Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Apoptosis ; 2024 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-38796567

RESUMEN

Podocyte apoptosis or loss is the pivotal pathological characteristic of diabetic kidney disease (DKD). Insulin-like growth factor-binding protein 2 (IGFBP2) have a proinflammatory and proapoptotic effect on diseases. Previous studies have shown that serum IGFBP2 level significantly increased in DKD patients, but the precise mechanisms remain unclear. Here, we found that IGFBP2 levels obviously increased under a diabetic state and high glucose stimuli. Deficiency of IGFBP2 attenuated the urine protein, renal pathological injury and glomeruli hypertrophy of DKD mice induced by STZ, and knockdown or deletion of IGFBP2 alleviated podocytes apoptosis induced by high concentration of glucose or in DKD mouse. Furthermore, IGFBP2 facilitated apoptosis, which was characterized by increase in inflammation and oxidative stress, by binding with integrin α5 (ITGA5) of podocytes, and then activating the phosphorylation of focal adhesion kinase (FAK)-mediated mitochondrial injury, including membrane potential decreasing, ROS production increasing. Moreover, ITGA5 knockdown or FAK inhibition attenuated the podocyte apoptosis caused by high glucose or IGFBP2 overexpression. Taken together, these findings unveiled the insight mechanism that IGFBP2 increased podocyte apoptosis by mitochondrial injury via ITGA5/FAK phosphorylation pathway in DKD progression, and provided the potential therapeutic strategies for diabetic kidney disease.

2.
Am J Pathol ; 193(11): 1845-1862, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37517685

RESUMEN

The transcription factor forkhead box protein (FOX)-O3 is a core regulator of cellular homeostasis, stress response, and longevity. The cellular localization of FOXO3 is closely related to its function. Herein, the role of FOXO3 in cataract formation was explored. FOXO3 showed nuclear translocation in lens epithelial cells (LECs) arranged in a single layer on lens capsule tissues from both human cataract and N-methyl-N-nitrosourea (MNU)-induced rat cataract, also in MNU-injured human (H)-LEC lines. FOXO3 knockdown inhibited the MNU-induced increase in expression of genes related to cell cycle arrest (GADD45A and CCNG2) and apoptosis (BAK and TP53). H2 is highly effective in reducing oxidative impairments in nuclear DNA and mitochondria. When H2 was applied to MNU-injured HLECs, FOXO3 underwent cleavage by MAPK1 and translocated into mitochondria, thereby increasing the transcription of oxidative phosphorylation-related genes (MTCO1, MTCO2, MTND1, and MTND6) in HLECs. Furthermore, H2 mediated the translocation of FOXO3 from the nucleus to the mitochondria within the LECs of cataract capsule tissues of rats exposed to MNU. This intervention ameliorated MNU-induced cataracts in the rat model. In conclusion, there was a correlation between the localization of FOXO3 and its function in cataract formation. It was also determined that H2 protects HLECs from injury by leading FOXO3 mitochondrial translocation via MAPK1 activation. Mitochondrial FOXO3 can increase mtDNA transcription and stabilize mitochondrial function in HLECs.

3.
Gerontology ; 69(5): 628-640, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36720215

RESUMEN

INTRODUCTION: Youthful blood environment was shown to decelerate the aging process of the kidney and to attenuate senile renal fibrosis in a young-old parabiotic animal model; in addition, we identified a stem cell factor (SCF) that is closely linked with the process. This research was to investigate the effect of youthful blood environment on senile renal interstitial fibrosis and the role of SCF. METHODS: We bred SCF receptor c-Kit gene loss-of-function Wps/Wps mice and established a combination mice model that was subjected to unilateral ureteral obstructive (UUO) and parabiotic surgeries. Parabiotic mice were divided into isochronic parabiotic (young-young [Y-IP] and old-old [O-IP]) and heterochronic parabiotic (young-old [HP]) groups. UUO surgery was performed in one of the parabiotic pairs in the IP group (Y-IPuuo and O-IPuuo) and in the elderly mice in the HP group (O-HPuuo). In order to study the role of SCF/c-kit on renal interstitial fibrosis, UUO surgery was performed in wildtype (WT) and Wps/Wps mice. RESULTS: Fourteen days after UUO surgery, the kidney interstitial fibrosis area, kidney function, and the expressions of SCF/c-Kit, pNF-κB, and fibrosis-related proteins in the O-HPuuo group were significantly lower than those in the Ouuo and O-IPuuo groups. Compared with WT UUO mice, the expressions of pNF-κB and fibrosis-related proteins and the kidney function were all significantly decreased in Wps/Wps UUO mice. CONCLUSION: Youthful blood environment downregulated the expressions of SCF/c-Kit in elderly UUO mice and ameliorated UUO-induced kidney fibrosis and function loss.


Asunto(s)
Enfermedades Renales , Obstrucción Ureteral , Ratones , Animales , Factor de Células Madre/genética , Factor de Células Madre/metabolismo , Factor de Células Madre/farmacología , Obstrucción Ureteral/complicaciones , Obstrucción Ureteral/metabolismo , Obstrucción Ureteral/patología , Enfermedades Renales/genética , Enfermedades Renales/metabolismo , Riñón/patología , Fibrosis , Modelos Animales de Enfermedad
4.
Acta Pharmacol Sin ; 43(2): 342-353, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34811512

RESUMEN

Panax notoginseng, a traditional Chinese medicine, exerts beneficial effect on diabetic kidney disease (DKD), but its mechanism is not well clarified. In this study we investigated the effects of ginsenoside Rb1 (Rb1), the main active ingredients of Panax notoginseng, in alleviating podocyte injury in diabetic nephropathy and the underlying mechanisms. In cultured mouse podocyte cells, Rb1 (10 µM) significantly inhibited high glucose-induced cell apoptosis and mitochondrial injury. Furthermore, Rb1 treatment reversed high glucose-induced increases in Cyto c, Caspase 9 and mitochondrial regulatory protein NOX4, but did not affect the upregulated expression of aldose reductase (AR). Molecular docking analysis revealed that Rb1 could combine with AR and inhibited its activity. We compared the effects of Rb1 with eparestat, a known aldose reductase inhibitor, in high glucose-treated podocytes, and found that both alleviated high glucose-induced cell apoptosis and mitochondrial damage, and Rb1 was more effective in inhibiting apoptosis. In AR-overexpressing podocytes, Rb1 (10 µM) inhibited AR-mediated ROS overproduction and protected against high glucose-induced mitochondrial injury. In streptozotocin-induced DKD mice, administration of Rb1 (40 mg·kg-1·d-1, ig, for 7 weeks) significantly mitigated diabetic-induced glomerular injuries, such as glomerular hypertrophy and mesangial matrix expansion, and reduced the expression of apoptotic proteins. Collectively, Rb1 combines with AR to alleviate high glucose-induced podocyte apoptosis and mitochondrial damage, and effectively mitigates the progression of diabetic kidney disease.


Asunto(s)
Aldehído Reductasa/antagonistas & inhibidores , Nefropatías Diabéticas/tratamiento farmacológico , Ginsenósidos/uso terapéutico , Podocitos/efectos de los fármacos , Albuminuria/metabolismo , Animales , Apoptosis/efectos de los fármacos , Glucemia/análisis , Western Blotting , Células Cultivadas , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/tratamiento farmacológico , Nefropatías Diabéticas/enzimología , Nefropatías Diabéticas/patología , Citometría de Flujo , Riñón/efectos de los fármacos , Riñón/patología , Masculino , Ratones , Simulación del Acoplamiento Molecular , Podocitos/enzimología
5.
Mediators Inflamm ; 2022: 5515305, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35399795

RESUMEN

Activation of NOD-like receptor (NLR) signaling pathway can promote downstream cytokine and proinflammatory cytokines release, and inflammation induced by excess nutrients leads to renal metabolic injury. How the NLRs influence metabolic progress and then lead to the renal injury remains poorly investigated. Compared with rodents, minipigs are more similar to humans and are more ideal animal models for human disease research. In this study, we established a diabetic minipig model through a high-sugar and high-fat diet combined with streptozotocin (STZ) injection. Blood biological markers and renal pathological markers, expression of NLRP subfamily members (NLRP1 and NLRP3) and their downstream cytokines (precursors of IL-1ß and IL-18 and mature forms of IL-1ß and IL-18), expression of NLRC subfamily members (NLRC1, NLRC2, and NLRC5) and their downstream nuclear factor-κB (NF-κB) signaling pathway molecules (IKKß, IκBα, and NF-κB p65), and inflammatory cytokines (TNF-α and interleukin-6 (IL-6)) were systematically evaluated. The expression of NLRP3 and its downstream cytokine signaling molecules, the precursors of IL-1ß and IL-18, and the mature forms of IL-1ß and IL-18 was significantly upregulated. The expression levels of NLRC1, NLRC2, and NLRC5 and activation of the downstream NF-κB pathway molecules phospho-IKKß, phospho-IκBα, NF-κB p65, and phospho-NF-κB p65 were significantly increased. The TNF-α and IL-6 levels were significantly increased in diabetic pig kidneys. The TGF-ß/Smad signaling molecules, TGF-ß and P-SMAD2/3, were also increased. These results suggested that the metabolic inflammation activated by NLRs might play an important role in diabetic renal injuries.


Asunto(s)
Diabetes Mellitus , FN-kappa B , Animales , Citocinas/metabolismo , Quinasa I-kappa B , Inflamación , Interleucina-18 , Interleucina-6/metabolismo , Riñón/metabolismo , Inhibidor NF-kappaB alfa , FN-kappa B/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Proteínas NLR , Porcinos , Porcinos Enanos/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Factor de Necrosis Tumoral alfa
6.
Dev Biol ; 464(2): 103-110, 2020 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-32562756

RESUMEN

Congenital anomalies of the urinary tract are a significant cause of morbidity in infancy, and many congenital anomalies are linked to ureter development; however, the mechanism by which congenital anomalies control ureter development remains unknown. The loss of Robo2 can cause ureter defects and vesicoureteral reflux. However, how Robo2 impacts ureter development is unclear. We found that ROBO2 is expressed in the common nephric duct (CND) and primitive bladder, and impacts CND migration and fusion with the primitive bladder via its novel binding partner retinaldehyde dehydrogenase-2 (RALDH2). Delayed apoptosis that is due to the failure of CND fusion with the primitive bladder in the Robo2-/-embryo results in an abnormal ureter connection to the CND, which is required for ureter development. We define a novel pathway in which the CND is remodeled by ROBO2 and retinoic acid rescued the ureter anomalies in the Robo2-/-embryo. These findings may be relevant to diverse disease conditions that are associated with altered signaling in the primitive bladder.


Asunto(s)
Aldehído Oxidorreductasas/metabolismo , Receptores Inmunológicos/metabolismo , Transducción de Señal , Uréter/embriología , Vejiga Urinaria/embriología , Aldehído Oxidorreductasas/genética , Animales , Ratones , Ratones Noqueados , Receptores Inmunológicos/genética , Uréter/citología , Vejiga Urinaria/citología
7.
J Immunol ; 203(10): 2644-2654, 2019 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-31591147

RESUMEN

Previous studies of Jak-STAT inhibitors have shown promise in treating kidney diseases. The activation of Jak-STAT components is important in cell fate determination in many cell types, including bone marrow-derived cells, which are important contributors in renal interstitial fibrosis. In this study, we tested the effect of a new STAT3 inhibitor, BP-1-102, on monocyte-to-fibrocyte transition and the progression of renal interstitial fibrosis. We tested the effect of BP-1-102 in a mouse model of unilateral ureteral obstruction in vivo and IL-33-treated bone marrow-derived monocytes in vitro. BP-1-102 treatment alleviated renal interstitial fibrosis, reduced collagen deposition and extracellular matrix protein production, inhibited inflammatory cell infiltration, suppressed the percentage of CD45+ PDGFRß+, CD45+ CD34- Col I+ and CD45+ CD11b+ Col I+ cells within the obstructed kidney and reduced the mRNA levels of the proinflammatory and profibrotic cytokines IL-1ß, TGF-ß, TNF-α, ICAM-1, and CXCL16. In vitro, BP-1-102 inhibited the IL-33-induced phenotypic transition into fibroblast precursors in bone marrow-derived monocytes, marked by reduced CD45+ CD34- Col I+ and CD45+ CD11b+ Col I+ cell percentage. Our results indicate a potential mechanism by which the STAT3 inhibitor BP-1-102 inhibits bone marrow-derived monocyte transition into fibroblast precursors in an IL-33/STAT3-dependent manner and thereby alleviates renal interstitial fibrosis.


Asunto(s)
Adaptación Fisiológica/efectos de los fármacos , Ácidos Aminosalicílicos/farmacología , Médula Ósea/metabolismo , Fibroblastos/metabolismo , Interleucina-33/farmacología , Riñón/patología , Monocitos/metabolismo , Factor de Transcripción STAT3/antagonistas & inhibidores , Sulfonamidas/farmacología , Obstrucción Ureteral/metabolismo , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Fibrosis , Interleucina-33/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL
8.
Biochem Biophys Res Commun ; 522(2): 322-327, 2020 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-31761321

RESUMEN

BACKGROUND: Hepcidin is a key regulator of iron homeostasis. Some studies showed that exogenous hepcidin decreased the expression of divalent metal transporter (DMT1) rather than ferroportin(FPN1) to regulate renal iron metabolism. This study explored the effects of hepcidin synthesized by the kidney and its mechanism of iron regulation. METHODS: In the in vivo experiments, mice were divided into a unilateral ureter obstruction (UUO) model group and a sham operation group, and mice in the UUO model group were sacrificed on days 1, 3, 5 and 7. The expression of renal hepcidin, FPN1, DMT1 and the retention of renal iron were studied. In the in vitro experiments, we overexpressed hepcidin in HK-2 cells. Then we tested the expression of renal hepcidin, FPN1, DMT1 and observed the production of intracellular ferrous ions. RESULTS: Renal hepcidin expression was consistently higher in the UUO group than in the sham group from the first day. The expression of FPN1 gradually decreased, and the expression of DMT1 gradually increased in the UUO model. Intracellular ferrous ions significantly increased on the first day of the UUO model. In hepcidin overexpressed HK-2 cells, the expression of FPN1 was decreased, while the expression of DMT1 has no significant change. In addition, production of intracellular ferrous ions increased. CONCLUSION: local hepcidin can regulate iron metabolism in the kidney by adjusting the expression of FPN1.


Asunto(s)
Proteínas de Transporte de Catión/metabolismo , Hepcidinas/metabolismo , Espacio Intracelular/metabolismo , Sobrecarga de Hierro/metabolismo , Riñón/metabolismo , Animales , Ferritinas/metabolismo , Hierro/metabolismo , Masculino , Ratones Endogámicos C57BL
9.
Mediators Inflamm ; 2020: 8387654, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32565731

RESUMEN

Renin angiotensin (Ang) system (RAS) activation in metabolic syndrome (MS) patients is associated with elevated uric acid (UA) levels, resulting in endothelial system dysfunction. Our previous study demonstrated that excessive UA could cause endothelial injury through the aldose reductase (AR) pathway. This study is the first to show that a high concentration of Ang II in human umbilical vein endothelial cells (HUVECs) increases reactive oxygen species (ROS) components, including O2 ·- and H2O2, and further aggravates endothelial system injury induced by high UA (HUA). In a MS/hyperuricemia model, nitric oxide (NO) production was decreased, followed by a decrease in total antioxidant capacity (TAC), and the concentration of the endothelial injury marker von Willebrand factor (vWF) in the serum was increased. Treatment with catalase and polyethylene glycol covalently linked to superoxide dismutase (PEG-SOD) to individually remove H2O2 and O2 ·- or treatment with the AR inhibitor epalrestat decreased ROS and H2O2, increased NO levels and TAC, and reduced vWF release. Taken together, these data indicate that HUA and Ang II act additively to cause endothelial dysfunction via oxidative stress, and specific elimination of O2 ·- and H2O2 improves endothelial function. We provide theoretical evidence to prevent or delay endothelial injury caused by metabolic diseases.


Asunto(s)
Angiotensina II/metabolismo , Endotelio Vascular/metabolismo , Ácido Úrico/metabolismo , Alimentación Animal , Animales , Endotelio Vascular/patología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Peróxido de Hidrógeno/química , Hiperuricemia/metabolismo , Masculino , NADPH Oxidasas/metabolismo , Óxido Nítrico/metabolismo , Estrés Oxidativo/efectos de los fármacos , Oxígeno/química , Ratas , Ratas Endogámicas SHR , Especies Reactivas de Oxígeno , Superóxido Dismutasa/metabolismo , Factor de von Willebrand/metabolismo
10.
Cell Physiol Biochem ; 42(5): 2118-2129, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28810249

RESUMEN

BACKGROUND/AIMS: IFN-γ-inducible protein 10 (IP-10, CXCL10) has been widely demonstrated to be involved in chemotaxis, cell growth regulation and angiogenesis inhibition. It has been reported that CXCL10 expression is significantly increased in patients with MesPGN (Mesangial proliferative glomerulonephritis). However, the underlying mechanism of CXCL10 in MesPGN reminds unclear. METHODS: Wildtype (Cxcl10+/+) mice and Cxcl10-deficient (Cxcl10-/-) mice were used to generate a murine model of MesPGN. The histological changes in glomeruli were examined by PAS staining (Periodic Acid-Schiff staining), and cell proliferation was detected by PCNA immunohistochemistry staining. The expression of cell cycle regulatory proteins was analyzed by Western blotting and the effects of CXCL10 on primary mouse renal mesangial cells (MRMC) proliferation were detected using the EDU assay. Furthermore, the specific mechanisms by which CXCL10 affected mesangial cells were investigated in vitro using a specific inhibitor. RESULTS: Typical pathological phenotypes were observed in both mouse types, while the Cxcl10-/- mice had lighter accumulation of extracellular matrix, less cell proliferation and diminished up-regulation of cell cycle regulatory proteins compared to Cxcl10+/+ mice at day 7. Furthermore, we observed that CXCL10 inhibition resulted in less activation of ERK phosphorylation, and ERK pathway inhibition by a specific inhibitor, U0126, prevented CXCL10 induced MRMC proliferation and the activation of phosphorylated ERK. CONCLUSIONS: CXCL10 may aggravate mesangial proliferation in MesPGN by activating the ERK signaling pathway. These results provide a novel insight into the mechanism and potential therapy target of MesPGN.


Asunto(s)
Quimiocina CXCL10/metabolismo , Glomerulonefritis Membranoproliferativa/patología , Animales , Proteínas de Ciclo Celular/metabolismo , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Quimiocina CXCL10/genética , Quimiocina CXCL10/farmacología , Modelos Animales de Enfermedad , Glomerulonefritis Membranoproliferativa/metabolismo , Inmunohistoquímica , Riñón/citología , Riñón/patología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Células Mesangiales/citología , Células Mesangiales/efectos de los fármacos , Células Mesangiales/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Fluorescente , Fosforilación/efectos de los fármacos , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/farmacología , Venenos de Serpiente/toxicidad , Regulación hacia Arriba/efectos de los fármacos
11.
Biochem Biophys Res Commun ; 489(3): 319-325, 2017 07 29.
Artículo en Inglés | MEDLINE | ID: mdl-28554841

RESUMEN

Periodontal ligament stem cells (PDLSCs) are promising tools for the investigations of cell differentiation and bone regeneration. However, the limited life span significantly restricts their usefulness. In this study, we established an immortalized PDLSC cell line by the introduction of Bmi1 (PDLSC-Bmi1). Several genes related to cell cycle, cell replication and stemness were found to be changed with the overexpression of Bmi1. Compared with primary PDLSCs, the immortalized cells had a slower aging rate, maintained in a proliferative state without crisis for more than 30 passages, and retained the molecular markers and biological functions of primary ones. Using the PDLSC-Bmi1, we confirmed the promotive effect of naringin on osteogenesis. Naringin promoted the osteogenic differentiation of PDLSC-Bmi1 manifested as the increased activity of alkaline phosphatase (ALP), expression of the runt-related transcription factor 2 (Runx2) and osteocalcin (OCN), and formation of mineralized nodules. In addition, the extracellular regulated protein kinases (ERK) 1/2 was found to be activated by naringin, and the ERK1/2 specific inhibitor significantly inhibited naringin-induced osteogenic differentiation in PDLSC-Bmi1. Our results indicated that the overexpression of Bmi1 extended the life span of PDLSCs without perturbing their biological functions, and that naringin promoted the osteogenesis of PDLSC-Bmi1 at least partially through the ERK1/2 signaling pathway.


Asunto(s)
Flavanonas/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Ligamento Periodontal/citología , Adolescente , Adulto , Diferenciación Celular/efectos de los fármacos , Femenino , Flavanonas/química , Células HEK293 , Humanos , Masculino , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Adulto Joven
12.
Cell Commun Signal ; 15(1): 3, 2017 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-28057038

RESUMEN

BACKGROUND: Uric acid (UA) is an antioxidant found in human serum. However, high UA levels may also have pro-oxidant functions. According to previous research, aldose reductase (AR) plays a vital role in the oxidative stress-related complications of diabetes. We sought to determine the mechanism by which UA becomes deleterious at high concentrations as well as the effect of AR in this process. METHOD: Endothelial cells were divided into three groups cultured without UA or with 300 µM or 600 µM UA. The levels of total reactive oxygen species (ROS), of four ROS components, and of NO and NOX4 expression were measured. Changes in the above molecules were detected upon inhibiting NOX4 or AR, and serum H2O2 and vWF levels were measured in vivo. RESULTS: Increased AR expression in high UA-treated endothelial cells enhanced ROS production by activating NADPH oxidase. These effects were blocked by the AR inhibitor epalrestat. 300 µM UA decreased the levels of the three major reactive oxygen species (ROS) components: O2•-, •OH, and 1O2. However, when the UA concentration was increased, both O2•- levels and downstream H2O2 production significantly increased. Finally, an AR inhibitor reduced H2O2 production in hyperuricemic mice and protected endothelial cell function. CONCLUSIONS: Our findings indicate that inhibiting AR or degrading H2O2 could protect endothelial function and maintain the antioxidant activities of UA. These findings provide new insight into the role of UA in chronic kidney disease.


Asunto(s)
Aldehído Reductasa/metabolismo , Células Endoteliales de la Vena Umbilical Humana/enzimología , Células Endoteliales de la Vena Umbilical Humana/patología , Ácido Úrico/farmacología , Aldehído Reductasa/antagonistas & inhibidores , Animales , Catalasa/farmacología , Citoprotección/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Peróxido de Hidrógeno/metabolismo , Hiperuricemia/metabolismo , Ratones , Modelos Biológicos , NADPH Oxidasas/metabolismo , Óxido Nítrico/metabolismo , Estrés Oxidativo/efectos de los fármacos , Polietilenglicoles/farmacología , Rodanina/análogos & derivados , Rodanina/farmacología , Tiazolidinas/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
13.
Proteomics ; 14(21-22): 2485-97, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25103794

RESUMEN

The apoptosis of mesangial cells (MCs) plays a critical role in the pathological progress of MesPGN. Septin2, a filamentous GTPase, is implicated in the apoptotic progress of MCs in the rat MesPGN model. However, the molecular mechanism of SEPT2 in MCs apoptosis is not clear. Here, we present the FHL2-driven molecular network as the main mechanism of SEPT2-mediated rat primary MCs apoptosis. First, we proved that the expression of FHL2 and Septin2 were closely related with MCs apoptosis in anti-Thy1 nephritis model. Then, it was found that FHL2 was a new interaction protein of Septin2 and Septin2 knockdown could induce MC apoptosis by FHL2-mediatied signal pathways including p-ERK1 and p-AKT. We applied label-Free quantitative proteomics to identify the mechanism of Septin2/FHL2-regulated apoptosis. Bioinformatics analysis revealed that FHL2-driven molecular network composed of biological functions including glycolysis, oxidative stress, ribonucleotide metabolism, actin cytoskeleton regulation, and signaling pathway, was the main mechanism of SETP2-mediated apoptosis. Furthermore, we showed that the effect of Septin2 knockdown on MC apoptosis could be alleviated by the overexpression of FHL2. Overall, this study illustrated the FHL2-driven molecular network controlling SEPT2-mediated apoptosis in MCs and their potential roles in mesangial proliferative nephritis.


Asunto(s)
Proteínas con Homeodominio LIM/metabolismo , Células Mesangiales/citología , Proteínas Musculares/metabolismo , Factores de Crecimiento Nervioso/metabolismo , Septinas/metabolismo , Factores de Transcripción/metabolismo , Animales , Apoptosis , Células Cultivadas , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Proteínas con Homeodominio LIM/genética , Células Mesangiales/metabolismo , Proteínas Musculares/genética , Factores de Crecimiento Nervioso/genética , Mapeo de Interacción de Proteínas , Ratas , Septinas/genética , Transducción de Señal , Factores de Transcripción/genética
14.
Lab Invest ; 94(4): 439-54, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24566932

RESUMEN

Podocyte injury has a critical role in the pathogenesis of proteinuria. Induction of endoplasmic reticulum (ER) stress is thought to lead to podocyte injury; however, no effective strategy for reducing ER stress-induced injury has been identified. We investigated specific mechanisms for reducing podocyte injury caused by ER stress. We found that the induction of ER stress in podocytes was related to cytoskeleton injury and increased proteinuria, which was associated with autophagy activation and downregulation of Na(+)/H(+) exchanger-1 (NHE-1) in the rat model of passive Heymann nephritis. Using mouse podocyte cells (MPCs), we showed that ER stress could lead to podocyte injury accompanied by autophagy activation, and the disturbance of autophagy aggravated cytoskeleton loss under conditions of ER stress. The balance between autophagy activation and ER stress was critical to podocyte survival, in which the efficiency of autophagy could have a pivotal role. Strikingly, the overexpression and small interfering RNA knockdown of NHE-1 results suggested that NHE-1 exerts a protective effect by reducing the loss of synaptopodin in MPCs exposed to ER stress. This protective mechanism involves NHE-1 activation of autophagy via the PI3K/Akt pathway to reduce ER stress injury in podocytes. This mechanism may provide a new pathway to prevent podocyte injury.


Asunto(s)
Autofagia , Estrés del Retículo Endoplásmico , Glomerulonefritis Membranosa/metabolismo , Podocitos/fisiología , Intercambiadores de Sodio-Hidrógeno/metabolismo , Animales , Células Cultivadas , Citoesqueleto/fisiología , Modelos Animales de Enfermedad , Glomerulonefritis Membranosa/fisiopatología , Masculino , Ratones , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Sprague-Dawley , Intercambiador 1 de Sodio-Hidrógeno
15.
BMC Complement Med Ther ; 24(1): 80, 2024 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-38331805

RESUMEN

BACKGROUND: Astragalus polysaccharides (APS) have been verified to have antioxidative and antiaging activities in the mouse liver and brain. However, the effect of APS on aortic endothelial senescence in old rats and its underlying mechanism are currently unclear. Here, we aimed to elucidate the effects of APS on rat aortic endothelial oxidative stress and senescence in vitro and in vivo and investigate the potential molecular targets. METHODS: Twenty-month-old natural aging male rats were treated with APS (200 mg/kg, 400 mg/kg, 800 mg/kg daily) for 3 months. Serum parameters were tested using corresponding assay kits. Aortic morphology was observed by staining with hematoxylin and eosin (H&E) and Verhoeff Van Gieson (VVG). Aging-related protein levels were evaluated using immunofluorescence and western blot analysis. Primary rat aortic endothelial cells (RAECs) were isolated by tissue explant method. RAEC mitochondrial function was evaluated by the mitochondrial membrane potential (MMP) measured with the fluorescent lipophilic cationic dye JC­1. Intracellular total antioxidant capacity (T-AOC) was detected by a commercial kit. Cellular senescence was assessed using senescence-associated-ß-galactosidase (SA-ß-Gal) staining. RESULTS: Treatment of APS for three months was found to lessen aortic wall thickness, renovate vascular elastic tissue, improve vascular endothelial function, and reduce oxidative stress levels in 20-month-old rats. Primary mechanism analysis showed that APS treatment enhanced Sirtuin 1 (SIRT-1) protein expression and decreased the levels of the aging marker proteins p53, p21 and p16 in rat aortic tissue. Furthermore, APS abated hydrogen peroxide (H2O2)-induced cell senescence and restored H2O2-induced impairment of the MMP and T-AOC in RAECs. Similarly, APS increased SIRT-1 and decreased p53, p21 and p16 protein levels in senescent RAECs isolated from old rats. Knockdown of SIRT-1 diminished the protective effect of APS against H2O2-induced RAEC senescence and T-AOC loss, increased the levels of the downstream proteins p53 and p21, and abolished the inhibitory effect of APS on the expression of these proteins in RAECs. CONCLUSION: APS may reduce rat aortic endothelial oxidative stress and senescence via the SIRT-1/p53 signaling pathway.


Asunto(s)
Células Endoteliales , Sirtuina 1 , Ratones , Masculino , Ratas , Animales , Células Endoteliales/metabolismo , Sirtuina 1/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Peróxido de Hidrógeno/farmacología , Senescencia Celular/fisiología , Antioxidantes/farmacología , Antioxidantes/metabolismo , Transducción de Señal , Polisacáridos/farmacología , Polisacáridos/metabolismo
16.
Adv Mater ; : e2311845, 2024 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-38720198

RESUMEN

Sweat gland (SwG) regeneration is crucial for the functional rehabilitation of burn patients. In vivo chemical reprogramming that harnessing the patient's own cells in damaged tissue is of substantial interest to regenerate organs endogenously by pharmacological manipulation, which could compensate for tissue loss in devastating diseases and injuries, for example, burns. However, achieving in vivo chemical reprogramming is challenging due to the low reprogramming efficiency and an unfavorable tissue environment. Herein, this work has developed a functionalized proteinaceous nanoformulation delivery system containing prefabricated epidermal growth factor structure for on-demand delivery of a cocktail of seven SwG reprogramming components to the dermal site. Such a chemical reprogramming system can efficiently induce the conversion of epidermal keratinocytes into SwG myoepithelial cells, resulting in successful in situ regeneration of functional SwGs. Notably, in vivo chemical reprogramming of SwGs is achieved for the first time with an impressive efficiency of 30.6%, surpassing previously reported efficiencies. Overall, this proteinaceous nanoformulation provides a platform for coordinating the target delivery of multiple pharmacological agents and facilitating in vivo SwG reprogramming by chemicals. This advancement greatly improves the clinical accessibility of in vivo reprogramming and offers a non-surgical, non-viral, and cell-free strategy for in situ SwG regeneration.

17.
Kidney Int ; 84(3): 521-31, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23615497

RESUMEN

The mechanism of mesenchymal stem cell therapy in acute kidney injury remains uncertain. Previous studies indicated that mesenchymal stem cells could attenuate inflammation-related organ injury by induction of regulatory T cells. Whether regulatory T-cell induction is a potential mechanism of mesenchymal stem cell therapy in ischemic acute kidney injury and how these induced regulatory T cells orchestrate local inflammation are unknown. Here we found that mesenchymal stem cells decrease serum creatinine and urea nitrogen levels, improve tubular injury, and downregulate IFN-γ production of T cells in the ischemic kidney. In addition to the lung, mesenchymal stem cells persisted mostly in the spleen. Mesenchymal stem cells increased the percentage of regulatory T cells in the spleen and the ischemic kidney. Antibody-dependent depletion of regulatory T cells blunted the therapeutic effect of mesenchymal stem cells, while coculture of splenocytes with mesenchymal stem cells caused an increase in the percentage of regulatory T cells. Splenectomy abrogated attenuation of ischemic injury, and downregulated IFN-γ production and the induction of regulatory T cells by mesenchymal stem cells. Thus, mesenchymal stem cells ameliorate ischemic acute kidney injury by inducing regulatory T cells through interactions with splenocytes. Accumulated regulatory T cells in ischemic kidney might be involved in the downregulation of IFN-γ production.


Asunto(s)
Lesión Renal Aguda/patología , Lesión Renal Aguda/terapia , Comunicación Celular/fisiología , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/patología , Bazo/patología , Linfocitos T Reguladores/patología , Lesión Renal Aguda/fisiopatología , Animales , Nitrógeno de la Urea Sanguínea , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Creatinina/sangre , Modelos Animales de Enfermedad , Interferón gamma/metabolismo , Riñón/patología , Ratones , Ratones Endogámicos C57BL , Bazo/cirugía , Esplenectomía
18.
Nephron Exp Nephrol ; 124(3-4): 19-27, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24401898

RESUMEN

BACKGROUND: The therapeutic use of the antineoplastic drug cisplatin (DDP) in the elderly is limited by its nephrotoxic effects. The aim of this study was to examine the effect of short-term calorie restriction (CR) on DDP-induced nephrotoxicity in aged rats. METHODS: A group of 25-month-old male Sprague-Dawley rats were divided into two groups: ad libitum (AL) and CR, which were fed 60% of the food consumed by AL rats for 8 weeks. The two groups were each further randomly divided into two subgroups: OAL control, OAL+DDP, OCR control, and OCR+DDP. A single dose of DDP (6 mg/kg) was injected intraperitoneally. Functional and structural changes of the kidneys were evaluated quantitatively by biochemical, histopathological, and morphometric analyses. RESULTS: At the end of the 8 weeks, rats in the OCR group lost 14.8% more body mass than rats in the OAL group. Pretreatment with CR had several effects: (1) it reduced the levels of blood urea nitrogen and serum creatinine, (2) it reduced the magnitude of the renal tubular epithelial damage, and (3) it significantly reduced the incidence of activated caspase-3 and TUNEL-positive cells in kidneys injured by DDP. However, SIRT1 had the opposite trend after DDP application between the two groups. CONCLUSIONS: Short-term CR exhibits a renoprotective effect in experimental DDP-induced renal injury, the mechanism of which may involve CR antiapoptotic effects and promotion of SIRT1.


Asunto(s)
Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/prevención & control , Restricción Calórica , Cisplatino/efectos adversos , Lesión Renal Aguda/metabolismo , Envejecimiento/metabolismo , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Caspasa 3/metabolismo , Cisplatino/farmacología , Creatinina/metabolismo , Modelos Animales de Enfermedad , Riñón/efectos de los fármacos , Riñón/metabolismo , Riñón/patología , Masculino , Ratas , Ratas Sprague-Dawley , Sirtuina 1/metabolismo , Resultado del Tratamiento
19.
Metabolism ; 145: 155592, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37230215

RESUMEN

BACKGROUND AND AIMS: Acute kidney injury (AKI) is associated with high morbidity and mortality and is recognized as a long-term risk factor for progression to chronic kidney disease (CKD). The AKI to CKD transition is characterized by interstitial fibrosis and the proliferation of collagen-secreting myofibroblasts. Pericytes are the major source of myofibroblasts in kidney fibrosis. However, the underlying mechanism of pericyte-myofibroblast transition (PMT) is still unclear. Here we investigated the role of metabolic reprogramming in PMT. METHODS: Unilateral ischemia/reperfusion-induced AKI to CKD mouse model and TGF-ß-treated pericyte-like cells were used to detect the levels of fatty acid oxidation (FAO) and glycolysis, and the critical signaling pathways during PMT under the treatment of drugs regulating metabolic reprogramming. RESULTS: PMT is characterized by a decrease in FAO and an increase in glycolysis. Enhancement of FAO by the peroxisome proliferator-activated receptor gamma coactivator-1α (PGC1α) activator ZLN-005 or suppression of glycolysis by the hexokinase 2 (HK2) inhibitor 2-DG can inhibit PMT, preventing the transition of AKI to CKD. Mechanistically, AMPK modulates various pathways involved in the metabolic switch from glycolysis to FAO. Specifically, the PGC1α-CPT1A pathway activates FAO, while inhibition of the HIF1α-HK2 pathway drives glycolysis inhibition. The modulations of these pathways by AMPK contribute to inhibiting PMT. CONCLUSIONS: Metabolic reprogramming controls the fate of pericyte transdifferentiation and targets the abnormal metabolism of pericytes can effectively prevent AKI to CKD transition.


Asunto(s)
Lesión Renal Aguda , Insuficiencia Renal Crónica , Ratones , Animales , Pericitos , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Insuficiencia Renal Crónica/etiología , Lesión Renal Aguda/patología , Fibrosis , Riñón
20.
Biochem Biophys Res Commun ; 421(3): 494-500, 2012 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-22521888

RESUMEN

Roundabout 2 (Robo2) is a member of the membrane protein receptor family. The chemorepulsive effect of Slit2-Robo2 signaling plays vital roles in nervous system development and neuron migration. Slit2-Robo2 signaling is also important for maintaining the normal morphogenesis of the kidney and urinary collecting system, especially for the branching of the ureteric bud (UB) at the proper site. Slit2 or Robo2 mouse mutants exhibit multilobular kidneys, multiple ureters, and dilatation of the ureter, renal pelvis, and collecting duct system, which lead to vesicoureteral reflux. To understand the effect of Robo2 on kidney development, we used microinjection and electroporation to overexpress GFP-Robo2 in an in vitro embryonic kidney model. Our results show reduced UB branching and decreased glomerular number after in vitro Robo2 overexpression in the embryonic kidneys. We found fewer metanephric mesenchymal (MM) cells surrounding the UB but no abnormal morphology in the branching epithelial UB. Meanwhile, no significant change in MM proliferation or apoptosis was observed. These findings indicate that Robo2 is involved in the development of embryonic kidneys and that the normal expression of Robo2 can help maintain proper UB branching and glomerular morphogenesis. Overexpression of Robo2 leads to reduced UB branching caused by fewer surrounding MM cells, but MM cell apoptosis is not involved in this effect. Our study demonstrates that overexpression of Robo2 by microinjection in embryonic kidneys is an effective approach to study the function of Robo2.


Asunto(s)
Mesodermo/citología , Mesodermo/embriología , Morfogénesis , Receptores Inmunológicos/biosíntesis , Uréter/embriología , Animales , Apoptosis , Cadherinas/metabolismo , Proliferación Celular , Electroporación , Riñón/embriología , Glomérulos Renales/embriología , Ratones , Ratones Endogámicos C57BL , Microinyecciones , Receptores Inmunológicos/genética , Uréter/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA