Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters











Publication year range
1.
Pharmaceuticals (Basel) ; 17(8)2024 Jul 30.
Article in English | MEDLINE | ID: mdl-39204110

ABSTRACT

Alzheimer's disease (AD) is a neurodegenerative disorder associated with age. A wealth of evidence indicates that the amyloid ß (Aß) aggregates result from dyshomeostasis between Aß production and clearance, which plays a pivotal role in the pathogenesis of AD. Consequently, therapies targeting Aß reduction represent a promising strategy for AD intervention. Tetramethylpyrazine nitrone (TBN) is a novel tetramethylpyrazine derivative with potential for the treatment of AD. Previously, we demonstrated that TBN markedly enhanced cognitive functions and decreased the levels of Aß, APP, BACE 1, and hyperphosphorylated tau in 3×Tg-AD mice. However, the mechanism by which TBN inhibits Aß deposition is still unclear. In this study, we employed APP/PS1 mice treated with TBN (60 mg/kg, ig, bid) for six months, and N2a/APP695swe cells treated with TBN (300 µM) to explore the mechanism of TBN in Aß reduction. Our results indicate that TBN significantly alleviated cognitive impairment and reduced Aß deposition in APP/PS1 mice. Further investigation of the underlying mechanisms revealed that TBN decreased the expression of APP and BACE1, activated the AMPK/mTOR/ULK1 autophagy pathway, inhibited the PI3K/AKT/mTOR/ULK1 autophagy pathway, and decreased the phosphorylation levels of JNK and ERK in APP/PS1 mice. Moreover, TBN was found to significantly reduce the mRNA levels of APP and BACE1, as well as those of SP1, CTCF, TGF-ß, and NF-κB, transcription factors involved in regulating gene expression. Additionally, TBN was observed to decrease the level of miR-346 and increase the levels of miR-147 and miR-106a in the N2a/APP695swe cells. These findings indicate that TBN may reduce Aß levels likely by reducing APP expression by regulating APP gene transcriptional factors and miRNAs, reducing BACE1 expression, and promoting autophagy activities.

2.
Org Lett ; 25(46): 8387-8392, 2023 Nov 24.
Article in English | MEDLINE | ID: mdl-37966124

ABSTRACT

By extending the photoinduced oxidative mechanism of aryl tertiary amines proposed earlier to an alkaline environment based on the prediction of quantum mechanics computations and the validation of meticulous experiments, we discovered a photoinduced oxidative N-dealkylation method for both aryl tertiary amines and amides. The dealkylation was achieved in an alkaline environment under mild conditions accompanied by excellent functional group tolerance.

3.
Comput Struct Biotechnol J ; 21: 5476-5490, 2023.
Article in English | MEDLINE | ID: mdl-38022698

ABSTRACT

Precise diagnosis of early prostate cancer (PCa) is critical for preventing tumor progression. However, the diagnostic outcomes of currently used markers are far from satisfactory due to the low sensitivity or specificity. Here, we identified a diagnostic subpopulation in PCa tissue with the integrating analysis of single-cell and bulk RNA-seq. The representative markers of this subpopulation were extracted to perform intersection analysis with early-PCa-related gene module generated from weighted correlation network analysis (WGCNA). A total of 24 overlapping genes were obtained, the diagnostic roles of which were validated by distinguishing normal and tumorous prostate samples from the public dataset. A least absolute shrinkage and selection operator (LASSO) model was constructed based on these genes and the obtained 24-gene panel showed high sensitivity and specificity for PCa diagnosis, with better identifying capability of PCa than the commercially used gene panel of Oncotype DX. The top two risk factors, TRPM4 and PODXL2, were verified to be highly expressed in early PCa tissues by multiplex immunostaining, and PODXL2 was more sensitive and specific compared to TRPM4 and the pathologically used marker AMACR for early PCa diagnosis, suggesting a novel and promising pathology marker.

4.
Front Pharmacol ; 14: 1082602, 2023.
Article in English | MEDLINE | ID: mdl-36950017

ABSTRACT

The pathophysiology of Alzheimer's disease (AD) is multifactorial with characteristic extracellular accumulation of amyloid-beta (Aß) and intraneuronal aggregation of hyperphosphorylated tau in the brain. Development of disease-modifying treatment for AD has been challenging. Recent studies suggest that deleterious alterations in neurovascular cells happens in parallel with Aß accumulation, inducing tau pathology and necroptosis. Therefore, therapies targeting cellular Aß and tau pathologies may provide a more effective strategy of disease intervention. Tetramethylpyrazine nitrone (TBN) is a nitrone derivative of tetramethylpyrazine, an active ingredient from Ligusticum wallichii Franchat (Chuanxiong). We previously showed that TBN is a potent scavenger of free radicals with multi-targeted neuroprotective effects in rat and monkey models of ischemic stroke. The present study aimed to investigate the anti-AD properties of TBN. We employed AD-related cellular model (N2a/APPswe) and transgenic mouse model (3×Tg-AD mouse) for mechanistic and behavioral studies. Our results showed that TBN markedly improved cognitive functions and reduced Aß and hyperphosphorylated tau levels in mouse model. Further investigation of the underlying mechanisms revealed that TBN promoted non-amyloidogenic processing pathway of amyloid precursor protein (APP) in N2a/APPswe in vitro. Moreover, TBN preserved synapses from dendritic spine loss and upregulated synaptic protein expressions in 3×Tg-AD mice. Proteomic analysis of 3×Tg-AD mouse hippocampal and cortical tissues showed that TBN induced neuroprotective effects through modulating mitophagy, MAPK and mTOR pathways. In particular, TBN significantly upregulated PINK1, a key protein for mitochondrial homeostasis, implicating PINK1 as a potential therapeutic target for AD. In summary, TBN improved cognitive functions in AD-related mouse model, inhibited Aß production and tau hyperphosphorylation, and rescued synaptic loss and neuronal damage. Multiple mechanisms underlie the anti-AD effects of TBN including the modulation of APP processing, mTOR signaling and PINK1-related mitophagy.

5.
Eur J Pharmacol ; 947: 175671, 2023 May 15.
Article in English | MEDLINE | ID: mdl-37001578

ABSTRACT

Renal tubulointerstitial fibrosis (TIF) is one of the main pathological changes induced by diabetic kidney disease (DKD), and epithelial-to-mesenchymal transition (EMT) induced by high glucose (HG) can promote TIF. Our previous study has shown that ubiquitin-specific protease 22 (USP22) could affect the process of DKD by deubiquitinating and stabilizing Sirt1 in glomerular mesangial cells. However, whether USP22 could regulate EMT occurrence in renal tubular epithelial cells and further aggravate the pathological process of TIF in DKD remains to be elucidated. In this study, we found that USP22 expression was upregulated in kidney tissues of db/db mice and HG-treated NRK-52E cells. In vitro, USP22 overexpression promoted the EMT process of NRK-52E cells stimulated by HG and further increased the levels of extracellular matrix (ECM) components such as fibronectin, Collagen I, and Collagen Ⅳ. Meanwhile, USP22 deficiency exhibited the opposite effects. Mechanism studies showed that USP22, depending on its deubiquitinase activity, deubiquitinated and stabilized the EMT transcriptional factor Snail1. In vivo experiment showed that interfering with USP22 could improve the renal pathological damages and renal function of the db/db spontaneous diabetic mice by decreasing Snail1 expression, which could inhibit EMT occurrence, and reduce the production of ECM components. These results suggested that USP22 could accelerate renal EMT and promote the pathological progression of diabetic TIF by deubiquitinating Snail1, providing an experimental basis for using USP22 as a potential target for DKD.


Subject(s)
Diabetes Mellitus, Experimental , Diabetic Nephropathies , Snail Family Transcription Factors , Ubiquitin Thiolesterase , Animals , Mice , Collagen/metabolism , Diabetes Mellitus, Experimental/metabolism , Diabetic Nephropathies/pathology , Epithelial-Mesenchymal Transition , Fibrosis , Kidney , Rats , Cell Line , Snail Family Transcription Factors/metabolism , Ubiquitin Thiolesterase/metabolism
6.
Front Pharmacol ; 13: 853383, 2022.
Article in English | MEDLINE | ID: mdl-35401165

ABSTRACT

Diabetic nephropathy (DN) is quickly becoming the largest cause of end-stage renal disease (ESRD) in diabetic patients, as well as a major source of morbidity and mortality. Our previous studies indicated that the activation of Nrf2/ARE pathway via Connexin43 (Cx43) considerably contribute to the prevention of oxidative stress in the procession of DN. Fraxin (Fr), the main active glycoside of Fraxinus rhynchophylla Hance, has been demonstrated to possess many potential pharmacological activities. Whereas, whether Fr could alleviate renal fibrosis through regulating Cx43 and consequently facilitating the activation of Nrf2/ARE pathway needs further investigation. The in vitro results showed that: 1) Fr increased the expression of antioxidant enzymes including SOD1 and HO-1 to inhibit high glucose (HG)-induced fibronectin (FN) and inflammatory cell adhesion molecule (ICAM-1) overexpression; 2) Fr exerted antioxidant effect through activating the Nrf2/ARE pathway; 3) Fr significantly up-regulated the expression of Cx43 in HG-induced glomerular mesangial cells (GMCs), while the knock down of Cx43 largely impaired the activation of Nrf2/ARE pathway induced by Fr; 4) Fr promoted the activation of Nrf2/ARE pathway via regulating the interaction between Cx43 and AKT. Moreover, in accordance with the results in vitro, elevated levels of Cx43, phosphorylated-AKT, Nrf2 and downstream antioxidant enzymes related to Nrf2 were observed in the kidneys of Fr-treated group compared with model group. Importantly, Fr significantly improved renal dysfunction pathological changes of renal fibrosis in diabetic db/db mice. Collectively, Fr could increase the Cx43-AKT-Nrf2/ARE pathway activation to postpone the diabetic renal fibrosis and the up-regulation of Cx43 is probably a novel mechanism in this process.

7.
Biochem Pharmacol ; 188: 114562, 2021 06.
Article in English | MEDLINE | ID: mdl-33857489

ABSTRACT

Epithelial-to-mesenchymal transition (EMT) plays an important role in diabetic nephropathy (DN). Ubiquitin-specific protease 9X (USP9X/FAM) is closely linked to TGF-ß and fibrosis signaling pathway. However, it remains unknown whether USP9X is involved in the process of EMT in DN. Our previous study has shown that connexin 43 (Cx43) activation attenuated the development of diabetic renal tubulointerstitial fibrosis (RIF). Here, we showed that USP9X is a novel negative regulator of EMT and the potential mechanism is related to the deubiquitination and degradation of Cx43. To explore the potential regulatory mechanism of USP9X, the expression and activity of USP9X were studied by CRISPR/Cas9-based synergistic activation mediator (SAM) system, short hairpin RNAs, and selective inhibitor. The following findings were observed: (1) Expression of USP9X was down-regulated in the kidney tissue of db/db diabetic mice; (2) overexpression of USP9X suppressed high glucose (HG)-induced expressions of EMT markers and extra cellular matrix (ECM) in NRK-52E cells; (3) depletion of USP9X further aggravated EMT process and ECM production in NRK-52E cells; (4) USP9X deubiquitinated Cx43 and suppressed its degradation to regulate EMT process; (5) USP9X deubiquitinated Cx43 by directly binding to the C-terminal Tyr286 of Cx43. The current study determined the protective role of USP9X in the process of EMT and the molecular mechanism clarified that the protective effects of USP9X on DN were associated with the deubiquitination of Cx43.


Subject(s)
Connexin 43/metabolism , Epithelial-Mesenchymal Transition/drug effects , Glucose/toxicity , Kidney Tubules/metabolism , Ubiquitin Thiolesterase/biosynthesis , Animals , Connexin 43/genetics , Deubiquitinating Enzymes/biosynthesis , Deubiquitinating Enzymes/genetics , Dose-Response Relationship, Drug , Epithelial-Mesenchymal Transition/physiology , HEK293 Cells , Humans , Kidney Tubules/cytology , Kidney Tubules/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Rats , Ubiquitin Thiolesterase/genetics
8.
Clin Sci (Lond) ; 134(13): 1573-1592, 2020 07 17.
Article in English | MEDLINE | ID: mdl-32558900

ABSTRACT

Hyperglycemia-induced renal epithelial-to-mesenchymal transition (EMT) is a key pathological factor in diabetic renal tubulointerstitial fibrosis (RIF). Our previous studies have shown that connexin 43 (Cx43) activation attenuated the development of diabetic renal fibrosis. However, whether Cx43 regulates the EMT of renal tubular epithelial cells (TECs) and the pathological process of RIF under the diabetic conditions remains to be elucidated. In the present study, we identified that Cx43 protein expression was down-regulated in the kidney tissues of db/db mice as well as in high glucose (HG)-induced NRK-52E cells. Overexpression of Cx43 improved renal function in db/db spontaneous diabetic model mice, increased SIRT1 levels, decreased hypoxia-inducible factor (HIF)-1α expression, and reduced production of EMT markers and extracellular matrix (ECM) components. Additionally, Cx43 overexpression inhibited the EMT process and reduced the expression of ECM components such as fibronectin (FN), Collagen I, and Collagen IV in HG-induced NRK-52E cells, whereas Cx43 deficiency had the opposite effects. Mechanistically, Cx43 in a carboxyl-terminal signal transduction-dependent manner could up-regulate SIRT1 expression and enhance SIRT1-dependent deacetylation of HIF-1α to reduce HIF-1α activity, which eventually ameliorated renal EMT and diabetic RIF. Our study indicates the essential role of Cx43 in regulating renal EMT and diabetic RIF via regulating the SIRT1-HIF-1α signaling pathway and provides an experimental basis for Cx43 as a potential target for diabetic nephropathy (DN).


Subject(s)
Connexin 43/metabolism , Diabetic Nephropathies/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Kidney Tubules/metabolism , Sirtuin 1/metabolism , Animals , Connexin 43/genetics , Diabetic Nephropathies/genetics , Diabetic Nephropathies/pathology , Disease Progression , Fibrosis/genetics , Fibrosis/metabolism , Fibrosis/pathology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Kidney Tubules/pathology , Male , Mice , Mice, Inbred C57BL , Signal Transduction , Sirtuin 1/genetics
9.
Exp Cell Res ; 393(2): 112100, 2020 08 15.
Article in English | MEDLINE | ID: mdl-32442538

ABSTRACT

Oxidative stress is a key pathological factor for diabetic renal fibrosis by activating TGF-ß/Smad pathway in glomerular mesangial cells (GMCs) to promote the synthesis of extracellular matrix such as fibronectin (FN). Nuclear factor-E2-related factor (Nrf2)- anti-oxidant response element (ARE) anti-oxidative pathway has crucial renoprotective effects, and inhibiting ubiquitin-mediated degradation of Nrf2 delays diabetic renal fibrosis development. Ubiquitin-specific protease 9X (USP9X) has close relationship with oxidative stress and TGF-ß/Smad pathway, but whether it regulate diabetic renal fibrosis remains unclarified. Here, we found that advanced glycation-end products (AGEs) dose- and time-dependently reduced the protein expression and deubiquitinase activity of USP9X in GMCs. USP9X overexpression attenuated AGEs-induced upregulation of FN, TGF-ß1, and Collagen Ⅳ, three fibrosis-related marker proteins, in a deubiquitinase activity-dependent manner. While USP9X depletion with siRNAs further promoted the expressions of those proteins in AGEs-treated GMCs. Under AGEs treatment conditions, USP9X overexpression markedly increased the total and nuclear levels, ARE-binding ability, and transcriptional activity of Nrf2, upregulated the protein expressions of Nrf2 downstream genes HO-1 and NQO1, and eventually reduced the excessive production of ROS. Overexpression of the deubiquitinase catalytically inactive USP9X-C1556S mutant failed to exert such effects. Silencing Nrf2 abolished the renoprotective effects of USP9X. Further study showed that upon AGEs stimulation, Nrf2 transferred into the nucleus and the interaction between USP9X and Nrf2 was weakened. AGEs also increased Nrf2 ubiquitination level, and overexpression of USP9X, instead of USP9X-C1556S, significantly reduced the ubiquitination level of Nrf2. Taken together, USP9X reduced Nrf2 ubiquitination level and promoted Nrf2-ARE pathway activation to prevent the accumulation of extracellular matrix, eventually alleviated the pathological process of diabetic renal fibrosis.


Subject(s)
Diabetes Mellitus, Experimental/prevention & control , Mesangial Cells/drug effects , Transforming Growth Factor beta1/metabolism , Ubiquitin Thiolesterase/pharmacology , Animals , Antioxidant Response Elements/drug effects , Antioxidant Response Elements/physiology , Diabetes Mellitus, Experimental/metabolism , Diabetic Nephropathies/metabolism , Glycation End Products, Advanced/pharmacology , Mesangial Cells/metabolism , Rats, Sprague-Dawley , Transforming Growth Factor beta1/drug effects , Ubiquitin-Specific Proteases/metabolism , Up-Regulation
10.
Br J Pharmacol ; 177(1): 145-160, 2020 01.
Article in English | MEDLINE | ID: mdl-31465542

ABSTRACT

BACKGROUND AND PURPOSE: Nox4 is the major isoform of NADPH oxidase found in the kidney and contributes to the pathogenesis of diabetic nephropathy. However, the molecular mechanisms of increased Nox4 expression induced by hyperglycaemia remain to be elucidated. Here, the role of the connexin32-Nox4 signalling axis in diabetic nephropathy and its related mechanisms were investigated. EXPERIMENTAL APPROACH: Diabetes was induced in mice by low-dose streptozotocin (STZ) combined with a high-fat diet. Effects of connexin32 on Nox4 expression and on renal function and fibrosis in STZ-induced diabetic mice were investigated using adenovirus-overexpressing connexin32 and connexin32-deficient mice. Interactions between connexin32 and Nox4 were analysed by co-immunoprecipitation and immunofluorescence assays. KEY RESULTS: Connexin32 was down-regulated in the kidneys of STZ-induced diabetic mice. Overexpression of connexin32 reduced expression of Nox4 and improved renal function and fibrosis in diabetic mice, whereas connexin32 deficiency had opposite effects. Down-regulation of fibronectin expression by connexin32 was not dependent on gap junctional intercellular communication involving connexin32. Connexin32 interacted with Nox4 and reduced the generation of hydrogen peroxide, leading to the down-regulation of fibronectin expression. Mechanistically, connexin32 decreased Nox4 expression by promoting its K48-linked polyubiquitination. Interestingly, Smurf1 overexpression inhibited K48-linked polyubiquitination of Nox4. Furthermore, connexin32 interacted with Smurf1 and inhibited its expression. CONCLUSION AND IMPLICATIONS: Connexin32 ameliorated renal fibrosis in diabetic mice by promoting K48-linked Nox4 polyubiquitination and degradation via inhibition of Smurf1 expression. Targeting the connexin32-Nox4 signalling axis may contribute to the development of novel treatments for diabetic nephropathy.


Subject(s)
Bacterial Capsules/metabolism , Connexins/metabolism , Diabetes Mellitus, Experimental/metabolism , Diabetic Nephropathies/metabolism , NADPH Oxidase 4/metabolism , Animals , Cells, Cultured , Connexins/antagonists & inhibitors , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/prevention & control , Diabetic Nephropathies/chemically induced , Diabetic Nephropathies/prevention & control , Diet, High-Fat/adverse effects , Fibrosis , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NADPH Oxidase 4/antagonists & inhibitors , Proteolysis/drug effects , Rats , Rats, Sprague-Dawley , Streptozocin/toxicity , Ubiquitination/drug effects , Ubiquitination/physiology , Gap Junction beta-1 Protein
11.
Biol Pharm Bull ; 41(6): 891-898, 2018.
Article in English | MEDLINE | ID: mdl-29863077

ABSTRACT

Previous investigations on diabetic rats and palmitic corrosive instigated insulin-resistant HepG2 cells have shown that polydatin exhibits hypoglycemic and hypolipidemic impacts. The AMP-activated protein kinase (AMPK) pathway assumes a crucial part in glucose and lipid digestion. We aimed to investigate the regulatory system of polydatin on the glucose and lipid metabolism through the AMPK pathway. Glucose take-up, utilization levels, and oil red O recoloring were distinguished to confirm their impact on improving insulin resistance. A Western blot examination was utilized to investigate the phosphorylation levels of protein kinase B (Akt), glycogen synthase kinase (GSK)-3ß, AMPK, acetyl-CoA carboxylase (ACC), and in addition the protein levels of the low-density lipoprotein receptor (LDLR) and sterol regulatory element-binding protein (SREBP)-1c. SREBP-1c nuclear translocation levels were recognized by a laser checking confocal magnifying instrument. One hundred nanomolar insulin treated for 24 h significantly declined the phosphorylation of Akt and AMPK, and increased the nucleoproteins of SREBP-1c compared with HepG2 cells without insulin. The insulin-resistant HepG2 cells prompted by insulin mediated the impact of polydatin on glucose and lipid digestion. Polydatin decreased glucose and lipid digestion of insulin-resistant HepG2 cells. Moreover, polydatin markedly raised phosphorylated Akt, GSK-3ß, AMPK, ACC, diminished nuclear protein levels of SREBP-1c, and upgraded the protein levels of LDLR. Regulation of the AMPK pathway and changes in LDLR protein expression are potential focuses of polydatin in the treatment of insulin protection in insulin-resistant HepG2 cells.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Glucose/metabolism , Glucosides/pharmacology , Lipid Metabolism/drug effects , Stilbenes/pharmacology , Acetyl-CoA Carboxylase/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Hep G2 Cells , Humans , Insulin Resistance , Proto-Oncogene Proteins c-akt/metabolism , Receptors, LDL/metabolism , Signal Transduction/drug effects , Sterol Regulatory Element Binding Protein 1/metabolism
12.
Eur J Pharmacol ; 832: 138-144, 2018 Aug 05.
Article in English | MEDLINE | ID: mdl-29782856

ABSTRACT

Mesangial proliferative glomerulonephritis (MsPGN) is characterized by glomerular mesangial cells proliferation and extracellular matrix deposition in mesangial area, which develop into glomerulosclerosis. Both silent information regulator 2-related protein 1 (Sirt1) and nuclear factor erythroid 2-related factor 2/anti-oxidant response element (Nrf2/ARE) pathway had remarkable renoprotective effects. However, whether Sirt1 and Nrf2/ARE pathway can regulate the pathological process of MsPGN remains unknown. Here, we found that Sirt1 activation by SRT1720 decreased mesangial hypercellularity and mesangial matrix areas, reduced renal Col4 and α-SMA expressions, lowered 24 h proteinuria, and eventually reduced FN and TGF-ß1 expressions in rats received anti-Thy 1.1 IgG. Further study showed that SRT1720 markedly enhanced the activity of Nrf2/ARE pathway including promoting the nuclear content and ARE-binding ability of Nrf2, elevating the protein levels of HO-1 and SOD1, two target genes of Nrf2, which eventually increased total SOD activity and decreased malondialdehyde level in the kidney tissues of experimental anti-Thy 1.1 MsPGN rats. Taken together, Sirt1 prevented the pathological process of experimental anti-Thy 1.1 MsPGN through promoting the activation of Nrf2/ARE pathway, which warrants further elucidation. Sirt1 might be a potential therapeutic target for treating MsPGN.


Subject(s)
Antioxidants/metabolism , Glomerulonephritis/pathology , Glomerulonephritis/prevention & control , Mesangial Cells/pathology , NF-E2-Related Factor 2/metabolism , Response Elements/drug effects , Sirtuin 1/metabolism , Animals , Enzyme Activation/drug effects , Glomerulonephritis/enzymology , Male , Mesangial Cells/drug effects , Rats , Rats, Sprague-Dawley , Rats, Wistar
13.
Exp Cell Res ; 361(1): 63-72, 2017 12 01.
Article in English | MEDLINE | ID: mdl-28986066

ABSTRACT

Oxidative stress aroused by advanced glycation-end products (AGEs) is a culprit in the pathological progression of diabetic nephropathy. Both Sirt1 and the Keap1/Nrf2/ARE anti-oxidative pathway exert crucial inhibitory effects on the development of diabetic nephropathy. Our previous study has confirmed that Sirt1 activation can inhibit the upregulation of fibronectin (FN) and transforming growth factor-ß1 (TGF-ß1) by promoting Keap1/Nrf2/ARE pathway in glomerular mesangial cells (GMCs) challenged with AGEs. However, the underlying mechanism needs further investigation. Here, we found that concomitant with deacetylating and reducing the ubiquitination levels of Nrf2, Sirt1 significantly enhanced the activity of Keap1/Nrf2/ARE pathway including decreasing Keap1 expression, promoting the nuclear content, ARE-binding ability, and transcriptional activity of Nrf2, augmenting the protein levels of heme oxygenase 1, a target gene of Nrf2, which eventually quenched ROS overproduction and alleviating FN and TGF-ß1 accumulation in AGEs-treated GMCs. And depletion of Nrf2 blocked those renoprotective effects of Sirt1. Interestingly, Nrf2 also positively regulated Sirt1 at the protein expression and deacetylase activity levels as evidenced by tert-Butylhydroquinone and specific siRNA targeting Nrf2 to downregulate FN and TGF-ß1. In conclusion, the current study basically demonstrated that the crosstalk between Sirt1 and Keap1/Nrf2/ARE anti-oxidative pathway forms a positive feedback loop to inhibit the protein expressions of FN and TGF-ß1 in AGEs-treated GMCs.


Subject(s)
Carboxylic Ester Hydrolases/metabolism , Fibronectins/antagonists & inhibitors , Kelch-Like ECH-Associated Protein 1/metabolism , Kidney Glomerulus/metabolism , Mesangial Cells/metabolism , NF-E2-Related Factor 2/metabolism , Sirtuin 1/metabolism , Transforming Growth Factor beta1/antagonists & inhibitors , Animals , Cells, Cultured , Feedback, Physiological/drug effects , Glycation End Products, Advanced/pharmacology , Kidney Glomerulus/cytology , Kidney Glomerulus/drug effects , Mesangial Cells/cytology , Mesangial Cells/drug effects , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects
14.
Article in English | MEDLINE | ID: mdl-27034698

ABSTRACT

Systemic lupus erythematosus (SLE) is a chronic autoimmune disease that leads to severe multiorgan damage. Lang Chuang Fang (LCF) is a Chinese herbal medicine that is clinically prescribed for treating SLE. In this study, we examined the therapeutic effects of LCF granule on lupus-prone MRL/lpr mice. Female mice were randomly separated into six groups, and LCF treatment groups received LCF granule at the dosage of 0.97 g/kg/d, 1.95 g/kg/d, and 3.90 g/kg/d, respectively. Here, we found that, compared to the MRL/lpr mice, both the spleen coefficient and thymus coefficient were reduced in the LCF granule-treated mice. There was a marked downregulation in CRP and anti-dsDNA autoantibody and an evident upregulation of CH50 in LCF granule-treated mice. LCF granule treatment also obviously reduced the proteinuria, BUN, and SCr levels in MRL/lpr mice at the dosage of 0.97 g/kg/d, 1.95 g/kg/d, and 3.90 g/kg/d, indicating that LCF granule alleviated the renal injury of MRL/lpr mice. Furthermore, LCF granule decreased p65 NF-κB levels and increased Sirt1 and Nrf2 levels in the kidney tissues of MRL/lpr mice, which might elucidate the beneficial effects of LCF on lupus nephritis. In conclusion, this study demonstrates that LCF granule has therapeutic effects on lupus-prone MRL/lpr mice.

15.
Mol Cell Endocrinol ; 427: 45-56, 2016 May 15.
Article in English | MEDLINE | ID: mdl-26948947

ABSTRACT

We previously demonstrated that activation of sphingosine kinase 1 (SphK1)- sphingosine 1- phosphate (S1P) signaling pathway by high glucose (HG) plays a pivotal role in increasing the expression of fibronectin (FN), an important fibrotic component, by promoting the DNA-binding activity of transcription factor activator protein 1 (AP-1) in glomerular mesangial cells (GMCs) under diabetic conditions. As a multi-target anti-oxidative drug, polydatin (PD) has been shown to have renoprotective effects on experimental diabetes. However, whether PD could resist diabetic nephropathy (DN) by regulating SphK1-S1P signaling pathway needs further investigation. Here, we found that PD significantly reversed the upregulated FN and ICAM-1 expression in GMCs exposed to AGEs. Simultaneously, PD dose-dependently inhibited SphK1 levels at the protein expression and kinase activity and attenuated S1P production under AGEs treatment conditions. In addition, PD reduced SphK activity in GMCs transfected with wild-type SphK(WT) plasmid and significantly suppressed SphK1-mediated increase of FN and ICAM-1 levels under normal conditions. Furthermore, we found that the AGEs-induced upregulation of phosphorylation of c-Jun at Ser63 and Ser73 and c-Fos at Ser32, DNA-binding activity and transcriptional activity of AP-1 were blocked by PD. In comparison with db/db model group, PD treatment suppressed SphK1 levels (mRNA, protein expression, and activity) and S1P production, reversed the upregulation of FN, ICAM-1, c-Jun, and c-Fos in the kidney tissues of diabetic mice, and finally ameliorated renal injury in db/db mice. These findings suggested that the downregulation of SphK1-S1P signaling pathway is probably a novel mechanism by which PD suppressed AGEs-induced FN and ICAM-1 expression and improved renal dysfunction of diabetic models.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Fibronectins/metabolism , Glomerular Mesangium/drug effects , Glucosides/pharmacology , Hypoglycemic Agents/pharmacology , Intercellular Adhesion Molecule-1/metabolism , Signal Transduction/drug effects , Stilbenes/pharmacology , Animals , Cells, Cultured , Diabetes Mellitus, Experimental/drug therapy , Drugs, Chinese Herbal/pharmacology , Female , Glomerular Mesangium/metabolism , Glycation End Products, Advanced/antagonists & inhibitors , Glycation End Products, Advanced/pharmacology , Humans , Lysophospholipids/antagonists & inhibitors , Lysophospholipids/metabolism , Male , Mice , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Rats , Rats, Sprague-Dawley , Sphingosine/analogs & derivatives , Sphingosine/antagonists & inhibitors , Sphingosine/metabolism , Up-Regulation/drug effects
16.
Endocrinology ; 156(1): 268-79, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25375034

ABSTRACT

We previously demonstrated that advanced glycation-end products (AGEs) promote the pathological progression of diabetic nephropathy by decreasing silent information regulator 2-related protein 1 (Sirt1) expression in glomerular mesangial cells (GMCs). Here, we investigated whether AGEs-receptor for AGEs (RAGE) system down-regulated Sirt1 expression through ubiquitin-proteasome pathway and whether Sirt1 ubiquitination affected fibronectin (FN) and TGF-ß1, 2 fibrotic indicators in GMCs. Sirt1 was polyubiquitinated and subsequently degraded by proteasome. AGEs increased Sirt1 ubiquitination and proteasome-mediated degradation, shortened Sirt1 half-life, and promoted FN and TGF-ß1 expression. Ubiquitin-specific protease 22 (USP22) reduced Sirt1 ubiquitination and degradation and decreased FN and TGF-ß1 expression in GMCs under both basal and AGEs-treated conditions. USP22 depletion enhanced Sirt1 degradation and displayed combined effects with AGEs to further promote FN and TGF-ß1 expression. RAGE functioned crucial mediating roles in these processes via its C-terminal cytosolic domain. Inhibiting Sirt1 by EX-527 substantially suppressed the down-regulation of FN and TGF-ß1 resulting from USP22 overexpression under both normal and AGEs-treated conditions, eventually leading to their up-regulation in GMCs. These results indicated that the AGEs-RAGE system increased the ubiquitination and subsequent proteasome-mediated degradation of Sirt1 by reducing USP22 level, and AGEs-RAGE-USP22-Sirt1 formed a cascade pathway that regulated FN and TGF-ß1 level, which participated in the pathological progression of diabetic nephropathy.


Subject(s)
Fibronectins/metabolism , Gene Expression Regulation/physiology , Glycation End Products, Advanced/metabolism , Receptors, Immunologic/metabolism , Transforming Growth Factor beta1/metabolism , Animals , Fibronectins/genetics , Male , Rats , Rats, Sprague-Dawley , Receptor for Advanced Glycation End Products , Receptors, Immunologic/genetics , Sirtuin 1/genetics , Sirtuin 1/metabolism , Thiolester Hydrolases/genetics , Thiolester Hydrolases/metabolism , Transforming Growth Factor beta1/genetics
17.
Mol Cell Endocrinol ; 399: 178-89, 2015 Jan 05.
Article in English | MEDLINE | ID: mdl-25192797

ABSTRACT

Sirt1 and nuclear factor-E2 related factor 2 (Nrf2)-anti-oxidant response element (ARE) anti-oxidative pathway play important regulatory roles in the pathological progression of diabetic nephropathy (DN) induced by advanced glycation-end products (AGEs). Polydatin (PD), a glucoside of resveratrol, has been shown to possess strong anti-oxidative bioactivity. Our previous study demonstrated that PD markedly resists the progression of diabetic renal fibrosis and thus, inhibits the development of DN. Whereas, whether PD could resist DN through regulating Sirt1 and consequently promoting Nrf2-ARE pathway needs further investigation. Here, we found that concomitant with decreasing RAGE (the specific receptor for AGEs) expression, PD significantly reversed the downregulation of Sirt1 in terms of protein expression and deacetylase activity and attenuated FN and TGF-ß1 expression in GMCs exposed to AGEs. Under AGEs-treatment condition, PD could decrease Keap1 expression and promote the nuclear content, ARE-binding ability, and transcriptional activity of Nrf2. In addition, PD increased the protein levels of heme oxygenase 1 (HO-1) and superoxide dismutase 1 (SOD1), two target genes of Nrf2. The activation of Nrf2-ARE pathway by PD eventually led to the quenching of ROS overproduction sharply boosted by AGEs. Depletion of Sirt1 blocked Nrf2-ARE pathway activation and reversed FN and TGF-ß1 downregulation induced by PD in GMCs challenged with AGEs. Along with reducing HO-1 and SOD1 expression, silencing of Nrf2 increased FN and TGF-ß1 levels. PD treatment elevated Sirt1 and Nrf2 levels in the kidney tissues of diabetic rats, then improved the anti-oxidative capacity and renal dysfunction of diabetic models, and finally reversed the upregulation of FN and TGF-ß1. Taken together, the resistance of PD on upregulated FN and TGF-ß1 induced by AGEs via oxidative stress in GMCs is closely associated with its activation of Sirt1-Nrf2-ARE pathway.


Subject(s)
Fibronectins/biosynthesis , Glucosides/pharmacology , Glycation End Products, Advanced/metabolism , Mesangial Cells/metabolism , NF-E2-Related Factor 2/metabolism , Response Elements , Sirtuin 1/metabolism , Stilbenes/pharmacology , Transforming Growth Factor beta1/biosynthesis , Up-Regulation/drug effects , Animals , Cells, Cultured , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetic Nephropathies/genetics , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Fibronectins/genetics , Gene Silencing , Glycation End Products, Advanced/genetics , Mesangial Cells/pathology , NF-E2-Related Factor 2/genetics , Oxidative Stress/drug effects , Oxidative Stress/genetics , Rats , Rats, Sprague-Dawley , Receptor for Advanced Glycation End Products , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Sirtuin 1/genetics , Transforming Growth Factor beta1/genetics , Up-Regulation/genetics
18.
Eur J Pharmacol ; 745: 152-65, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25310908

ABSTRACT

Recently, the effect of polydatin on lipid regulation has gained considerable attention. And previous study has demonstrated that polydatin has hypoglycemic effect on experimental diabetic rats. Repressed Akt pathway contributes to glucose and lipid disorders in diabetes. Thus, whether polydatin regulates glucose and lipid metabolism in experimental diabetic models through the Akt pathway arouses interest. The purpose was to explore the regulatory mechanism of polydain on glucose and lipid through Akt pathway. We used a diabetic rat model induced by high-fat and -sugar diet with low-dose of streptozocin and an insulin resistant HepG2 cell model induced by palmitic acid to clarify the role of polydatin on glucose and lipid metabolism. Here, we found that polydatin significantly attenuated fasting blood­glucose, glycosylated hemoglobin, glycosylated serum protein, total cholesterol, triglyceride, and low-density lipoprotein cholesterol in diabetic rats. Furthermore, polydatin significantly increased glucose uptake and consumption and decreased lipid accumulation in insulin resistant HepG2 cells. Polydatin markedly increased serum insulin levels in diabetic rats, and obviously activated the Akt signaling pathway in diabetic rat livers and insulin resistant HepG2 cells. Polydatin markedly increased phosphorylated GSK-3ß, decreased the protein levels of G6Pase and SREBP-1c, and increased protein levels of GCK, LDLR, and phosphorylated IRS in livers and HepG2 cells. Overall, the results indicate that polydatin regulates glucose and lipid metabolism in experimental diabetic models, the underlying mechanism is probably associated with regulating the Akt pathway. The effect of polydatin on increased Akt phosphorylation is independent of prompting insulin secretion, but dependent of increasing IRS phosphorylation.


Subject(s)
Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Glucose/metabolism , Glucosides/pharmacology , Lipid Metabolism/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Stilbenes/pharmacology , Animals , Blood Glucose/metabolism , Diabetes Mellitus, Experimental/blood , Drugs, Chinese Herbal/pharmacology , Glucokinase/metabolism , Glucose-6-Phosphatase/metabolism , Hep G2 Cells , Humans , Hypoglycemic Agents/pharmacology , Insulin/blood , Insulin Resistance , Liver/drug effects , Liver/metabolism , Male , Rats , Rats, Sprague-Dawley , Receptors, LDL/metabolism , Signal Transduction/drug effects , Sterol Regulatory Element Binding Protein 1/metabolism
19.
Exp Cell Res ; 327(2): 276-86, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25064462

ABSTRACT

RhoA/Rho kinase (ROCK) signaling has been suggested to be involved in diabetic nephropathy (DN) pathogenesis. Altered expression of connexin43 (Cx43) has been found in kidneys of diabetic animals. Both of them have been found to regulate nuclear factor kappa-B (NF-κB) activation in high glucose-treated glomerular mesangial cells (GMCs). The aim of this study was to investigate the relationship between RhoA/ROCK signaling and Cx43 in the DN pathogenesis. We found that upregulation of Cx43 expression inhibited NF-κB p65 nuclear translocation induced by RhoA/ROCK signaling in GMCs. Inhibition of RhoA/ROCK signaling attenuated the high glucose-induced decrease in Cx43. F-actin accumulation and an enhanced interaction between zonula occludens-1 (ZO-1) and Cx43 were observed in high glucose-treated GMCs. ZO-1 depletion or disruption of F-actin formation also inhibited the reduction in Cx43 protein levels induced by high glucose. In conclusion, activated RhoA/ROCK signaling induces Cx43 degradation in GMCs cultured in high glucose, depending on F-actin regulation. Increased F-actin induced by RhoA/ROCK signaling promotes the association between ZO-1 and Cx43, which possibly triggered Cx43 endocytosis, a mechanism of NF-κB activation in high glucose-treated GMCs.


Subject(s)
Connexin 43/metabolism , Glucose/pharmacology , Mesangial Cells/metabolism , Signal Transduction/drug effects , Zonula Occludens-1 Protein/metabolism , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Apoptosis/drug effects , Blotting, Western , Cell Proliferation/drug effects , Cells, Cultured , Immunoprecipitation , Male , Mesangial Cells/cytology , Mesangial Cells/drug effects , Rats , Rats, Sprague-Dawley , Sweetening Agents/pharmacology
20.
Cell Signal ; 26(3): 629-38, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24342046

ABSTRACT

Our previous studies have confirmed that the sphingosine kinase 1 (SphK1)-sphingosine 1-phosphate (S1P) signaling pathway in the kidney under diabetic conditions is closely correlated with the pathogenesis of diabetic nephropathy (DN). The activation of SphK1-S1P pathway by high glucose (HG) can increase the expression of fibronectin (FN), an important fibrotic component, in glomerular mesangial cells (GMCs) by promoting the DNA-binding activity of transcription factor AP-1. However, the mechanism responsible for the sustained activation of SphK1-S1P pathway remains unclear. Given the binding motifs for AP-1 within the first intron of the SphK1 gene, we speculated that the activated AP-1 in the kidney under HG condition possibly regulates SphK1 expression in a positive feedback manner, thereby promoting the sustained activation of SphK1-S1P pathway and mediating the pathological progression of DN. Here, we observed the effect of AP-1 on SphK1 expression in GMCs and explored the molecular mechanism involved in the sustained activation of SphK1-S1P pathway. We found two consensus binding motifs for AP-1 in the promoter sequences and non-coding region downstream of the transcriptional initiation of the rat SphK1 gene by chromatin immunoprecipitation assay. The treatment of GMCs with both HG and S1P significantly increased the protein expression of c-Jun and c-Fos, and obviously enhanced the phosphorylation of c-Jun at Ser63 and Ser73, and c-Fos at Ser32. Knockdown of c-Jun and c-Fos with siRNAs substantially inhibited the expression of SphK1 and FN, whereas overexpression of c-Jun and c-Fos significantly increased the expression of SphK1 and FN. Curcumin treatment greatly decreased the levels of c-Jun, c-Fos, SphK1, and FN in the kidney tissues of diabetic rats. SiRNAs targeting SphK1 and S1P2 receptor respectively inhibited the phosphorylation of c-Jun (ser63 and ser73) and c-Fos (ser32), as well as FN expression under both normal and HG conditions. Our data demonstrated that the activated SphK1-S1P signaling pathway in GMCs under diabetic conditions is closely associated with AP-1 to form a positive feedback loop. This positive feedback loop functions as an important molecular basis for the sustained activation of SphK1-S1P pathway and increased FN expression that lead to the initiation and progression of DN.


Subject(s)
Diabetic Nephropathies/pathology , Glucose/pharmacology , Mesangial Cells/metabolism , Phosphotransferases (Alcohol Group Acceptor)/genetics , Transcription Factor AP-1/genetics , Animals , Binding Sites/genetics , Cells, Cultured , Curcumin/pharmacology , DNA-Binding Proteins , Diabetes Mellitus/pathology , Enzyme Inhibitors/pharmacology , Fibronectins/biosynthesis , JNK Mitogen-Activated Protein Kinases/biosynthesis , JNK Mitogen-Activated Protein Kinases/genetics , Lysophospholipids/biosynthesis , Lysophospholipids/pharmacology , Male , Mesangial Cells/drug effects , Phosphorylation/genetics , Phosphotransferases (Alcohol Group Acceptor)/biosynthesis , Promoter Regions, Genetic/genetics , Protein Binding/genetics , Proto-Oncogene Proteins c-fos/biosynthesis , Proto-Oncogene Proteins c-fos/genetics , RNA Interference , RNA, Small Interfering , Rats , Receptors, Lysosphingolipid/genetics , Signal Transduction/drug effects , Sphingosine/analogs & derivatives , Sphingosine/biosynthesis , Sphingosine/pharmacology , Sweetening Agents/pharmacology , Transcription Factor AP-1/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL