Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
1.
Pharmacol Res ; 208: 107384, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39209083

RESUMEN

Energy metabolism disorder, mainly exhibiting the inhibition of fatty acid degradation and lipid accumulation, is highly related with aging acceleration. However, the intervention measures are deficient. Here, we reported Omega-3 polyunsaturated fatty acids (Omega-3 PUFAs), especially EPA, exerted beneficial effects on maintaining energy metabolism and lipid homeostasis to slow organ aging. As the endogenous agonist of peroxisome proliferator-activated receptor α (PPARα), Omega-3 PUFAs significantly boosted fatty acid ß-oxidation and ATP production in multiple aged organs. Consequently, Omega-3 PUFAs effectively inhibited age-related pathological changes, preserved organ function, and retarded aging process. The beneficial effects of Omega-3 PUFAs were also testified in mfat-1 transgenic mice, which spontaneously generate abundant endogenous Omega-3 PUFAs. In conclusion, our study innovatively demonstrated Omega-3 PUFAs administration in diet slow aging through promoting energy metabolism. The supplement of Omega-3 PUFAs or fat-1 transgene provides a promising therapeutic approach to promote healthy aging in the elderly.


Asunto(s)
Envejecimiento , Metabolismo Energético , Ácidos Grasos Omega-3 , Ratones Transgénicos , PPAR alfa , Animales , Metabolismo Energético/efectos de los fármacos , Ácidos Grasos Omega-3/farmacología , Ácidos Grasos Omega-3/metabolismo , Envejecimiento/efectos de los fármacos , Envejecimiento/metabolismo , PPAR alfa/metabolismo , PPAR alfa/genética , Masculino , Metabolismo de los Lípidos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Humanos
2.
Acta Pharmacol Sin ; 45(2): 366-377, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37770579

RESUMEN

Diabetic nephropathy (DN) is characterized by chronic low-grade renal inflammatory responses, which greatly contribute to disease progression. Abnormal glucose metabolism disrupts renal lipid metabolism, leading to lipid accumulation, nephrotoxicity, and subsequent aseptic renal interstitial inflammation. In this study, we investigated the mechanisms underlying the renal inflammation in diabetes, driven by glucose-lipid metabolic rearrangement with a focus on the role of acetyl-CoA synthetase 2 (ACSS2) in lipid accumulation and renal tubular injury. Diabetic models were established in mice by the injection of streptozotocin and in human renal tubular epithelial HK-2 cells cultured under a high glucose (HG, 30 mmol/L) condition. We showed that the expression levels of ACSS2 were significantly increased in renal tubular epithelial cells (RTECs) from the diabetic mice and human diabetic kidney biopsy samples, and ACSS2 was co-localized with the pro-inflammatory cytokine IL-1ß in RTECs. Diabetic ACSS2-deficient mice exhibited reduced renal tubular injury and inflammatory responses. Similarly, ACSS2 knockdown or inhibition of ACSS2 by ACSS2i (10 µmol/L) in HK-2 cells significantly ameliorated HG-induced inflammation, mitochondrial stress, and fatty acid synthesis. Molecular docking revealed that ACSS2 interacted with Sirtuin 1 (SIRT1). In HG-treated HK-2 cells, we demonstrated that ACSS2 suppressed SIRT1 expression and activated fatty acid synthesis by modulating SIRT1-carbohydrate responsive element binding protein (ChREBP) activity, leading to mitochondrial oxidative stress and inflammation. We conclude that ACSS2 promotes mitochondrial oxidative stress and renal tubular inflammation in DN by regulating the SIRT1-ChREBP pathway. This highlights the potential therapeutic value of pharmacological inhibition of ACSS2 for alleviating renal inflammation and dysregulation of fatty acid metabolic homeostasis in DN. Metabolic inflammation in the renal region, driven by lipid metabolism disorder, is a key factor in renal injury in diabetic nephropathy (DN). Acetyl-CoA synthetase 2 (ACSS2) is abundantly expressed in renal tubular epithelial cells (RTECs) and highly upregulated in diabetic kidneys. Deleting ACSS2 reduces renal fatty acid accumulation and markers of renal tubular injury in diabetic mice. We demonstrate that ACSS2 deletion inhibits ChREBP-mediated fatty acid lipogenesis, mitochondrial oxidative stress, and inflammatory response in RTECs, which play a major role in the progression of diabetic renal tubular injury in the kidney. These findings support the potential use of ACSS2 inhibitors in treating patients with DN.


Asunto(s)
Diabetes Mellitus Experimental , Nefropatías Diabéticas , Humanos , Ratones , Animales , Sirtuina 1/metabolismo , Nefropatías Diabéticas/patología , Acetilcoenzima A/metabolismo , Acetilcoenzima A/uso terapéutico , Diabetes Mellitus Experimental/tratamiento farmacológico , Simulación del Acoplamiento Molecular , Riñón/patología , Factores de Transcripción/metabolismo , Metabolismo de los Lípidos , Glucosa/metabolismo , Ácidos Grasos/metabolismo , Inflamación/metabolismo , Ligasas/metabolismo , Lípidos
3.
BMC Med Educ ; 24(1): 712, 2024 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-38956620

RESUMEN

BACKGROUND: The online-to-offline (O2O) teaching method is recognized as a new educational model that integrates network learning into offline classroom education, while problem-based learning (PBL) is a teaching modality that guides students to apply acquired theoretical knowledge to solve practical problems. However, implementing O2O combined with PBL has not been extensively explored in nephrology residency training. This study aims to explore the efficacy of O2O combined with PBL in the standardized residency training of nephrology by comparing it with the traditional lecture-based teaching (LBT). METHODS: Sixty residency trainees who participated in the standardized training of internal medicine in the nephrology department of the Second Affiliated Hospital of Zhejiang University School of Medicine were equally allocated into O2O combined with PBL (O2O/PBL) or the LBT group demographically matched. Examinations of theory, practice skills, clinical thinking and teaching satisfaction surveys were utilized to assess the teaching effects of the two groups. RESULTS: Participants from the O2O/PBL group outperformed those from the LBT group in the examination of theory (81.233 ± 9.156 vs. 75.800 ± 7.009, mean ± SEM), practice skills (104.433 ± 3.569 vs.100.316 ± 4.628, mean ± SEM) and clinical thinking (88.933 ± 4.473 vs. 86.667 ± 3.844, mean ± SEM). There was no significant difference in the teaching satisfaction between the two groups. CONCLUSION: The current study shows the positive impact of O2O combined with PBL approach on standardized residency training in nephrology without reducing teaching satisfaction.


Asunto(s)
Internado y Residencia , Nefrología , Aprendizaje Basado en Problemas , Aprendizaje Basado en Problemas/métodos , Humanos , Nefrología/educación , Masculino , Femenino , Competencia Clínica , Evaluación Educacional , Enseñanza , Adulto , Instrucción por Computador/métodos , Educación a Distancia
4.
FASEB J ; 33(11): 12630-12643, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31451021

RESUMEN

The discovery of hypoxia-inducible factor (HIF)-prolyl hydroxylase inhibitor (PHI) has revolutionized the treatment strategy for renal anemia. However, the presence of multiple transcription targets of HIF raises safety concerns regarding HIF-PHI. Here, we explored the dose-dependent effect of MK-8617 (MK), a kind of HIF-PHI, on renal fibrosis. MK was administered by oral gavage to mice for 12 wk at doses of 1.5, 5, and 12.5 mg/kg. In vitro, the human proximal tubule epithelial cell line HK-2 was treated with increasing doses of MK administration. Transcriptome profiling was performed, and fibrogenesis was evaluated. The dose-dependent biphasic effects of MK on tubulointerstitial fibrosis (TIF) were observed in chronic kidney disease mice. Accordingly, high-dose MK treatment could significantly enhance TIF. Using RNA-sequencing, combined with in vivo and in vitro experiments, we found that Krüppel-like factor 5 (KLF5) expression level was significantly increased in the proximal tubular cells, which could be transcriptionally regulated by HIF-1α with high-dose MK treatment but not low-dose MK. Furthermore, our study clarified that HIF-1α-KLF5-TGF-ß1 signaling activation is the potential mechanism of high-dose MK-induced TIF, as knockdown of KLF5 reduced TIF in vivo. Collectively, our study demonstrates that high-dose MK treatment initiates TIF by activating HIF-1α-KLF5-TGF-ß1 signaling. These findings provide novel insights into TIF induction by high-dose MK (HIF-PHI), suggesting that the safety dosage window needs to be emphasized in future clinical applications.-Li, Z.-L., Lv, L.-L., Wang, B., Tang, T.-T., Feng, Y., Cao, J.-Y., Jiang, L.-Q., Sun, Y.-B., Liu, H., Zhang, X.-L., Ma, K.-L., Tang, R.-N., Liu, B.-C. The profibrotic effects of MK-8617 on tubulointerstitial fibrosis mediated by the KLF5 regulating pathway.


Asunto(s)
Enfermedades Renales/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Piridazinas/efectos adversos , Pirimidinas/efectos adversos , Transducción de Señal/efectos de los fármacos , Animales , Fibrosis , Perfilación de la Expresión Génica , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Enfermedades Renales/inducido químicamente , Enfermedades Renales/patología , Masculino , Ratones , Piridazinas/farmacología , Pirimidinas/farmacología , Factor de Crecimiento Transformador beta1/metabolismo
5.
Acta Pharmacol Sin ; 41(8): 1111-1118, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32203081

RESUMEN

Some studies have shown that gut microbiota along with its metabolites is closely associated with diabetic mellitus (DM). In this study we explored the relationship between gut microbiota and kidney injuries of early diabetic nephropathy (DN) and its underlying mechanisms. Male SD rats were intraperitoneally injected with streptozotocin to induce DM. DM rats were orally administered compound broad-spectrum antibiotics for 8 weeks. After the rats were sacrificed, their blood, urine, feces, and renal tissues were harvested for analyses. We found that compared with the control rats, DM rats had abnormal intestinal microflora, increased plasma acetate levels, increased proteinuria, thickened glomerular basement membrane, and podocyte foot process effacement in the kidneys. Furthermore, the protein levels of angiotensin II, angiotensin-converting enzyme, and angiotensin II type 1 receptor in the kidneys of DM rats were significantly increased. Administration of broad-spectrum antibiotics in DM rats not only completely killed most intestinal microflora, but also significantly lowered the plasma acetate levels, inhibited intrarenal RAS activation, and attenuated kidney damage. Finally, we showed that plasma acetate levels were positively correlated with intrarenal angiotensin II protein expression (r = 0.969, P < 0.001). In conclusion, excessive acetate produced by disturbed gut microbiota might be involved in the kidney injuries of early DN through activating intrarenal RAS.


Asunto(s)
Acetatos/metabolismo , Diabetes Mellitus Experimental/fisiopatología , Nefropatías Diabéticas/fisiopatología , Disbiosis/fisiopatología , Microbioma Gastrointestinal/fisiología , Sistema Renina-Angiotensina/fisiología , Acetatos/sangre , Animales , Antibacterianos/farmacología , Diabetes Mellitus Experimental/patología , Nefropatías Diabéticas/patología , Microbioma Gastrointestinal/efectos de los fármacos , Riñón/patología , Masculino , Ratas Sprague-Dawley
6.
J Cell Biochem ; 120(5): 7353-7362, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30525213

RESUMEN

The mechanisms that underlie the profibrotic effect of interleukin (IL)-1ß are complicated and not fully understood. Recent evidence has suggested the involvement of the calcium-sensing receptor (CaSR) in tubular injury. Therefore, the current study aimed to investigate whether CaSR mediates IL-1ß-induced collagen expression in cultured mouse inner medullary collecting duct cells (mIMCD3) and to determine the possible downstream signaling effector. The results showed that IL-1ß significantly upregulated the expression of type I and III collagens in a concentration- and time-dependent manner. Moreover, CaSR was expressed in mIMCD3 cells, and its expression was increased by increasing the concentrations and times of IL-1ß treatment. Selective inhibitors (Calhex231 or NPS2143) or the siRNA of CaSR attenuated the enhanced expression of type I and III collagens. Furthermore, IL-1ß increased nuclear ß-catenin protein levels and decreased cytoplasmic ß-catenin expression in cells. In contrast, blockage of CaSR by the pharmacological antagonists or siRNA could partially attenuate such changes in the IL-1ß-induced nuclear translocation of ß-catenin. DKK1, an inhibitor of ß-catenin nuclear translocation, further inhibited the expression of type I and III collagens in cells treated with IL-1ß plus CaSR antagonist. In summary, these data demonstrated that IL-1ß-induced collagen I and III expressions in collecting duct cells might be partially mediated by CaSR and the downstream nuclear translocation of ß-catenin.

7.
J Cell Biochem ; 120(3): 4291-4300, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30260039

RESUMEN

Artemisinin (Art) is isolated from Artemisia annua L. and known as the most effective antimalaria drugs. Previous studies demonstrated that it could exert an immune-regulatory effect on autoimmune diseases. In this study, we first investigated its potential role in tubulointerstitial inflammation and fibrosis in rats with 5/6 nephrectomy. Subtotal nephrectomized (SNx) rats were orally administered Art (100 mg·kg -1 ·d - 1) for 16 weeks. Blood and urine samples were collected for biochemical examination. Kidney tissues were collected for immunohistochemistry and Western blot analyses. Ang II-induced injury of the human kidney 2 (HK-2) cells was used for in vitro study. It was shown that Art could significantly attenuate the renal function decline in SNx rats compared with control. More importantly, Art treatment significantly reduced the tubulointerstitial inflammation and fibrosis, as demonstrated by the evaluation of renal pathology. Furthermore, Art inhibited the activation of NLRP3 inflammasome and NF-κB in the kidneys. In in vitro study, Art pretreatment could significantly prevent the activation of NLRP3 inflammasome and NF-κB in Ang II-treated HK-2 cells, while BAY11-7082 (an inhibitor of NF-κB) significantly inhibited Ang II-induced NLRP3 inflammasome activation. This study suggested that Art could provide renoprotective role by attenuating the tubulointerstitial inflammation and fibrosis in SNx rats by downregulating the NF-κB/NLRP3 signaling pathway.


Asunto(s)
Antiinflamatorios/uso terapéutico , Artemisininas/uso terapéutico , FN-kappa B/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Nefrectomía/efectos adversos , Nefritis Intersticial/tratamiento farmacológico , Nefritis Intersticial/etiología , Animales , Antiinflamatorios/farmacología , Artemisia/química , Artemisininas/farmacología , Línea Celular , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Fibrosis , Humanos , Inflamasomas/efectos de los fármacos , Inflamasomas/metabolismo , Riñón/citología , Riñón/patología , Masculino , Extractos Vegetales/uso terapéutico , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos
8.
J Transl Med ; 17(1): 59, 2019 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-30819181

RESUMEN

BACKGROUND: Diabetic kidney disease (DKD) is the leading cause of end-stage kidney disease (ESKD) in the world. Emerging evidence has shown that urinary mRNAs may serve as early diagnostic and prognostic biomarkers of DKD. In this article, we aimed to first establish a novel bioinformatics-based methodology for analyzing the "urinary kidney-specific mRNAs" and verify their potential clinical utility in DKD. METHODS: To select candidate mRNAs, a total of 127 Affymetrix microarray datasets of diabetic kidney tissues and other tissues from humans were compiled and analyzed using an integrative bioinformatics approach. Then, the urinary expression of candidate mRNAs in stage 1 study (n = 82) was verified, and the one with best performance moved on to stage 2 study (n = 80) for validation. To avoid potential detection bias, a one-step Taqman PCR assay was developed for quantification of the interested mRNA in stage 2 study. Lastly, the in situ expression of the selected mRNA was further confirmed using fluorescent in situ hybridization (FISH) assay and bioinformatics analysis. RESULTS: Our bioinformatics analysis identified sixteen mRNAs as candidates, of which urinary BBOX1 (uBBOX1) levels were significantly upregulated in the urine of patients with DKD. The expression of uBBOX1 was also increased in normoalbuminuric diabetes subjects, while remained unchanged in patients with urinary tract infection or bladder cancer. Besides, uBBOX1 levels correlated with glycemic control, albuminuria and urinary tubular injury marker levels. Similar results were obtained in stage 2 study. FISH assay further demonstrated that BBOX1 mRNA was predominantly located in renal tubular epithelial cells, while its expression in podocytes and urothelium was weak. Further bioinformatics analysis also suggested that tubular BBOX1 mRNA expression was quite stable in various types of kidney diseases. CONCLUSIONS: Our study provided a novel methodology to identify and analyze urinary kidney-specific mRNAs. uBBOX1 might serve as a promising biomarker of DKD. The performance of the selected urinary mRNAs in monitoring disease progression needs further validation.


Asunto(s)
Biología Computacional , Nefropatías Diabéticas/genética , Nefropatías Diabéticas/orina , gamma-Butirobetaína Dioxigenasa/genética , gamma-Butirobetaína Dioxigenasa/orina , Biomarcadores/orina , Bases de Datos Genéticas , Femenino , Humanos , Riñón/metabolismo , Riñón/patología , Masculino , Persona de Mediana Edad , ARN Mensajero/genética , ARN Mensajero/orina , Reproducibilidad de los Resultados , Regulación hacia Arriba/genética
9.
Acta Pharmacol Sin ; 40(7): 989-990, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31015737

RESUMEN

The REFERENCES 1-35 are wrong because of the error in the process of typesetting.

10.
Acta Pharmacol Sin ; 40(4): 468-476, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30446735

RESUMEN

Platelet microparticles (PMPs) are closely associated with diabetic macrovascular complications. The present study aimed to investigate the effects of PMPs in diabetes on aortic vascular endothelial injury and to explore the underlying mechanisms. Peritoneal injection of streptozotocin was used to generate a diabetic rat model in vivo, and human umbilical vein endothelial cells (HUVECs) treated with PMPs were used in vitro. PMP levels in the circulation and aorta tissues were time-dependently increased in streptozotocin-induced diabetic rats at weeks 4, 8, and 12 (P < 0.05). Aspirin significantly inhibited the PMP levels at each time point (P < 0.05). In diabetic rats, the endothelial nitric oxide levels were decreased significantly combined with increased endothelial permeability. PMPs were internalized by HUVECs and primarily accumulated around the nuclei. PMPs inhibited endothelial nitric oxide levels to about 50% and caused approximately twofold increase in reactive oxygen species production. Furthermore, PMPs significantly decreased the endothelial glycocalyx area and expression levels of glypican-1 and occludin (P < 0.05). Interestingly, the PMP-induced endothelial injuries were prevented by raptor siRNA and rapamycin. In conclusion, increased PMPs levels contribute to aortic vascular endothelial injuries in diabetes through activating the mTORC1 pathway.


Asunto(s)
Plaquetas/química , Micropartículas Derivadas de Células/metabolismo , Diabetes Mellitus Experimental/metabolismo , Endotelio Vascular/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Animales , Micropartículas Derivadas de Células/química , Células Cultivadas , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/patología , Endotelio Vascular/patología , Humanos , Masculino , Ratas , Ratas Sprague-Dawley , Estreptozocina
11.
Adv Exp Med Biol ; 1165: 195-232, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31399967

RESUMEN

Finding new therapeutic targets of glomerulosclerosis treatment is an ongoing quest. Due to a living environment of various stresses and pathological stimuli, podocytes are prone to injuries; moreover, as a cell without proliferative potential, loss of podocytes is vital in the pathogenesis of glomerulosclerosis. Thus, sufficient understanding of factors and underlying mechanisms of podocyte injury facilitates the advancement of treating and prevention of glomerulosclerosis. The clinical symptom of podocyte injury is proteinuria, sometimes with loss of kidney functions progressing to glomerulosclerosis. Injury-induced changes in podocyte physiology and function are actually not a simple passive process, but a complex interaction of proteins that comprise the anatomical structure of podocytes at molecular levels. This chapter lists several aspects of podocyte injuries along with potential mechanisms, including glucose and lipid metabolism disorder, hypertension, RAS activation, micro-inflammation, immune disorder, and other factors. These aspects are not technically separated items, but intertwined with each other in the pathogenesis of podocyte injuries.


Asunto(s)
Glomeruloesclerosis Focal y Segmentaria/fisiopatología , Podocitos/citología , Podocitos/patología , Humanos , Hipertensión , Inflamación , Trastornos del Metabolismo de los Lípidos , Proteinuria
12.
Kidney Blood Press Res ; 43(2): 568-581, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29642064

RESUMEN

BACKGROUND/AIMS: Chronic kidney disease (CKD) is a worldwide public health problem. Regardless of the underlying primary disease, CKD tends to progress to end-stage kidney disease, resulting in unsatisfactory and costly treatment. Its common pathogenesis, however, remains unclear. The aim of this study was to provide an unbiased catalog of common gene-expression changes of CKD and reveal the underlying molecular mechanism using an integrative bioinformatics approach. METHODS: We systematically collected over 250 Affymetrix microarray datasets from the glomerular and tubulointerstitial compartments of healthy renal tissues and those with various types of established CKD (diabetic kidney disease, hypertensive nephropathy, and glomerular nephropathy). Then, using stringent bioinformatics analysis, shared differentially expressed genes (DEGs) of CKD were obtained. These shared DEGs were further analyzed by the gene ontology (GO) and pathway enrichment analysis. Finally, the protein-protein interaction networks(PINs) were constructed to further refine our results. RESULTS: Our analysis identified 176 and 50 shared DEGs in diseased glomeruli and tubules, respectively, including many transcripts that have not been previously reported to be involved in kidney disease. Enrichment analysis also showed that the glomerular and tubulointerstitial compartments underwent a wide range of unique pathological changes during chronic injury. As revealed by the GO enrichment analysis, shared DEGs in glomeruli were significantly enriched in exosomes. By constructing PINs, we identified several hub genes (e.g. OAS1, JUN, and FOS) and clusters that might play key roles in regulating the development of CKD. CONCLUSION: Our study not only further reveals the unifying molecular mechanism of CKD pathogenesis but also provides a valuable resource of potential biomarkers and therapeutic targets.


Asunto(s)
Biología Computacional/métodos , Insuficiencia Renal Crónica/patología , Biomarcadores , Conjuntos de Datos como Asunto , Perfilación de la Expresión Génica , Ontología de Genes , Humanos , Glomérulos Renales/patología , Túbulos Renales/patología , Análisis por Micromatrices , Mapas de Interacción de Proteínas , Insuficiencia Renal Crónica/etiología , Insuficiencia Renal Crónica/genética
13.
Acta Pharmacol Sin ; 39(2): 222-229, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28858296

RESUMEN

Glomerular sclerosis is characterized by mesangial cell proliferation and progressive extracellular matrix (ECM) accumulation. CCN3 belongs to the CCN family of matrix proteins; increasing evidence suggests that CCN3 is an endogenous negative regulator of the ECM and fibrosis. However, the exact role of CCN3 in the accumulation of ECM remains unknown. The aim of the present study was to investigate the effects of CCN3 on TGF-ß1-induced production of ECM in human mesangial cells (HMCs) in vitro. Treatment with TGF-ß1 (0.5-2.0 ng/mL) suppressed the mRNA and protein expression of CCN3 in HMCs in dose- and time-dependent manners. Furthermore, treatment with TGF-ß1 significantly increased the expression of the two markers of renal fibrosis, fibronectin (FN) and type I collagen (COLI), in HMCs. Moreover, treatment with TGF-ß1 significantly decreased the expression of metalloproteinase (MMP)-2 and MMP-9, and markedly increased the expression of tissue inhibitor of metalloproteinase (TIMP)-1 in HMCs. Pretreatment of HMCs with exogenous CCN3 (5-500 ng/mL) or overexpression of CCN3 significantly attenuated TGF-ß1-induced changes in FN, COLI, MMP-2, MMP-9 and TIMP-1 in HMCs. These results suggest that CCN3 suppresses TGF-ß1-induced accumulation of ECM in HMCs. CCN3 may have potential as a novel therapeutic target for alleviating glomerulosclerosis.


Asunto(s)
Matriz Extracelular/metabolismo , Células Mesangiales/metabolismo , Proteína Hiperexpresada del Nefroblastoma/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Regulación hacia Abajo , Fibronectinas/genética , Fibronectinas/metabolismo , Humanos , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Proteína Hiperexpresada del Nefroblastoma/genética , ARN Mensajero/metabolismo , Regulación hacia Arriba
14.
Acta Pharmacol Sin ; 39(6): 1022-1033, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29620052

RESUMEN

Inflammation and lipid disorders play crucial roles in synergistically accelerating the progression of diabetic nephropathy (DN). In this study we investigated how inflammation and lipid disorders caused tubulointerstitial injury in DN in vivo and in vitro. Diabetic db/db mice were injected with 10% casein (0.5 mL, sc) every other day for 8 weeks to cause chronic inflammation. Compared with db/db mice, casein-injected db/db mice showed exacerbated tubulointerstitial injury, evidenced by increased secretion of extracellular matrix (ECM) and cholesterol accumulation in tubulointerstitium, which was accompanied by activation of the CXC chemokine ligand 16 (CXCL16) pathway. In the in vitro study, we treated HK-2 cells with IL-1ß (5 ng/mL) and high glucose (30 mmol/L). IL-1ß treatment increased cholesterol accumulation in HK-2 cells, leading to greatly increased ROS production, ECM protein expression levels, which was accompanied by the upregulated expression levels of proteins in the CXCL16 pathway. In contrast, after CXCL16 in HK-2 cells was knocked down by siRNA, the IL-1ß-deteriorated changes were attenuated. In conclusion, inflammation accelerates renal tubulointerstitial lesions in mouse DN via increasing the activity of CXCL16 pathway.


Asunto(s)
Quimiocina CXCL16/metabolismo , Nefropatías Diabéticas/metabolismo , Mediadores de Inflamación/metabolismo , Inflamación/metabolismo , Túbulos Renales/metabolismo , Animales , Caseínas , Línea Celular , Quimiocina CXCL16/genética , Colesterol/metabolismo , Nefropatías Diabéticas/genética , Nefropatías Diabéticas/patología , Modelos Animales de Enfermedad , Proteínas de la Matriz Extracelular/metabolismo , Humanos , Inflamación/inducido químicamente , Inflamación/patología , Túbulos Renales/patología , Masculino , Ratones Endogámicos C57BL , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Factores de Tiempo
15.
Am J Physiol Renal Physiol ; 312(3): F436-F444, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-27582099

RESUMEN

Previous studies have shown that increased parathyroid hormone (PTH) attributable to secondary hyperparathyroidism in chronic kidney disease accelerates the arteriosclerotic fibrosis and calcification. Although the underlying mechanisms remain largely unknown, endothelial cells (ECs) have recently been demonstrated to participate in calcification in part by providing chondrogenic cells via the endothelial-to-mesenchymal transition (EndMT). Therefore, this study aimed to investigate whether elevated PTH could induce endothelial-to-chondrogenic transition in aortic ECs and to determine the possible underlying signaling pathway. We found that treatment of ECs with PTH significantly upregulated the expression of EndMT-related markers. Accordingly, ECs treated with PTH exhibited chondrogenic potential. In vivo, lineage-tracing model-subjected mice with endothelial-specific green fluorescent protein fluorescence to chronic PTH infusion showed a marked increase in the aortic expression of chondrocyte markers, and confocal microscopy revealed the endothelial origin of cells expressing chondrocyte markers in the aorta after PTH infusion. Furthermore, this in vitro study showed that PTH enhanced the nuclear localization of ß-catenin in ECs, whereas ß-catenin siRNA or DKK1, an inhibitor of ß-catenin nuclear translocation, attenuated the upregulation of EndMT-associated and chondrogenic markers induced by PTH. In summary, our study demonstrated that elevated PTH could induce the transition of ECs to chondrogenic cells via EndMT, possibly mediated by the nuclear translocation of ß-catenin.


Asunto(s)
Aorta/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Condrogénesis/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Hormona Paratiroidea/farmacología , Transporte Activo de Núcleo Celular , Animales , Aorta/metabolismo , Aorta/patología , Linaje de la Célula , Células Cultivadas , Relación Dosis-Respuesta a Droga , Células Endoteliales/metabolismo , Células Endoteliales/patología , Humanos , Ratones Transgénicos , Fenotipo , Interferencia de ARN , Transducción de Señal/efectos de los fármacos , Transfección , beta Catenina/genética , beta Catenina/metabolismo
16.
J Biol Chem ; 290(29): 18018-18028, 2015 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-26025362

RESUMEN

Albuminuria contributes to the development and progression of chronic kidney disease by inducing tubulointerstitial inflammation (TI) and fibrosis. However, the exact mechanisms of TI in response to albuminuria are unresolved. We previously demonstrated that NLRP3 and inflammasomes mediate albumin-induced lesions in tubular cells. Here, we further investigated the role of endocytic receptors and lysosome rupture in NLRP3 inflammasome activation. A murine proteinuric nephropathy model was induced by albumin overload as described previously. The priming and activation signals for inflammasome complex formation were evoked simultaneously by albumin excess in tubular epithelial cells. The former signal was dependent on a albumin-triggered NF-κB pathway activation. This process is mediated by the endocytic receptor, megalin and cubilin. However, the silencing of megalin or cubilin inhibited the albumin-induced NLRP3 signal. Notably, subsequent lysosome rupture and the corresponding release of lysosomal hydrolases, especially cathepsin B, were observed in tubular epithelial cells exposed to albumin. Cathepsin B release and distribution are essential for NLRP3 signal activation, and inhibitors of cathepsin B suppressed the NLRP3 signal in tubular epithelial cells. Taken together, our findings suggest that megalin/cubilin and lysosome rupture are involved in albumin-triggered tubular injury and TI. This study provides novel insights into albuminuria-induced TI and implicates the active control of albuminuria as a critical strategy to halt the progression of chronic kidney disease.


Asunto(s)
Albuminuria/inmunología , Proteínas Portadoras/inmunología , Inflamasomas/inmunología , Túbulos Renales/patología , Proteína 2 Relacionada con Receptor de Lipoproteína de Baja Densidad/inmunología , Receptores de Superficie Celular/inmunología , Albúmina Sérica/inmunología , Albuminuria/complicaciones , Albuminuria/patología , Animales , Catepsina B/inmunología , Línea Celular , Humanos , Inflamación/inmunología , Inflamación/patología , Interleucina-18/inmunología , Interleucina-1beta/inmunología , Enfermedades Renales/etiología , Enfermedades Renales/inmunología , Enfermedades Renales/patología , Túbulos Renales/inmunología , Lisosomas/inmunología , Lisosomas/patología , Masculino , Proteína con Dominio Pirina 3 de la Familia NLR , Ratas Wistar
17.
Cardiovasc Diabetol ; 15: 42, 2016 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-26944557

RESUMEN

BACKGROUND: Previous studies have shown that high glucose (HG) induced endothelial cell (EC) damage via a phenotypic transition of EC. There is increasing evidence suggesting the role of inflammatory cytokines in mediated HG-induced EC damage. However, little is known about the potential role of interleukin-1ß (IL-1ß) in the process. The aim of present study was to investigate whether IL-1ß mediated HG-induced phenotypic transition in human aortic endothelial cells (HAECs) and to determine the possible underlying mechanism. METHODS: Primary HAECs were exposed to normal glucose (NG, 5.5 nM), high glucose (HG,30 nM), IL-1ß (10 ng/ml), HG + IL-1ß (10 ng/ml) and HG + anti-IL-1ß antibodies (1000 ng/ml) or HG + IL-1ß small interfering RNA (siRNA). Pathological changes were investigated using confocal microscopy and electron microscopy. Confocal microscopy was performed to detect the co-expression of CD31 and fibroblast specific protein 1 (FSP1). To study the effect of protein kinase C-ß (PKCß) activation on IL-1ß in HAECs, HAECs were stimulated with 30 nM PMA (PKCß activator) and 0.3 µM PKCß inhibition (LY317615) for 48 h in the NG or HG group. The expressions of PKCß and IL-1ß were detected by RT-PCR and Western blot. And the concentration of IL-1ß in the supernatant of HAECs was measured by ELISA. The expressions of FSP1, a-SMA and CD31 were detected by Western blot. RESULTS: It was shown that the HG resulted in significant increase in the expressions of PKCß and IL-1ß in dose-and time-dependent manners. The HG or exogenous IL-1ß alone inhibited the expression of CD31 and markly increased the expressions of FSP1 and α-SMA. Furthermore, we observed that the HG and IL-1ß synergistically increased FSP1 and a-SMA expressions compared with the HG or IL-1ß alone group (P < 0.05). Confocal microscopy revealed a colocalization of CD31 and FSP1 and that some cells acquired spindle-shaped morphologies and a loss of CD31 staining. Electron microscopy showed that the HG resulted in the increased microfilamentation and a roughened endoplasmic reticulum structure in the cytoplasm. However, the changes above were attenuated by the intervention of anti-IL-1ß antibodies or IL-1ß siRNA (P < 0.05). In addition, the PMA induced the expressions of PKCß and IL-1ß in HAECs. The PKCß activation may mediate the effect of the HG on IL-1ß production, which could be attenuated by the PKCß selective inhibitor (LY317615) (P < 0.05). CONCLUSIONS: Our findings suggested that HG-induced phenotypic transition of HAECs might require IL-ß activation via the PKCß pathway.


Asunto(s)
Aorta/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Glucosa/farmacología , Interleucina-1beta/metabolismo , Actinas/metabolismo , Aorta/metabolismo , Aorta/ultraestructura , Proteínas de Unión al Calcio/metabolismo , Células Cultivadas , Relación Dosis-Respuesta a Droga , Células Endoteliales/metabolismo , Células Endoteliales/ultraestructura , Activación Enzimática , Regulación de la Expresión Génica , Glucosa/metabolismo , Humanos , Interleucina-1beta/genética , Interleucina-1beta/farmacología , Fenotipo , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Proteína Quinasa C beta/antagonistas & inhibidores , Proteína Quinasa C beta/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Interferencia de ARN , Proteína de Unión al Calcio S100A4 , Transducción de Señal/efectos de los fármacos , Transfección
18.
BMC Nephrol ; 17(1): 60, 2016 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-27295981

RESUMEN

BACKGROUND: There are limited data on the trends of incidence or prevalence of end stage renal disease (ESRD) in China. To assist in future planning for the ESRD program, the trends of incidence, prevalence and health care costs were analyzed and forecasted to the year 2025 by modeling of historical data from 2004 through 2014. METHODS: Nanjing urban employee basic medical insurance (NJUEBMI) data were obtained from the Nanjing Medical Insurance Information System from 2004 to 2014. The time series forecasting system in SAS 9.4 was used. Each variable was independently forecasted by the fittest model, which was selected automatically or manually. RESULTS: The forecasting models demonstrated mean percent errors of -2.49 to 5.62 %, relative to the observed values. The R-square values for the forecasting models ranged from 0.756 to 0.997. On the basis of trends in the historical data, the models projected that the average annual increase in the NJUEBMI population was 4.77 %, with forecasted values of 5,029,270 in 2025 (95 % CI, 4,960,423-5,098,117). The incidence and prevalence of ESRD were projected to increase by 1.19 and 1.95 % annually and were expected to reach 250.5 pmp (95 % CI, 247.7-253.3) and 1505 pmp(95 % CI, 1450-1560) by 2025. Additionally, the costs associated with ESRD were forecasted to increase at a growth rate of 5.80 % for healthcare costs and 7.25‰ for per capita medical expenses, with forecasted values of ¥600.3 million ($92.4 million) (95 % CI, 541.8-658.9) and ¥99.0 thousand ($15.2 thousand) (95 % CI, 98.6-99.3), respectively, by 2025. The incidence and prevalence of kidney transplantation were projected to decrease by 6.58 and 9.79 % annually. CONCLUSIONS: These projections suggest that the incidence, prevalence, healthcare costs, and per capita medical expenses of ESRD would increase in the NJUEBMI population. They provide a basis for discussing the trends of ESRD in China and facing the challenges from the ESRD program.


Asunto(s)
Costo de Enfermedad , Planes de Asistencia Médica para Empleados/economía , Planes de Asistencia Médica para Empleados/tendencias , Costos de la Atención en Salud/tendencias , Fallo Renal Crónico/economía , Fallo Renal Crónico/epidemiología , China/epidemiología , Predicción , Humanos , Incidencia , Fallo Renal Crónico/terapia , Prevalencia
19.
Acta Pharmacol Sin ; 36(7): 821-30, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26005910

RESUMEN

AIM: NLRP3 inflammasome plays an important role in renal injury and may be a therapeutic target in the treatment of patients with progressive chronic kidney disease. In this study we investigated whether angiotensin II (Ang II)-induced NLRP3 inflammasome activation was linked to endoplasmic reticulum stress (ERS) in human renal proximal tubular cells in vitro. METHODS: Human kidney proximal epithelial cells (HK-2) were pretreated with telmisartan or 4-PBA, and then treated with Ang II. The expression levels of mRNAs and proteins related to NLRP3 inflammasomes and ERS was examined by real-time PCR, Western blot and immunofluorescence. RESULTS: Treatment with Ang II (10, 100, and 1000 nmol/L) increased the expression of the inflammasome markers NLRP3 and ASC, as well as caspase-1, IL-1ß, and IL-18 in dose- and time-dependent manners with peak levels detected at 100 nmol/L and 12 h. Ang II-induced increases in the expression of NLRP3, ASC, caspase-1, IL-1ß, and IL-18 were significantly reduced by pretreatment with telmisartan (1 µmol/L). Immunofluorescence studies showed that Ang II increased the expression of NLRP3 and ASC, which was inhibited by telmisartan. Furthermore, Ang II treatment increased the expression of ERS markers GRP78 and p-eIF2α in dose- and time-dependent manners, which was significantly reduced by telmisartan. Moreover, Ang II-induced increases in the expression of NLRP3, ASC, caspase-1, IL-1ß, and IL-18 were significantly inhibited by pretreatment with the ERS inhibitor 4-PBA (5 mmol/L). CONCLUSION: Ang II treatment induces NLRP3 inflammasome activation in HK-2 cells in vitro and ER stress is involved in this process, which may represent a new mechanism for the renal rennin-angiotensin system to induce tubulointerstitial inflammation.


Asunto(s)
Angiotensina II/toxicidad , Proteínas Portadoras/metabolismo , Estrés del Retículo Endoplásmico/efectos de los fármacos , Estrés del Retículo Endoplásmico/fisiología , Túbulos Renales Proximales/efectos de los fármacos , Túbulos Renales Proximales/metabolismo , Línea Celular , Chaperón BiP del Retículo Endoplásmico , Humanos , Túbulos Renales Proximales/citología , Proteína con Dominio Pirina 3 de la Familia NLR
20.
J Cell Biochem ; 115(10): 1692-701, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24821601

RESUMEN

Emerging evidence shows that increased parathyroid hormone (PTH) accelerates endothelial injury and subsequent organ fibrosis. Although the underlying mechanisms remain largely unknown, the endothelial-to-mesenchymal transition (EndMT) has recently been demonstrated to be a crucial event during fibrotic disorders. Therefore, the present study aimed to investigate whether elevated PTH could induce EndMT in primary human renal glomerular endothelial cells (GECs) and to determine the possible underlying signaling pathway. The expression of EndMT-related markers was determined by real-time PCR, Western blotting, and confocal microscopy. The results showed that PTH receptor (PTHR) was expressed in GECs and its expression was decreased by increasing concentration of PTH. Moreover, PTH significantly inhibited the expression of endothelial marker CD31 and increased the expression of mesenchymal markers FSP1 and α-SMA in concentration- and time-dependent manners. Confocal microscopy revealed an increasing overlap of CD31 with FSP1 in some GECs after PTH treatment. The expression of type I collagen was upregulated by PTH. Furthermore, PTH enhanced the nuclear ß-catenin protein levels, and decreased cytoplasmic ß-catenin expression in GECs was observed. In contrast, DKK1, an inhibitor of ß-catenin nuclear translocation, attenuated such changes in EndMT-related markers induced by PTH. In summary, these data demonstrated that elevated PTH-induced EndMT in human GECs might be partially mediated by the nuclear translocation of ß-catenin.


Asunto(s)
Células Endoteliales/citología , Mesodermo/citología , Hormona Paratiroidea/farmacología , Proteínas Recombinantes/farmacología , beta Catenina/metabolismo , Actinas/biosíntesis , Transporte Activo de Núcleo Celular , Proteínas de Unión al Calcio/biosíntesis , Diferenciación Celular , Células Cultivadas , Colágeno Tipo I/biosíntesis , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Péptidos y Proteínas de Señalización Intercelular/farmacología , Células Mesangiales/citología , Células Mesangiales/metabolismo , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/biosíntesis , Receptor de Hormona Paratiroídea Tipo 1/biosíntesis , Proteína de Unión al Calcio S100A4 , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA