Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Redox Biol ; 67: 102868, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37690165

RESUMO

Kidney fibrosis is associated with tubular injury, oxidative stress and activation of interstitial fibroblasts. However, whether these events are somehow connected is poorly understood. In this study, we show that glutathione peroxidase-3 (GPX3) depletion in renal tubular epithelium after kidney injury plays a central role in orchestrating an oxidatively stressed extracellular microenvironment, which drives interstitial fibroblast activation and proliferation. Through transcriptional profiling by RNA-sequencing, we found that the expression of GPX3 was down-regulated in various models of chronic kidney disease (CKD), which was correlated with induction of nicotinamide adenine dinucleotide phosphate (NAPDH) oxidase-4 (NOX4). By using decellularized extracellular matrix (ECM) scaffold, we demonstrated that GPX3-depleted extracellular microenvironment spontaneously induced NOX4 expression and reactive oxygen species (ROS) production in renal fibroblasts and triggered their activation and proliferation. Activation of NOX4 by advanced oxidation protein products (AOPPs) mimicked the loss of GPX3, increased the production of ROS, stimulated fibroblast activation and proliferation, and activated protein kinase C-α (PKCα)/mitogen-activated protein kinase (MAPK)/signal transducer and activator of transcription 3 (STAT3) signaling. Silencing NOX4 or inhibition of MAPK with small molecule inhibitors hampered fibroblast activation and proliferation. In mouse model of CKD, knockdown of NOX4 repressed renal fibroblast activation and proliferation and alleviated kidney fibrosis. These results indicate that loss of GPX3 orchestrates an oxidatively stressed extracellular microenvironment, which promotes fibroblast activation and proliferation through a cascade of signal transduction. Our studies underscore the crucial role of extracellular microenvironment in driving fibroblast activation and kidney fibrosis.


Assuntos
Rim , Insuficiência Renal Crônica , Camundongos , Animais , Espécies Reativas de Oxigênio/metabolismo , Rim/metabolismo , Insuficiência Renal Crônica/metabolismo , Fibroblastos/metabolismo , NADPH Oxidase 4/genética , NADPH Oxidase 4/metabolismo , Fibrose
2.
Inflamm Res ; 72(8): 1567-1581, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37438583

RESUMO

BACKGROUND: Intercellular communication between macrophages and peritoneal mesothelial cells (PMCs) has been suggested as a key factor regulating peritonitis development. Here, we explored whether PPARγ (peroxisome proliferator-activated receptor gamma) can be packaged into macrophage exosomes to mediate intercellular communication and regulate peritonitis. METHODS: Macrophage exosomes were isolated by ultracentrifugation and identified by nanoparticle tracking analysis and transmission electron microscopy. Proteomic analysis of macrophage-derived exosomes was performed using mass spectrometry. Co-culture models of supernatants or exosomes with PMCs, as well as a mouse peritonitis model induced by lipopolysaccharide (LPS), were employed. RESULTS:  In this study, using stable Raw264.7 cells overexpressing GFP-FLAG-PPARγ (OE-PPARγ), we found that PPARγ inhibited LPS-induced inflammatory responses in Raw264.7 cells and that PPARγ was incorporated into macrophage exosomes during this process. Overexpression of PPARγ mainly regulated the secretion of differentially expressed exosomal proteins involved in the biological processes of protein transport, lipid metabolic process, cell cycle, apoptotic process, DNA damage stimulus, as well as the KEGG pathway of salmonella infection. Using co-culture models and mouse peritonitis model, we showed that exosomes from Raw264.7 cells overexpressing PPARγ inhibited LPS-induced inflammation in co-cultured human PMCs and in mice through downregulating CD14 and TLR4, two key regulators of the salmonella infection pathway. Pretreatment of the PPARγ inhibitor GW9662 abolished the anti-inflammatory effect of exosomes from Raw264.7 OE-PPARγ cells on human PMCs. CONCLUSIONS: These results suggested that overexpression of PPARγ largely altered the proteomic profile of macrophage exosomes and that exosomal PPARγ from macrophages acted as a regulator of intercellular communication to suppress LPS-induced inflammatory responses in vitro and in vivo via negatively regulating the CD14/TLR4 axis.


Assuntos
Fenômenos Biológicos , Peritonite , Camundongos , Humanos , Animais , PPAR gama/metabolismo , Lipopolissacarídeos/farmacologia , Receptor 4 Toll-Like/metabolismo , Proteômica , Macrófagos/metabolismo , Peritonite/induzido quimicamente
3.
Sheng Li Xue Bao ; 75(1): 1-9, 2023 Feb 25.
Artigo em Chinês | MEDLINE | ID: mdl-36859829

RESUMO

This study was designed to evaluate the protective effect of CPD1, a novel phosphodiesterase 5 inhibitor, on renal interstitial fibrosis after unilateral renal ischemia-reperfusion injury (UIRI). Male BALB/c mice were subjected to UIRI, and treated with CPD1 once daily (i.g, 5 mg/kg). Contralateral nephrectomy was performed on day 10 after UIRI, and the UIRI kidneys were harvested on day 11. Hematoxylin-eosin (HE), Masson trichrome and Sirius Red staining methods were used to observe the renal tissue structural lesions and fibrosis. Immunohistochemical staining and Western blot were used to detect the expression of proteins related to fibrosis. HE, Sirius Red and Masson trichrome staining showed that CPD1-treated UIRI mice had lower extent of tubular epithelial cell injury and deposition of extracellular matrix (ECM) in renal interstitium compared with those in the fibrotic mouse kidneys. The results from immunohistochemistry and Western blot assay indicated significantly decreased protein expressions of type I collagen, fibronectin, plasminogen activator inhibitor-1 (PAI-1) and α-smooth muscle actin (α-SMA) after CPD1 treatment. In addition, CPD1 dose-dependently inhibited the expression of ECM-related proteins induced by transforming growth factor ß1 (TGF-ß1) in normal rat kidney interstitial fibroblasts (NRK-49F) and human renal tubular epithelial cell line (HK-2). In summary, the novel PDE inhibitor, CPD1, displays strong protective effects against UIRI and fibrosis by suppressing TGF-ß signaling pathway and regulating the balance between ECM synthesis and degradation through PAI-1.


Assuntos
Nefropatias , Inibidores da Fosfodiesterase 5 , Animais , Humanos , Masculino , Camundongos , Ratos , Proteínas da Matriz Extracelular , Fibrose , Rim , Inibidor 1 de Ativador de Plasminogênio
5.
Front Endocrinol (Lausanne) ; 13: 864407, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35923621

RESUMO

Background: This study aimed to identify biological markers for diabetic nephropathy (DN) and explore their underlying mechanisms. Methods: Four datasets, GSE30528, GSE47183, GSE104948, and GSE96804, were downloaded from the Gene Expression Omnibus (GEO) database. The differentially expressed genes (DEGs) were identified using the "limma" package, and the "RobustRankAggreg" package was used to screen the overlapping DEGs. The hub genes were identified using cytoHubba of Cytoscape. Logistic regression analysis was used to further analyse the hub genes, followed by receiver operating characteristic (ROC) curve analysis to predict the diagnostic effectiveness of the hub genes. Correlation analysis and enrichment analysis of the hub genes were performed to identify the potential functions of the hub genes involved in DN. Results: In total, 55 DEGs, including 38 upregulated and 17 downregulated genes, were identified from the three datasets. Four hub genes (FN1, CD44, C1QB, and C1QA) were screened out by the "UpSetR" package, and FN1 was identified as a key gene for DN by logistic regression analysis. Correlation analysis and enrichment analysis showed that FN1 was positively correlated with four genes (COL6A3, COL1A2, THBS2, and CD44) and with the development of DN through the extracellular matrix (ECM)-receptor interaction pathway. Conclusions: We identified four candidate genes: FN1, C1QA, C1QB, and CD44. On further investigating the biological functions of FN1, we showed that FN1 was positively correlated with THBS2, COL1A2, COL6A3, and CD44 and involved in the development of DN through the ECM-receptor interaction pathway. THBS2, COL1A2, COL6A3, and CD44 may be novel biomarkers and target therapeutic candidates for DN.


Assuntos
Diabetes Mellitus , Nefropatias Diabéticas , Biomarcadores , Biologia Computacional , Nefropatias Diabéticas/genética , Perfilação da Expressão Gênica , Humanos , Transdução de Sinais/genética
6.
Mol Cell Biochem ; 477(7): 1959-1971, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-35380292

RESUMO

OBJECTIVE: Peritoneal fibrosis (PF) is commonly induced by bioincompatible dialysate exposure during peritoneal dialysis, but the underlying mechanisms remain elusive. This study aimed to investigate the roles of peroxisome proliferator-activated receptor gamma (PPARγ) in PF pathogenesis. METHODS: Rat and cellular PF models were established by high glucose dialysate and lipopolysaccharide treatments. Serum creatinine, urea nitrogen, and glucose contents were detected by ELISA. Histological evaluation was done through H&E and Masson staining. GLUT1, PPARγ, and other protein expression were measured by qRT-PCR, western blotting, and IHC. PPARγ and GLUT1 subcellular distribution were detected using confocal microscopy. Cell proliferation was assessed by MTT and Edu staining. RESULTS: Serum creatinine, urea nitrogen and glucose, and PPARγ and GLUT1 expression in rat PF model were reduced by PPARγ agonists Rosiglitazone or 15d-PGJ2 and elevated by antagonist GW9662. Rosiglitazone or 15d-PGJ2 repressed and GW9662 aggravated peritoneal fibrosis in rat PF model. PPARγ and GLUT1 were mainly localized in nucleus and cytosols of peritoneal mesothelial cells, respectively, which were reduced in cellular PF model, enhanced by Rosiglitazone or 15d-PGJ2, and repressed by GW9662. TGF-ß and a-SMA expression was elevated in cellular PF model, which was inhibited by Rosiglitazone or 15d-PGJ2 and promoted by GW9662. PPARγ silencing reduced GLUT1, elevated a-SMA and TGF-b expression, and promoted peritoneal mesothelial cell proliferation, which were oppositely changed by PPARγ overexpression. CONCLUSION: PPARγ inhibited high glucose-induced peritoneal fibrosis progression through elevating GLUT1 expression and repressing peritoneal mesothelial cell proliferation.


Assuntos
Transportador de Glucose Tipo 1 , PPAR gama , Fibrose Peritoneal , Tiazolidinedionas , Animais , Proliferação de Células , Creatinina , Soluções para Diálise/farmacologia , Glucose/farmacologia , Transportador de Glucose Tipo 1/metabolismo , Nitrogênio/metabolismo , Nitrogênio/farmacologia , PPAR gama/agonistas , PPAR gama/genética , Fibrose Peritoneal/induzido quimicamente , Fibrose Peritoneal/genética , Prostaglandina D2 , Ratos , Rosiglitazona/farmacologia , Tiazolidinedionas/farmacologia , Fator de Crescimento Transformador beta/metabolismo , Ureia
7.
Nat Commun ; 13(1): 438, 2022 01 21.
Artigo em Inglês | MEDLINE | ID: mdl-35064106

RESUMO

Loss of Klotho, an anti-aging protein, plays a critical role in the pathogenesis of chronic kidney diseases. As Klotho is a large transmembrane protein, it is challenging to harness it as a therapeutic remedy. Here we report the discovery of a Klotho-derived peptide 1 (KP1) protecting kidneys by targeting TGF-ß signaling. By screening a series of peptides derived from human Klotho protein, we identified KP1 that repressed fibroblast activation by binding to TGF-ß receptor 2 (TßR2) and disrupting the TGF-ß/TßR2 engagement. As such, KP1 blocked TGF-ß-induced activation of Smad2/3 and mitogen-activated protein kinases. In mouse models of renal fibrosis, intravenous injection of KP1 resulted in its preferential accumulation in injured kidneys. KP1 preserved kidney function, repressed TGF-ß signaling, ameliorated renal fibrosis and restored endogenous Klotho expression. Together, our findings suggest that KP1 recapitulates the anti-fibrotic action of Klotho and offers a potential remedy in the fight against fibrotic kidney diseases.


Assuntos
Rim/metabolismo , Rim/patologia , Proteínas Klotho/química , Peptídeos/farmacologia , Substâncias Protetoras/farmacologia , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo , Sequência de Aminoácidos , Animais , Linhagem Celular , Modelos Animais de Doenças , Fibrose , Humanos , Inflamação/patologia , Rim/lesões , Rim/fisiopatologia , Nefropatias/complicações , Nefropatias/patologia , Masculino , Camundongos Endogâmicos BALB C , Peptídeos/química , Fosforilação/efeitos dos fármacos , Ligação Proteica , Ratos , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Traumatismo por Reperfusão/complicações , Traumatismo por Reperfusão/patologia , Traumatismo por Reperfusão/fisiopatologia , Proteínas Smad/metabolismo , Obstrução Ureteral/complicações , Obstrução Ureteral/patologia
8.
Kidney Int ; 95(4): 830-845, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30770219

RESUMO

Podocyte injury is the major cause of proteinuria in primary glomerular diseases. Oxidative stress has long been thought to play a role in triggering podocyte damage; however, the underlying mechanism remains poorly understood. Here we show that the Wnt/ß-catenin pathway is involved in mediating oxidative stress-induced podocyte dysfunction. Advanced oxidation protein products, a marker and trigger of oxidative stress, were increased in the serum of patients with chronic kidney disease and correlated with impaired glomerular filtration, proteinuria, and circulating level of Wnt1. Both serum from patients with chronic kidney disease and exogenous advanced oxidation protein products induced Wnt1 and Wnt7a expression, activated ß-catenin, and reduced expression of podocyte-specific markers in vitro and in vivo. Blockade of Wnt signaling by Klotho or knockdown of ß-catenin by shRNA in podocytes abolished ß-catenin activation and the upregulation of fibronectin, desmin, matrix metalloproteinase-9, and Snail1 triggered by advanced oxidation protein products. Furthermore, conditional knockout mice with podocyte-specific ablation of ß-catenin were protected against podocyte injury and albuminuria after treatment with advanced oxidation protein products. The action of Wnt/ß-catenin was dependent on the receptor of advanced glycation end products (RAGE)-mediated NADPH oxidase induction, reactive oxygen species generation, and nuclear factor-κB activation. These studies uncover a novel mechanistic linkage of oxidative stress, Wnt/ß-catenin activation, and podocyte dysfunction.


Assuntos
Produtos da Oxidação Avançada de Proteínas/metabolismo , Podócitos/patologia , Proteinúria/patologia , Insuficiência Renal Crônica/patologia , Via de Sinalização Wnt , Adolescente , Adulto , Produtos da Oxidação Avançada de Proteínas/sangue , Idoso , Animais , Feminino , Glucuronidase/metabolismo , Voluntários Saudáveis , Humanos , Proteínas Klotho , Masculino , Camundongos , Camundongos Knockout , Pessoa de Meia-Idade , NF-kappa B/metabolismo , Estresse Oxidativo , Podócitos/metabolismo , Proteinúria/sangue , Proteinúria/urina , Espécies Reativas de Oxigênio/metabolismo , Receptor para Produtos Finais de Glicação Avançada/metabolismo , Insuficiência Renal Crônica/sangue , Insuficiência Renal Crônica/urina , Regulação para Cima , Proteínas Wnt/metabolismo , Proteína Wnt1/metabolismo , Adulto Jovem , beta Catenina/genética , beta Catenina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...