin in both mouse kidney tissues and renal tubular epithelial cells, accompanied by increased expression of the GSDMD-N fragment. In GSDMD knockout mice with cisplatin-induced AKI, we found that cisplatin-induced loss of renal function, renal tubular injury, and inflammation was significantly attenuated compared with wild-type mice. Furthermore, the GSDMD-N fragment was overexpressed by an established rapid plasmid tail vein injection approach to evaluate the role of this cleaved form of GSDMD in AKI. As expected, mice with GSDMD-N fragment overexpression in the kidney were more susceptible to cisplatin-induced AKI than control mice, as evidenced by further elevated serum levels of blood urea nitrogen and creatinine, aggravated renal pathology, increased expression of neutrophil gelatinase-associated lipocalin and kidney injury molecule-1, and enhanced renal inflammatory cytokine secretion, which indicates a pathogenic role of GSDMD-N in cisplatin-induced AKI by triggering cell pyroptosis. Similar results were also observed in renal tubular epithelial cells overexpressing the GSDMD-N fragment. Thus these findings suggested that the activation of GSDMD contributes to cisplatin-induced AKI, possibly through triggering pyroptosis.


Asunto(s)
Lesión Renal Aguda/metabolismo , Antineoplásicos/efectos adversos , Cisplatino/efectos adversos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Riñón/metabolismo , Proteínas de Unión a Fosfato/metabolismo , Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/patología , Animales , Inflamación/inducido químicamente , Inflamación/metabolismo , Inflamación/patología , Péptidos y Proteínas de Señalización Intracelular/genética , Riñón/patología , Masculino , Ratones , Ratones Noqueados , Proteínas de Unión a Fosfato/genética , Piroptosis/fisiología
14.
Am J Physiol Renal Physiol ; 318(5): F1199-F1209, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-32249612

RESUMEN

Lithium is widely used in psychiatry as the golden standard for more than 60 yr due to its effectiveness. However, its adverse effect has been limiting its long-term use in clinic. About 40% of patients taking lithium develop nephrogenic diabetes insipidus (NDI). Lithium can also induce proliferation of collecting duct cells, leading to microcyst formation in the kidney. Lithium was considered an autophagy inducer that might contribute to the therapeutic benefit of neuropsychiatric disorders. Thus, we hypothesized that autophagy may play a role in lithium-induced kidney nephrotoxicity. To address our hypothesis, we fed mice with a lithium-containing diet with chloroquine (CQ), an autophagy inhibitor, concurrently. Lithium-treated mice presented enhanced autophagy activity in the kidney cortex and medulla. CQ treatment significantly ameliorated lithium-induced polyuria, polydipsia, natriuresis, and kaliuresis accompanied with attenuated downregulation of aquaporin-2 and Na+-K+-2Cl- cotransporter protein. The protective effect of CQ on aquaporin-2 protein abundance was confirmed in cultured cortical collecting duct cells. In addition, we found that lithium-induced proliferation of collecting duct cells was also suppressed by CQ as detected by proliferating cell nuclear antigen staining. Moreover, both phosphorylated mammalian target of rapamycin and ß-catenin expression, which have been reported to be increased by lithium and associated with cell proliferation, were reduced by CQ. Taken together, our study demonstrated that CQ protected against lithium-induced NDI and collecting duct cell proliferation possibly through inhibiting autophagy.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Cloroquina/farmacología , Diabetes Insípida Nefrogénica/prevención & control , Túbulos Renales Colectores/efectos de los fármacos , Cloruro de Litio , Animales , Acuaporina 2/genética , Acuaporina 2/metabolismo , Autofagia/efectos de los fármacos , Línea Celular , Diabetes Insípida Nefrogénica/inducido químicamente , Diabetes Insípida Nefrogénica/metabolismo , Diabetes Insípida Nefrogénica/patología , Dinoprostona/orina , Modelos Animales de Enfermedad , Túbulos Renales Colectores/metabolismo , Túbulos Renales Colectores/patología , Masculino , Ratones de la Cepa 129 , Natriuresis/efectos de los fármacos , Fosforilación , Poliuria/inducido químicamente , Poliuria/metabolismo , Poliuria/patología , Poliuria/prevención & control , Miembro 1 de la Familia de Transportadores de Soluto 12/genética , Miembro 1 de la Familia de Transportadores de Soluto 12/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Sustancias Reactivas al Ácido Tiobarbitúrico/metabolismo , beta Catenina/metabolismo
15.
Apoptosis ; 25(1-2): 92-104, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31820187

RESUMEN

Cardiovascular disease (CVD) serves as the major cause of mortality in chronic kidney disease (CKD) patients. The injury of endothelium associated with the long-term challenge of uremic toxins including the toxic indoxyl sulfate (IS) is one of key pathological factors leading to CVD. However, the mechanisms of uremic toxins, especially the IS, resulting in endothelial injury, remain unclear. miR-214 was reported to contribute to the pathogenesis of cardiovascular diseases, while its role in IS-induced endothelial cell apoptosis is unknown. In this study, we investigated the role of microRNA-214 (miR-214) in IS-induced endothelial cell apoptosis and the underlying mechanisms using mouse aortic endothelial cells (MAECs). Following IS treatment, miR-214 was significantly downregulated in MAECs in line with enhanced cell apoptosis. Meanwhile, COX-2 was upregulated at both mRNA and protein levels along with increased secretion of PGE2 in medium. To define the role of miR-214 in IS-induced endothelial cell apoptosis, we modulated miR-214 level in MAECs and found that overexpression of miR-214 markedly attenuated endothelial cell apoptosis, while antagonism of miR-214 deteriorated cell death after IS challenge. Further analyses confirmed that COX-2 is a target gene of miR-214, and the inhibition of COX-2 by a specific COX-2 inhibitor NS-398 strikingly attenuated IS-induced endothelial cell apoptosis along with a significant blockade of PGE2 secretion. In conclusion, this study demonstrated an important role of miR-214 in protecting against endothelial cell damage induced by IS possibly by direct downregulation of COX-2/PGE2 axis.


Asunto(s)
Apoptosis/efectos de los fármacos , Ciclooxigenasa 2/metabolismo , Células Endoteliales/citología , Células Endoteliales/metabolismo , Indicán/toxicidad , MicroARNs/metabolismo , Animales , Ciclooxigenasa 2/genética , Células Endoteliales/efectos de los fármacos , Humanos , Masculino , Ratones , MicroARNs/genética
16.
Clin Sci (Lond) ; 134(12): 1357-1376, 2020 06 26.
Artículo en Inglés | MEDLINE | ID: mdl-32490513

RESUMEN

Non-specific inhibition of Rho-associated kinases (ROCKs) alleviated renal fibrosis in the unilateral ureteral obstruction (UUO) model, while genetic deletion of ROCK1 did not affect renal pathology in mice. Thus, whether ROCK2 plays a role in renal tubulointerstitial fibrosis needs to be clarified. In the present study, a selective inhibitor against ROCK2 or genetic approach was used to investigate the role of ROCK2 in renal tubulointerstitial fibrosis. In the fibrotic kidneys of chronic kidney diseases (CKDs) patients, we observed an enhanced expression of ROCK2 with a positive correlation with interstitial fibrosis. In mice, the ROCK2 protein level was time-dependently increased in the UUO model. By treating CKD animals with KD025 at the dosage of 50 mg/kg/day via intraperitoneal injection, the renal fibrosis shown by Masson's trichrome staining was significantly alleviated along with the reduced expression of fibrotic genes. In vitro, inhibiting ROCK2 by KD025 or ROCK2 knockdown/knockout significantly blunted the pro-fibrotic response in transforming growth factor-ß1 (TGF-ß1)-stimulated mouse renal proximal tubular epithelial cells (mPTCs). Moreover, impaired cellular metabolism was reported as a crucial pathogenic factor in CKD. By metabolomics analysis, we found that KD025 restored the metabolic disturbance, including the impaired glutathione metabolism in TGF-ß1-stimulated tubular epithelial cells. Consistently, KD025 increased antioxidative stress enzymes and nuclear erythroid 2-related factor 2 (Nrf2) in fibrotic models. In addition, KD025 decreased the infiltration of macrophages and inflammatory response in fibrotic kidneys and blunted the activation of macrophages in vitro. In conclusion, inhibition of ROCK2 may serve as a potential novel therapy for renal tubulointerstitial fibrosis in CKD.


Asunto(s)
Células Epiteliales/enzimología , Túbulos Renales Proximales/patología , Enfermedades Metabólicas/enzimología , Quinasas Asociadas a rho/antagonistas & inhibidores , Adolescente , Animales , Antiinflamatorios/farmacología , Niño , Preescolar , Modelos Animales de Enfermedad , Células Epiteliales/efectos de los fármacos , Femenino , Fibrosis , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Humanos , Lactante , Inflamación/patología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Enfermedades Metabólicas/patología , Ratones , Factor 2 Relacionado con NF-E2/metabolismo , Estrés Oxidativo/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Células RAW 264.7 , Proteína Smad2/metabolismo , Factor de Crecimiento Transformador beta1/farmacología , Regulación hacia Arriba/efectos de los fármacos , Obstrucción Ureteral/enzimología , Obstrucción Ureteral/patología , Quinasas Asociadas a rho/metabolismo
17.
Pediatr Nephrol ; 35(7): 1351-1357, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32333285

RESUMEN

Coronavirus disease 2019 (COVID-19) has rapidly spread not only in China but throughout the world. Children with kidney failure (chronic kidney disease (CKD) stage 5) are at significant risk for COVID-19. In turn, a set of recommendations for the prevention and control of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and COVID-19 in pediatric hemodialysis (HD) centers and in home peritoneal dialysis (PD) settings have been proposed. The recommendations are based on the epidemiological features of the SARS-CoV-2 virus and COVID-19 disease, susceptibility factors, and preventive and control strategies. These recommendations will be updated as new information regarding SARS-CoV-2 and COVID-19 becomes available.


Asunto(s)
Infecciones por Coronavirus/prevención & control , Transmisión de Enfermedad Infecciosa/prevención & control , Control de Infecciones/normas , Fallo Renal Crónico/terapia , Pandemias/prevención & control , Neumonía Viral/prevención & control , Diálisis Renal/normas , Betacoronavirus , COVID-19 , Niño , Consenso , Infecciones por Coronavirus/diagnóstico , Infecciones por Coronavirus/transmisión , Epidemias , Humanos , Control de Infecciones/métodos , Neumonía Viral/diagnóstico , Neumonía Viral/transmisión , Guías de Práctica Clínica como Asunto/normas , SARS-CoV-2
18.
BMC Pediatr ; 20(1): 218, 2020 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-32410653

RESUMEN

BACKGROUND: Lipin-1, encoded by LPIN1 gene, serves as an enzyme and a transcriptional co-regulator to regulate lipid metabolism and mitochondrial respiratory chain. Autosomal recessive mutations in LPIN1 were recognized as one of the most common causes of pediatric recurrent rhabdomyolysis in western countries. However, to date, there were only a few cases reported in Asian group. This study aims to report the first pediatric case of recurrent rhabdomyolysis with a novel LPIN1 mutation in China mainland in order to raise the awareness of both pediatricians and patients. CASE PRESENTATIONS: Here we report a Chinese pediatric case of recurrent rhabdomyolysis with compound heterozygous variants (p.Arg388* and p.Arg810Cys) in the LPIN1 gene. The c.2428C > T was a novel missense variant involved Arg-to-Cys substitution at position 810 (p.Arg810Cys), located in the highly conserved region which predicted to be damaging by multiple algorithms. The patient manifested as cola-colored urine, muscle weakness and tenderness, as well as acute kidney injury with peak blood creatine kinase level 109,570 U/l in 19-month old. In his second episode of 9 years old, the symtoms were relatively milder with peak creatine kinase level 50,948 U/l. He enjoyed quite normal life between the bouts but slightly elevation of serum creatine kinase level during the fever or long-term exercises. Prolonged weight training combined with calorie deprivation were speculated to be the triggers of his illness. Prompt symptomatic therapy including fluid therapy and nutritional support was given and the patient recovered soon. CONCLUSIONS: LPIN1-related rhabdomyolysis is still quite new to physicians due to its seemly low-incidence especially in Asian countries. In the future, more active genetic test strategy and detailed prophylactic care education should be taken in patients with severe recurrent rhabdomyolysis, who are the high risk group of LPIN1 genetic defects.


Asunto(s)
Fosfatidato Fosfatasa , Rabdomiólisis , Asia , Niño , China , Ejercicio Físico , Humanos , Masculino , Fosfatidato Fosfatasa/genética , Rabdomiólisis/diagnóstico , Rabdomiólisis/genética
19.
J Cell Biochem ; 120(10): 17767-17778, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31131474

RESUMEN

BACKGROUND: Atherosclerosis involves disorders in lipoprotein metabolism and inflammation. Mitochondrial dysfunction plays a critical role in promoting cell apoptosis, inflammation, and oxidative stress involved in the progress of atherosclerosis. Whereas the direct effect of mitochondrial activity modulation on atherogenesis remains unclear. This study evaluated the effect of a mitochondrial complex inhibitor on atherosclerosis in ApoE-deficient mice as well as the potential mechanisms. METHODS AND RESULTS: We treated ApoE-deficient mice with mitochondrial complex I inhibitor rotenone in the western diet and found that rotenone attenuated early and advanced atherosclerosis with no effect on serum lipoprotein levels. Mechanistic investigation showed that rotenone suppressed primary macrophage foam cell formation possibly by suppressing CD36. In addition, we also found that the inhibitory effect of rotenone on VSMC proliferation and migration possibly by targeting the PI3K/AKT signaling. Consistently, mitochondrial complex III inhibitor azoxystrobin also exhibited similar actions as rotenone in VSMCs but not in macrophages. CONCLUSIONS: Inhibition of mitochondrial activity could significantly attenuate atherosclerosis possibly by modulating CD36-mediated macrophage foam cell formation and PI3K/AKT signaling pathway-associated VSMC activation. Targeting mitochondrial activity might be the potential therapeutic strategy for atherosclerosis.


Asunto(s)
Apolipoproteínas E/deficiencia , Aterosclerosis/metabolismo , Aterosclerosis/patología , Células Espumosas/metabolismo , Mitocondrias/metabolismo , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/patología , Animales , Apolipoproteínas E/metabolismo , Antígenos CD36/metabolismo , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Complejo I de Transporte de Electrón/antagonistas & inhibidores , Complejo I de Transporte de Electrón/metabolismo , Complejo III de Transporte de Electrones/antagonistas & inhibidores , Complejo III de Transporte de Electrones/metabolismo , Células Espumosas/efectos de los fármacos , Lípidos/química , Lipoproteínas LDL/farmacología , Masculino , Ratones Noqueados , Mitocondrias/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Pirimidinas/farmacología , Rotenona/toxicidad , Estrobilurinas/farmacología
20.
Am J Physiol Renal Physiol ; 317(3): F593-F605, 2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31268353

RESUMEN

Diabetic nephropathy (DN) has become the main cause of end-stage renal disease worldwide, but the efficacy of current therapeutic strategies on DN remains unsatisfactory. Recent research has reported the involvement of metabolic rearrangement in the pathological process of DN, and of all the disturbances in metabolism, mitochondria serve as key regulatory hubs. In the present study, high-resolution mass spectrometry-based nontarget metabolomics was used to uncover the metabolic characteristics of the early diabetic kidney with or without the inhibition of mitochondrial activity. At first, we observed a moderate enhancement of mitochondrial complex-1 activity in the diabetic kidney, which was completely normalized by the specific mitochondrial complex-1 inhibitor rotenone (ROT). Meanwhile, metabolomics data indicated an overactivated pentose phosphate pathway, purine and pyrimidine metabolism, hexosamine biosynthetic pathway, and tricarboxylic acid cycle, which were strikingly corrected by ROT. In addition, ROT also strikingly corrected imbalanced redox homeostasis, possibly by increasing the ratio of antioxidant metabolites glutathione and NADPH against their oxidative form. In agreement with the improved metabolic status and oxidative response, ROT attenuated glomerular and tubular injury efficiently. Fibrotic markers (fibronectin, α-smooth muscle actin, collagen type I, and collagen type III), inflammatory factors (TNF-α, IL-1ß, and ICAM-1), and oxidative stress were all markedly blocked by ROT. In vitro, ROT dose dependently attenuated high glucose-induced proliferation and extracellular matrix production in mesangial cells. Collectively, these findings revealed that the overactivation of mitochondrial activity in the kidney could contribute to metabolic disorders and the pathogenesis of early DN.


Asunto(s)
Diabetes Mellitus Experimental/complicaciones , Nefropatías Diabéticas/etiología , Metabolismo Energético , Riñón/metabolismo , Mitocondrias/metabolismo , Animales , Glucemia/metabolismo , Línea Celular , Proliferación Celular , Cromatografía Líquida de Alta Presión , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/metabolismo , Nefropatías Diabéticas/tratamiento farmacológico , Nefropatías Diabéticas/metabolismo , Nefropatías Diabéticas/patología , Complejo I de Transporte de Electrón/antagonistas & inhibidores , Complejo I de Transporte de Electrón/metabolismo , Metabolismo Energético/efectos de los fármacos , Fibrosis , Homeostasis , Mediadores de Inflamación/metabolismo , Riñón/efectos de los fármacos , Riñón/patología , Masculino , Metabolómica/métodos , Ratones Endogámicos C57BL , Mitocondrias/efectos de los fármacos , Mitocondrias/patología , Nefrectomía , Estrés Oxidativo , Rotenona/farmacología , Espectrometría de Masa por Ionización de Electrospray , Estreptozocina , Espectrometría de Masas en Tándem , Desacopladores/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA