Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Transl Med ; 22(1): 26, 2024 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-38183100

RESUMEN

BACKGROUND: (Pro)renin receptor (PRR) is highly expressed in renal tubules, which is involved in physiological and pathological processes. However, the role of PRR, expressed in renal tubular epithelial cells, in diabetic kidney disease (DKD) remain largely unknown. METHODS: In this study, kidney biopsies, urine samples, and public RNA-seq data from DKD patients were used to assess PRR expression and cell pyroptosis in tubular epithelial cells. The regulation of tubular epithelial cell pyroptosis by PRR was investigated by in situ renal injection of adeno-associated virus9 (AAV9)-shRNA into db/db mice, and knockdown or overexpression of PRR in HK-2 cells. To reveal the underlined mechanism, the interaction of PRR with potential binding proteins was explored by using BioGrid database. Furthermore, the direct binding of PRR to dipeptidyl peptidase 4 (DPP4), a pleiotropic serine peptidase which increases blood glucose by degrading incretins under diabetic conditions, was confirmed by co-immunoprecipitation assay and immunostaining. RESULTS: Higher expression of PRR was found in renal tubules and positively correlated with kidney injuries of DKD patients, in parallel with tubular epithelial cells pyroptosis. Knockdown of PRR in kidneys significantly blunted db/db mice to kidney injury by alleviating renal tubular epithelial cells pyroptosis and the resultant interstitial inflammation. Moreover, silencing of PRR blocked high glucose-induced HK-2 pyroptosis, whereas overexpression of PRR enhanced pyroptotic cell death of HK-2 cells. Mechanistically, PRR selectively bound to cysteine-enrich region of C-terminal of DPP4 and augmented the protein abundance of DPP4, leading to the downstream activation of JNK signaling and suppression of SIRT3 signaling and FGFR1 signaling, and then subsequently mediated pyroptotic cell death. CONCLUSIONS: This study identified the significant role of PRR in the pathogenesis of DKD; specifically, PRR promoted tubular epithelial cell pyroptosis via DPP4 mediated signaling, highlighting that PRR could be a promising therapeutic target in DKD.


Asunto(s)
Nefropatías Diabéticas , Receptor de Prorenina , Animales , Humanos , Ratones , Diabetes Mellitus , Nefropatías Diabéticas/metabolismo , Dipeptidil Peptidasa 4 , Células Epiteliales , Sistema de Señalización de MAP Quinasas , Receptor de Prorenina/metabolismo , Piroptosis
2.
Am J Obstet Gynecol ; 230(2): 254.e1-254.e13, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37531989

RESUMEN

BACKGROUND: Hyperglycemia from pregestational diabetes mellitus induces neural tube defects in the developing fetus. Folate supplementation is the only effective way to prevent neural tube defects; however, some cases of neural tube defects are resistant to folate. Excess folate has been linked to higher maternal cancer risk and infant allergy. Therefore, additional interventions are needed. Understanding the mechanisms underlying maternal diabetes mellitus-induced neural tube defects can identify potential targets for preventing such defects. Despite not yet being in clinical use, growing evidence suggests that microRNAs are important intermediates in embryonic development and can serve as both biomarkers and drug targets for disease intervention. Our previous studies showed that maternal diabetes mellitus in vivo activates the inositol-requiring transmembrane kinase/endoribonuclease 1α (IRE1α) in the developing embryo and that a high glucose condition in vitro reduces microRNA-322 (miR-322) levels. IRE1α is an RNA endonuclease; however, it is unknown whether IRE1α targets and degrades miR-322 specifically or whether miR-322 degradation leads to neural tube defects via apoptosis. We hypothesize that IRE1α can inhibit miR-322 in maternal diabetes mellitus-induced neural tube defects and that restoring miR-322 expression in developing neuroepithelium ameliorates neural tube defects. OBJECTIVE: This study aimed to identify potential targets for preventing maternal diabetes mellitus-induced neural tube defects and to investigate the roles and relationship of a microRNA and an RNA endonuclease in mouse embryos exposed to maternal diabetes mellitus. STUDY DESIGN: To determine whether miR-322 reduction is necessary for neural tube defect formation in pregnancies complicated by diabetes mellitus, male mice carrying a transgene expressing miR-322 were mated with nondiabetic or diabetic wide-type female mice to generate embryos with or without miR-322 overexpression. At embryonic day 8.5 when the neural tube is not yet closed, embryos were harvested for the assessment of 3 miR-322 transcripts (primary, precursor, and mature miR-322), tumor necrosis factor receptor-associated factor 3 (TRAF3), and neuroepithelium cell survival. Neural tube defect incidences were determined in embryonic day 10.5 embryos when the neural tube should be closed if there is no neural tube defect formation. To identify which miR-322 transcript is affected by maternal diabetes mellitus and high glucose conditions, 3 miR-322 transcripts were assessed in embryos from dams with or without diabetes mellitus and in C17.2 mouse neural stem cells treated with different concentrations of glucose and at different time points. To determine whether the endonuclease IRE1α targets miR-322, small interfering RNA knockdown of IRE1α or overexpression of inositol-requiring transmembrane kinase/endoribonuclease 1α by DNA plasmid transfection was used to determine the effect of IRE1α deficiency or overexpression on miR-322 expression. RNA immunoprecipitation was performed to reveal the direct targets of inositol-requiring transmembrane kinase/endoribonuclease 1α. RESULTS: Maternal diabetes mellitus suppressed miR-322 expression in the developing neuroepithelium. Restoring miR-322 expression in the neuroepithelium blocked maternal diabetes mellitus-induced caspase-3 and caspase-8 cleavage and cell apoptosis, leading to a neural tube defect reduction. Reversal of maternal diabetes mellitus-inhibited miR-322 via transgenic overexpression prevented TRAF3 up-regulation in embryos exposed to maternal diabetes mellitus. Activated IRE1α acted as an endonuclease and degraded precursor miR-322, resulting in mature miR-322 reduction. CONCLUSION: This study supports the crucial role of the IRE1α-microRNA-TRAF3 circuit in the induction of neuroepithelial cell apoptosis and neural tube defect formation in pregnancies complicated by diabetes mellitus and identifies IRE1α and miR-322 as potential targets for preventing maternal diabetes mellitus-induced neural tube defects.


Asunto(s)
Diabetes Mellitus Experimental , Diabetes Gestacional , MicroARNs , Defectos del Tubo Neural , Embarazo en Diabéticas , Humanos , Embarazo , Masculino , Femenino , Ratones , Animales , MicroARNs/genética , MicroARNs/metabolismo , Factor 3 Asociado a Receptor de TNF/metabolismo , Endorribonucleasas/genética , Endorribonucleasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Defectos del Tubo Neural/genética , Defectos del Tubo Neural/patología , Embarazo en Diabéticas/genética , Embarazo en Diabéticas/metabolismo , Diabetes Gestacional/genética , Glucosa , Ácido Fólico , Inositol
3.
Int J Mol Sci ; 24(24)2023 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-38139429

RESUMEN

Diabetes nephropathy (DN) is the leading cause of end-stage renal disease (ESRD) worldwide, and podocyte injury is the central contributor to the progression of DN. Despite the emerging evidence that has established the importance of podocyte endoplasmic reticulum (ER) stress in the pathogenesis of DN, abnormal protein O-GlcNAcylation is also augmented. Currently, the mechanism associating these two hyperglycemia-induced disorders remains poorly understood. This study intended to elucidate whether ER stress drives hyper-protein O-GlcNAcylation to cause podocyte injury in DN. We used both type 1 and type 2 DN models to confirm the occurrence of ER stress and excessive protein O-GlcNAcylation, and then podocyte purification was also conducted for further investigation. Nephroseq V5 data were mined and in vitro studies were applied to reveal the involvement of ER stress and hyper-O-GlcNAcylation in podocyte injury. Our results indicated that ER stress was induced in both type 1 and type 2 DN, and the human RNA-seq data from Nephroseq V5 showed that O-GlcNAcylation-related genes were significantly upregulated in the DN patients. We further demonstrated that ER stress occurred prior to hyper-O-GlcNAc modification and that pharmacologically inhibited protein O-GlcNAcylation can help decrease the podocyte apoptosis induced by hyperglycemia. Together, these discoveries will aid in uncovering the activation of the ER stress-O-GlcNAcylation axis in podocyte injury under DN, which will help open up new therapeutic approaches for preventing DN progression.


Asunto(s)
Diabetes Mellitus Tipo 2 , Nefropatías Diabéticas , Hiperglucemia , Podocitos , Humanos , Podocitos/metabolismo , Nefropatías Diabéticas/metabolismo , Estrés del Retículo Endoplásmico/genética , Proteínas/metabolismo , Hiperglucemia/metabolismo , Diabetes Mellitus Tipo 2/metabolismo
4.
Gels ; 9(10)2023 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-37888412

RESUMEN

Aerogels emerge as captivating contenders within the realm of high-temperature thermal resistance and thermal insulation. Nevertheless, their practical applications are usually constrained by their inherent brittleness when subjected to rigorous conditions. Herein, employing hafnium dichloride oxide octahydrate (HfOCl2·8H2O) as the hafnium source and resorcinol-formaldehyde (RF) as the carbon precursor, hafnium carbide (HfC) aerogels are fabricated via the sol-gel method complemented with carbothermal reduction reaction. Investigations are conducted into the effects of various molar ratios, duration, and temperatures of calcination on the microstructural features and physico-chemical characteristics of the as-prepared HfC aerogel. The aerogel shows a high BET-specific surface area (601.02 m2/g), which is much larger than those of previously reported aerogels. Furthermore, the HfC aerogel exhibits a low thermal conductivity of 0.053 W/(m·K) and a compressive strength of up to 6.12 MPa after carbothermal reduction at 1500 °C. These excellent thermal insulation and mechanical properties ensure it is ideal for the utilization of high-temperature thermal resistance and thermal insulation in the fields of aerospace.

5.
Int J Biol Macromol ; 246: 125616, 2023 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-37391003

RESUMEN

Naturally-derived aerogels have attracted considerable attention owing to their good biocompatibility, biodegradability and sustainability, but their weak mechanical properties largely limit their applications in various fields. Herein, we proposed the use of a directional freeze-drying method to prepare an anisotropic honeycomb three-dimensional porous aerogel with water-soluble chitosan (CS) as a rigid skeleton and water-soluble biobased epoxy resin as cross-linked hard segments, which had low volume shrinkage and density of 13.9 % and 34.3 mg/cm3, respectively. The resultant aerogel had anisotropic mechanical properties, such as rigidity in the axial direction with a maximum axial modulus of 6.71 MPa, which was 51.6 times larger than that of the pure chitosan aerogel, demonstrating a good compressive elasticity in the radial direction. It also had anisotropic thermal management properties, with a lower thermal conductivity in the radial direction than in the axial direction, down to 0.029 W/mK. The introduction of biobased epoxy resin improved the overall thermal stability, flame retardancy, and increased the biomass content in the aerogel, reducing the carbon footprint of the material. This study paves the way for the construction of a special graded porous, structurally and functionally integrated thermal insulation aerogel, which is of great significance for the development of new thermal insulation materials.


Asunto(s)
Quitosano , Poríferos , Animales , Resinas Epoxi , Anisotropía , Biomasa , Desecación
7.
Exp Cell Res ; 424(2): 113512, 2023 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-36775185

RESUMEN

Excessive mitochondrial fission in podocytes is a critical feature of diabetic nephropathy (DN). Mitochondria-associated endoplasmic reticulum membranes (MAMs) are contact sites between the endoplasmic reticulum (ER) and mitochondria, which are suggested to be related to mitochondrial function. However, the role of MAMs in mitochondrial dynamics disorder in podocytes remains unknown. Here, we firstly reported a novel mechanism of MAMs' effects on mitochondrial dynamics in podocytes under diabetic conditions. Increased MAMs were found in diabetic podocytes in vivo and in vitro, which were positively correlated with excessive mitochondrial fission. What's more, we also found that A-kinase anchoring protein 1 (AKAP1) was located in MAMs, and its translocation to MAMs was increased in podocytes cultured with high glucose (HG). In addition, AKAP1 knockdown significantly reduced mitochondrial fission and attenuated high glucose induced-podocyte injury through regulating phosphorylation of dynamin-related protein 1 (Drp1) and its subsequent mitochondrial translocation. On the contrary, AKAP1 overexpression in these podocytes showed the opposite effect. Finally, pharmacological inhibition of Drp1 alleviated excessive mitochondrial fission and podocyte damage in AKAP1 overexpressed podocytes. Our data suggest that MAMs were increased in podocytes under diabetic conditions, leading to excessive mitochondrial fission and podocyte damage through AKAP1-Drp1 signaling.


Asunto(s)
Podocitos , Dinaminas/metabolismo , Retículo Endoplásmico/metabolismo , Glucosa/farmacología , Glucosa/metabolismo , Mitocondrias/metabolismo , Dinámicas Mitocondriales , Podocitos/metabolismo , Proteínas de Anclaje a la Quinasa A/metabolismo
8.
Biogerontology ; 24(5): 783-799, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-36683095

RESUMEN

Atherosclerosis threatens human health by developing cardiovascular diseases, the deadliest disease world widely. The major mechanism contributing to the formation of atherosclerosis is mainly due to vascular endothelial cell (VECs) senescence. We have shown that 17ß-estradiol (17ß-E2) may protect VECs from senescence by upregulating autophagy. However, little is known about how 17ß-E2 activates the autophagy pathway to alleviate cellular senescence. Therefore, the aim of this study is to determine the role of estrogen receptor (ER) α and ß in the effects of 17ß-E2 on vascular autophagy and aging through in vitro and in vivo models. Hydrogen peroxide (H2O2) was used to establish Human Umbilical Vein Endothelial Cells (HUVECs) senescence. Autophagy activity was measured through immunofluorescence and immunohistochemistry staining of light chain 3 (LC3) expression. Inhibition of ER activity was established using shRNA gene silencing and ER antagonist. Compared with ER-ß knockdown, we found that knockdown of ER-α resulted in a significant increase in the extent of HUVEC senescence and senescence-associated secretory phenotype (SASP) secretion. ER-α-specific shRNA was found to reduce 17ß-E2-induced autophagy, promote HUVEC senescence, disrupt the morphology of HUVECs, and increase the expression of Rb dephosphorylation and SASP. These in vitro findings were found consistent with the in vivo results. In conclusion, our data suggest that 17ß-E2 activates the activity of ER-α and then increases the formation of autophagosomes (LC3 high expression) and decreases the fusion of lysosomes with autophagic vesicles (P62 low expression), which in turn serves to decrease the secretion of SASP caused by H2O2 and consequently inhibit H2O2-induced senescence in HUVEC cells.


Asunto(s)
Receptor alfa de Estrógeno , Peróxido de Hidrógeno , Humanos , Receptor alfa de Estrógeno/metabolismo , Peróxido de Hidrógeno/farmacología , Células Cultivadas , Estradiol/farmacología , Células Endoteliales de la Vena Umbilical Humana , Autofagia
9.
Int J Biochem Cell Biol ; 151: 106281, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35995387

RESUMEN

Excessive mitochondrial fission in podocytes serves as a central hub for the pathogenesis of diabetic nephropathy (DN), and the thromboxane/prostaglandin receptor (TP receptor) plays a potential role in DN. However, regulation of the TP receptor during mitochondrial dynamics disorder in podocytes remains unknown. Here, we firstly reported novel mechanistic details of TP receptor effects on mitochondrial dynamics in podocytes under diabetic conditions. Expression of the TP receptor was significantly upregulated in podocytes under diabetic conditions both in vivo and in vitro. S18886 attenuated podocyte mitochondrial fission, glomerular injury and renal dysfunction in diabetic mice. Furthermore, inhibition of the TP receptor by both genetic and pharmacological methods dramatically reduced mitochondrial fission and attenuated podocyte injury induced by high glucose through regulating dynamin-related protein 1 (Drp1) phosphorylation and its subsequent translocation to mitochondria. In contrast, TP receptor overexpression and application of TP receptor agonist U46619 in these podocytes showed the opposite effect on mitochondrial fission and podocyte injury. Furthermore, treatment with Y27632, an inhibitor of Rho-associated kinase1 (ROCK1), significantly blunted more fragmented mitochondria and reduced podocyte injuries in podocytes with TP receptor overexpression or after U46619 treatment. Finally, pharmacological inhibition of Drp1 alleviated excessive mitochondrial fragmentation and podocyte damage in TP receptor overexpressing podocytes. Our data suggests that increased expression of the TP receptor can occur in a human cultured podocyte cell line and in podocytes derived from streptozotocin (STZ)-induced diabetic mice, which contributes to mitochondrial excessive fission and podocyte injury via ROCK1-Drp1 signaling.


Asunto(s)
Diabetes Mellitus Experimental , Nefropatías Diabéticas , Enfermedades Mitocondriales , Podocitos , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/metabolismo , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacología , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/uso terapéutico , Animales , Diabetes Mellitus Experimental/patología , Nefropatías Diabéticas/patología , Dinaminas/metabolismo , Glucosa/metabolismo , Glucosa/farmacología , Humanos , Ratones , Enfermedades Mitocondriales/metabolismo , Dinámicas Mitocondriales , Prostaglandinas/metabolismo , Prostaglandinas/farmacología , Prostaglandinas/uso terapéutico , Receptores de Prostaglandina/metabolismo , Receptores de Prostaglandina/uso terapéutico , Receptores de Tromboxanos/metabolismo , Receptores de Tromboxanos/uso terapéutico , Estreptozocina , Tromboxanos/metabolismo , Tromboxanos/farmacología , Tromboxanos/uso terapéutico , Quinasas Asociadas a rho/metabolismo
10.
J Clin Med ; 12(1)2022 Dec 25.
Artículo en Inglés | MEDLINE | ID: mdl-36614968

RESUMEN

Diabetic kidney disease (DKD) and primary glomerular disease (PGD) are the main causes of chronic kidney disease (CKD) and end-stage renal disease (ESRD). This study was conducted to compare the characteristics of ambulatory blood-pressure monitoring (ABPM) and its relationship with target-organ damage (TOD) in patients with DKD and PGD matched by propensity score. The assessment of TOD included macroalbuminuria, left ventricular hypertrophy (LVH) and macrovascular disease. Propensity-score weighting (PSW) was used in stratified analysis. Results: Patients with DKD had a higher prevalence of abnormal blood-pressure patterns such as reversed dipper pattern, nocturnal hypertension, and sustained hypertension and had a higher prevalence of TOD than did patients with PGD. Logistic regression indicated that patients with DKD were more related to TOD than to PGD. The stratified analysis indicated that DKD patients with white-coat hypertension, masked hypertension and sustained hypertension had closer relationships with TOD compared with PGD patients. Conclusion: Patients with type 2 diabetic kidney disease had more abnormal blood-pressure patterns and were more closely related to target organ damage than were patients with primary glomerular disease.

11.
Steroids ; 170: 108829, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33811924

RESUMEN

Intrinsic cardiac aging increases cardiovascular mortality and morbidity in the elderly. Estrogen helps reduce the risk of cardiovascular disease in women, with 17ß-estradiol (17ß-E2) activating the autophagy pathway and inhibiting vascular aging, mainly through estrogen receptor alpha (ER α) to prevent atherosclerosis. Abnormal methylation of autophagy-related genes can impact autophagic regulation. We hypothesized that 17ß-E2, specifically 17ß-E2 α, downregulates the methylation of autophagy factors and delays cardiac aging. Here, we used d-galactose, 17ß-E2, and ER α receptor antagonist methyl-piperidino-pyrazole (MPP) to establish different aging models in mice divided into four groups, namely negative control, D.gal, D.gal + 17ß-E2, and D.gal + 17ß-E2 + MPP groups. Echocardiography showed that compared with the D.gal group group, the D.gal + 17ß-E2 showed substantially increased cardiac function. The level of cardiac aging markers in mice in the D.gal + 17ß-E2 group was lower than that in mice in the D.gal group. Beclin1, LC3, and Atg5 mRNA and protein expression levels in mice in the D.gal + 17ß-E2 group were significantly increased compared with those in the D.gal group. Additionally, Beclin1, LC3, and Atg5 methylation levels were significantly decreased in the D.gal + 17ß-E2 group. All the above values of the D.gal + 17ß-E2 + MPP group were between those of the D.gal and D.gal + 17ß-E2 groups. The expression of Dnmt1, Dnmt2, and Dnmt3A genes was the highest in the D.gal group. In summary, our results suggest that 17ß-E2, specifically 17ß-E2 α, promotes autophagy by downregulating the methylation of autophagy factors, thereby inhibiting galactose-induced cardiac aging in mice. 17ß-E2 may be a potential therapeutic target to mitigate the effects of cardiac aging.


Asunto(s)
Galactosa , Envejecimiento/efectos de los fármacos , Autofagia/efectos de los fármacos , Estradiol/farmacología , Metilación
12.
Cell Death Dis ; 11(9): 814, 2020 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-32994406

RESUMEN

Excessive mitochondrial fission plays a key role in podocyte injury in diabetic kidney disease (DKD), and long noncoding RNAs (lncRNAs) are important in the development and progression of DKD. However, lncRNA regulation of mitochondrial fission in podocytes is poorly understood. Here, we studied lncRNA maternally expressed gene 3 (Meg3) in mitochondrial fission in vivo and in vitro using human podocytes and Meg3 podocyte-specific knockdown mice. Expression of lncRNA Meg3 in STZ-induced diabetic mice was higher, and correlated with the number of podocytes. Excessive mitochondrial fission of podocytes and renal histopathological and physiological parameters were improved in podocyte-specific Meg3 knockdown diabetic mice. Elongated mitochondria with attenuated podocyte damage, as well as mitochondrial translocation of dynamin-related protein 1 (Drp1), were decreased in Meg3 knockout podocytes. By contrast, increased fragmented mitochondria, podocyte injury, and Drp1 expression and phosphorylation were observed in lncRNA Meg3-overexpressing podocytes. Treatment with Mdivi1 significantly blunted more fragmented mitochondria and reduced podocyte injury in lncRNA Meg3-overexpressing podocytes. Finally, fragmented mitochondria and Drp1 mitochondrial translocation induced by high glucose were reduced following treatment with Mdivi1. Our data show that expression of Meg3 in podocytes in both human cells and diabetic mice was higher, which regulates mitochondrial fission and contributes to podocyte injury through increased Drp1 and its translocation to mitochondria.


Asunto(s)
Glucosa/metabolismo , Dinámicas Mitocondriales , Podocitos/metabolismo , ARN Largo no Codificante/metabolismo , Animales , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Dinaminas/genética , Dinaminas/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Podocitos/patología , ARN Largo no Codificante/biosíntesis , ARN Largo no Codificante/genética
13.
Biogerontology ; 21(5): 549-557, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32172411

RESUMEN

17ß-estradiol (17ß-E2) has been implicated in inhibiting the senescence of vascular endothelial cells (VEC) and slowing down the process of atherosclerosis. However, the underlying molecular mechanisms are still unknown. In this study, we examined the roles of SIRT3 in 17ß-E2-induced autophagy and 17ß-E2-mediated inhibition of hydrogen peroxide (H2O2)-induced senescence in Human umbilical vein endothelial cells (HUVEC). Cellular senescence was measured by immunoblot analysis with antibodies against phosphorylated Rb and senescence-associated ß-galactosidase staining. Immunoblot analysis with antibodies against LC3 and p62 was performed to determine autophagy flux. Our findings show that 17ß-E2 activates SIRT3 promoter and upregulates SIRT3 gene expression in HUVEC cells. siRNA-mediated silencing of SIRT3 gene expression inhibits 17ß-E2-induced processing of LC3-I to LC3-II and degradation of p62, two widely-used makers of autophagy. SIRT3 knockdown also blocks 17ß-E2-induced inhibition of cellular senescence triggered by H2O2. Our data further reveal that SIRT3 knockdown impairs 17ß-E2-induced co-localization of LC3 and VDAC1, a marker protein on mitochondria, when HUVEC cells were co-treated with H2O2. Together, our findings suggest that 17ß-E2 upregulates SIRT3 gene expression by activating SIRT3 promoter and then promotes autophagy, which in turn serves to remove dysfunctional mitochondria caused by H2O2 and consequently inhibit H2O2-induced senescence in HUVEC cells.


Asunto(s)
Autofagia , Senescencia Celular , Estradiol/farmacología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Sirtuina 3/metabolismo , Silenciador del Gen , Humanos , Peróxido de Hidrógeno , Mitocondrias/patología
14.
Exp Gerontol ; 114: 57-66, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30399406

RESUMEN

Vascular endothelial cell (VEC) senescence is an initiating factor in numerous cardiovascular diseases. Recent studies showed that 17ß-estradiol (17ß-E2), an estrogen with numerous biological activities such as inhibition of atherosclerosis, protects VECs from senescence. However, the effects of 17ß-E2 on human umbilical VECs (HUVECs) remain unknown. This study investigated the anti-senescent effect of 17ß-E2 on HUVECs and explored the underlying mechanism with respect to autophagy and p53 activity. First, rapamycin and 3-methyladenine were used to clarify the relationship between autophagy and senescence in HUVECs, and an inverse relationship was demonstrated. Next, the effect of 17ß-E2 on H2O2-induced senescence of HUVECs was examined. Increased autophagy induced by 17ß-E2 inhibited H2O2-induced senescence of HUVECs, increased cell viability, and maintained HUVEC morphology. 17ß-E2 pre-treatment also decreased cell cycle arrest, decreased the dephosphorylation of Rb, decreased the production of ET-1, and increased the production of NO. Most importantly, 17ß-E2 pre-treatment increased autophagy by activating p53 and its downstream effector p53-upregulated modulator of apoptosis (PUMA). Overall, our data indicate the critical role of autophagy in the anti-senescent effect of 17ß-E2 on HUVECs.


Asunto(s)
Autofagia/efectos de los fármacos , Senescencia Celular/efectos de los fármacos , Estradiol/farmacología , Células Endoteliales de la Vena Umbilical Humana/citología , Proteína p53 Supresora de Tumor/metabolismo , Apoptosis , Puntos de Control del Ciclo Celular , Supervivencia Celular , Estrógenos/farmacología , Humanos , Peróxido de Hidrógeno/toxicidad , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...