Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
J Nanobiotechnology ; 20(1): 205, 2022 Apr 27.
Article in English | MEDLINE | ID: mdl-35477452

ABSTRACT

BACKGROUND: The reactive oxygen species (ROS) and inflammation, a critical contributor to tissue damage, is well-known to be associated with various disease. The kidney is susceptible to hypoxia and vulnerable to ROS. Thus, the vicious cycle between oxidative stress and renal hypoxia critically contributes to the progression of chronic kidney disease and finally, end-stage renal disease. Thus, delivering therapeutic agents to the ROS-rich inflammation site and releasing the therapeutic agents is a feasible solution. RESULTS: We developed a longer-circulating, inflammation-sensing, ROS-scavenging versatile nanoplatform by stably loading catalase-mimicking 1-dodecanethiol stabilized Mn3O4 (dMn3O4) nanoparticles inside ROS-sensitive nanomicelles (PTC), resulting in an ROS-sensitive nanozyme (PTC-M). Hydrophobic dMn3O4 nanoparticles were loaded inside PTC micelles to prevent premature release during circulation and act as a therapeutic agent by ROS-responsive release of loaded dMn3O4 once it reached the inflammation site. CONCLUSIONS: The findings of our study demonstrated the successful attenuation of inflammation and apoptosis in the IRI mice kidneys, suggesting that PTC-M nanozyme could possess promising potential in AKI therapy. This study paves the way for high-performance ROS depletion in treating various inflammation-related diseases.


Subject(s)
Acute Kidney Injury , Acute Kidney Injury/drug therapy , Animals , Catalase , Female , Humans , Hypoxia , Inflammation/drug therapy , Male , Mice , Oxidative Stress , Reactive Oxygen Species
2.
Front Pharmacol ; 12: 708575, 2021.
Article in English | MEDLINE | ID: mdl-34588982

ABSTRACT

Maslinic acid (MA), also named crategolic acid, is a pentacyclic triterpene extracted from fruits and vegetables. Although various beneficial pharmacological effects of MA have been revealed, its effect on renal fibrosis remains unclear. This study was designed to clarify whether MA could attenuate renal fibrosis and determine the putative underlying molecular mechanisms. We demonstrated that MA-treated mice with unilateral ureteral obstruction (UUO) developed a histological injury of low severity and exhibited downregulated expression of fibrotic markers, including α-smooth muscle actin (α-SMA), vimentin, and fibronectin by 38, 44 and 40%, and upregulated expression of E-cadherin by 70% as compared with untreated UUO mice. Moreover, MA treatment restored the expression levels of α-SMA, connective tissue growth factor, and vimentin to 10, 7.8 and 38% of those induced by transforming growth factor (TGF)-ß in NRK49F cells. MA decreased expression of Smad2/3 phosphorylation and Smad4 in UUO kidneys and TGF-ß treated NRK49F cells (p < 0.05, respectively). Notably, MA specifically interferes with MyD88, an adaptor protein, thereby mitigating Smad4 nuclear expression (p < 0.01 compared to TGF-ß treated group) and ameliorating renal fibrotic changes (p < 0.01 for each fibrotic markers compared to TGF-ß induced cells). In addition, in the UUO model and lipopolysaccharide-induced NRK49F cells, MA treatment decreased the expression of IL-1ß, TGF-α and MCP-1, ICAM-1, associated with the suppression of NF-κB signaling. These findings suggest that MA is a potential agent that can reduce renal interstitial fibrosis, to some extent, via targeting TGF-ß/Smad and MyD88 signaling.

3.
Int J Pharm ; 600: 120497, 2021 May 01.
Article in English | MEDLINE | ID: mdl-33753165

ABSTRACT

ACE inhibitors or angiotensin II receptor blockers (ACEi/ARBs) have been a cornerstone of the management in kidney disease, but their use is often limited by undesired systemic effects, such as symptomatic hypotension. To minimize the extra-renal effects of ACEi/ARBs, we formulated hydrophobically modified glycol chitosan (HGC) nanomicelles releasing olmesartan (HGC-Olm) that specifically accumulated in the kidney, and investigated whether kidney-specific delivery of olmesartan by HGC nanomicelles could ameliorate organ damage in Col4a3-/- mouse, a murine model of progressive chronic kidney disease mimicking human Alport syndrome. Ex vivo tracing demonstrated that intravenously injected HGC-Olm nanomicelles were specifically delivered to the kidney, with sustained release of olmesartan for more than 48 h. Contrary to the conventional delivery of olmesartan via oral route, injection of HGC-Olm nanomicelles did not alter blood pressure in Col4a3-/- mice. Immunohistochemistry revealed that HGC nanomicelles were diffusely distributed from the cortex and glomeruli to the outer medulla, sparing the inner medulla. Phenotypic analysis showed that the attenuation of kidney fibrosis in the kidney of Col4a3-/- mice by HGC-Olm nanomicelles was comparable to that noted with conventionally delivered olmesartan. Therefore, our results suggest that HGC-Olm nanomicelles could be a safe and effective alternative drug delivery system for kidney diseases.


Subject(s)
Nephritis, Hereditary , Angiotensin Receptor Antagonists , Angiotensin-Converting Enzyme Inhibitors , Animals , Disease Models, Animal , Imidazoles , Kidney , Mice , Nephritis, Hereditary/genetics , Tetrazoles
4.
Sci Rep ; 10(1): 4225, 2020 03 06.
Article in English | MEDLINE | ID: mdl-32144368

ABSTRACT

Angiotensin-[1-7] (Ang-[1-7]) antagonize the actions of the renin-angiotensin-system via the Mas receptor and thereby exert renoprotective effects. Murine recombinant angiotensin-converting enzyme (ACE)2 was reported to show renoprotective effects in an experimental Alport syndrome model; however, the protective effect of direct administration of Ang-[1-7] is unknown. Here, we used Col4a3-/- mice as a model of Alport syndrome, which were treated with saline or Ang- [1-7]; saline-treated wild-type mice were used as a control group. The mice were continuously infused with saline or Ang-[1-7] (25 µg/kg/h) using osmotic mini-pumps. Col4a3-/- mice showed increased α-smooth muscle actin (SMA), collagen, and fibronectin expression levels, which were attenuated by Ang-[1-7] treatment. Moreover, Ang-[1-7] alleviated activation of transforming growth factor-ß/Smad signaling, and attenuated the protein expression of ED-1 and heme oxygenase-1, indicating reduction of renal inflammation. Ang-[1-7] treatment further reduced the expression levels of inflammatory cytokines and adhesion molecules and attenuated apoptosis in human kidney cells. Finally, Ang-[1-7] downregulated TNF-α converting enzyme and upregulated ACE2 expression. Thus, treatment with Ang-[1-7] altered the ACE2-Ang-[1-7]-Mas receptor axis in the kidneys of Col4a3-/- mice to attenuate the nephropathy progression of Alport syndrome.


Subject(s)
Acute Kidney Injury/drug therapy , Angiotensin I/pharmacology , Antihypertensive Agents/pharmacology , Autoantigens/physiology , Collagen Type IV/physiology , Disease Models, Animal , Inflammation/prevention & control , Nephritis, Hereditary/complications , Peptide Fragments/pharmacology , Acute Kidney Injury/etiology , Acute Kidney Injury/pathology , Animals , Apoptosis , Inflammation/etiology , Inflammation/pathology , Male , Mice , Mice, Knockout , Nephritis, Hereditary/pathology
5.
Int J Mol Sci ; 21(4)2020 Feb 21.
Article in English | MEDLINE | ID: mdl-32098220

ABSTRACT

Histone deacetylases have been a target of therapy for organ fibrosis. Here, we report the protective effect of CG200745 (CG), a novel histone deacetylase inhibitor, on tubulointerstitial fibrosis in Col4a3-/- mice, a murine model of Alport syndrome. Morphological analyses revealed CG treatment markedly alleviated kidney fibrosis in Col4a3-/- mice at the age of 7 weeks. CG prevented the activation of transforming growth factor ß (TGFß) and its downstream SMAD signaling in the kidney of Col4a3-/- mice. As critical upstream regulators of TGFß signaling, immunoblotting of whole kidney lysate of Col4a3-/- mice reveled that intra-renal renin-angiotensin system (RAS) was activated with concurrent upregulation of inflammation and apoptosis, which were effectively suppressed by CG treatment. CG suppressed both activation of RAS and up-regulation of TGFß signals in angiotensin II-stimulated HK-2 cells, a human kidney proximal tubular epithelial cell line. CG inhibited activation of TGFß-driven signals and fibrosis in NRK-49F cells, a rat kidney fibroblast cell line, under angiotensin II-rich conditions. Collectively, CG was found to be effective both in proximal tubular epithelial cells by inhibiting local RAS and TGFß signaling activation, as well as in fibroblasts by blocking their transition to myofibroblasts, attenuating renal fibrosis in a murine model of Alport syndrome.


Subject(s)
Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Kidney Tubules, Proximal/metabolism , Naphthalenes/pharmacology , Nephritis, Hereditary , Signal Transduction , Animals , Autoantigens/metabolism , Cell Line , Collagen Type IV/deficiency , Collagen Type IV/metabolism , Disease Models, Animal , Fibrosis , Humans , Kidney Tubules, Proximal/pathology , Male , Mice , Mice, Knockout , Nephritis, Hereditary/drug therapy , Nephritis, Hereditary/genetics , Nephritis, Hereditary/metabolism , Nephritis, Hereditary/pathology , Rats , Transforming Growth Factor beta/metabolism
6.
Int J Mol Sci ; 20(15)2019 Aug 06.
Article in English | MEDLINE | ID: mdl-31390839

ABSTRACT

Despite the wide use of angiotensin II receptor blockers in the treatment of Alport syndrome (AS), the mechanism as to how angiotensin II receptor blockers prevent interstitial fibrosis remains unclear. Here, we report that treatment of olmesartan effectively targets the feedback loop between the renin-angiotensin system (RAS) and transforming growth factor ß (TGFß) signals in tubular epithelial cells and preserves renal angiotensin-converting enzyme 2 (ACE2) expression in the kidney of Col4a3-/- mice, a murine model of experimental AS. Morphology analyses revealed amelioration of kidney fibrosis in Col4a3-/- mice by olmesartan treatment. Upregulation of TGFß and activation of its downstream in Col4a3-/- mice were attenuated by olmesartan in Col4a3-/- mice. Intriguingly, TGFß expression was preferentially upregulated in damaged tubular epithelial cells in Col4a3-/- mice. Concurrent upregulation of TNFα-converting enzyme and downregulation of ACE2 suggested RAS activation in Col4a3-/- mice, which was prevented by olmesartan. Mechanistically, olmesartan suppressed TGFß-induced RAS activation in tubular epithelial cells in vitro. Collectively, we concluded that olmesartan effectively suppresses the progression of tubulointerstitial fibrosis in AS by interrupting RAS-TGFß feedback loop to counterbalance intrarenal RAS activation.


Subject(s)
Antihypertensive Agents/pharmacology , Gene Expression Regulation/drug effects , Imidazoles/pharmacology , Kidney Tubules/drug effects , Kidney Tubules/metabolism , Nephritis, Hereditary/genetics , Nephritis, Hereditary/pathology , Tetrazoles/pharmacology , Transforming Growth Factor beta/genetics , Angiotensin-Converting Enzyme 2 , Animals , Apoptosis/drug effects , Biomarkers , Biopsy , Disease Models, Animal , Fibrosis , Kidney Tubules/pathology , Mice , Mice, Knockout , Nephritis, Hereditary/drug therapy , Nephritis, Hereditary/metabolism , Peptidyl-Dipeptidase A/genetics , Peptidyl-Dipeptidase A/metabolism , Transforming Growth Factor beta/metabolism , Treatment Outcome , ras Proteins/genetics , ras Proteins/metabolism
7.
Int J Mol Sci ; 20(14)2019 Jul 10.
Article in English | MEDLINE | ID: mdl-31295865

ABSTRACT

G-protein-coupled receptor 40 (GPR40) has an anti-apoptotic effect in pancreatic ß-cells. However, its role in renal tubular cell apoptosis remains unclear. To explore the role of GPR40 in renal tubular apoptosis, a two-week unilateral ureteral obstruction (UUO) mouse model was used. The protein expression of GPR40 was decreased, while the Bax/Bcl-2 protein expression ratio, the expression of tumor necrosis factor (TNF)-α mRNA, and angiotensin II type 1 receptor (AT1R) protein were increased in mice with UUO. In vitro, pretreatment of rat proximal tubular (NRK52E) cells with GW9508, a GPR40 agonist, attenuated the decreased cell viability, increased the Bax/Bcl-2 protein expression ratio, increased protein expression of cleaved caspase-3 and activated the nuclear translocation of nuclear factor-κB (NF-κB) p65 subunit induced by TNF-α treatment. TNF-α treatment significantly increased the expression of AT1R protein and the generation of reactive oxygen species (ROS), whereas GW9508 treatment markedly reversed these effects. Pretreatment with GW1100, a GPR40 antagonist, or silencing of GPR40 in NRK52E cells promoted the increased expression of the cleaved caspase-3 protein by TNF-α treatment. Our results demonstrate that decreased expression of GPR40 is associated with apoptosis via TNF-α and AT1R in the ureteral obstructed kidney. The activation of GPR40 attenuates TNF-α-induced apoptosis by inhibiting AT1R expression and ROS generation through regulation of the NF-κB signaling pathway.


Subject(s)
Acute Kidney Injury/metabolism , Apoptosis/drug effects , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/metabolism , Receptors, G-Protein-Coupled/metabolism , Tumor Necrosis Factor-alpha/metabolism , Acute Kidney Injury/drug therapy , Acute Kidney Injury/etiology , Acute Kidney Injury/pathology , Animals , Apoptosis/genetics , Biomarkers , Disease Models, Animal , Fluorescent Antibody Technique , Gene Expression , Immunohistochemistry , Kidney Tubules, Proximal/pathology , Male , Rats , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/genetics , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Ureteral Obstruction/complications , Ureteral Obstruction/genetics , Ureteral Obstruction/metabolism
8.
Sci Rep ; 9(1): 8751, 2019 06 19.
Article in English | MEDLINE | ID: mdl-31217524

ABSTRACT

Activation of the epidermal growth factor receptor (EGFR)/signal transducer and activator of transcription 3 (Stat3) signaling pathway has been reported to be associated with renal fibrosis. We have recently demonstrated that peroxiredoxin V (PrdxV) acted as an antifibrotic effector by inhibiting the activity of Stat3 in TGF-ß-treated NRK49F cells. However, the underlying mechanism of PrdxV remains poorly understood. To investigate molecular mechanism of PrdxV, we used a transgenic mouse model expressing PrdxV siRNA (PrdxVsi mice) and performed unilateral ureteral obstruction (UUO) for 7 days. 209/MDCT cells were transiently transfected with HA-tagged WT PrdxV and C48S PrdxV. Transgenic PrdxVsi mice displayed an exacerbated epithelial-to-mesenchymal transition (EMT) as well as an increase in oxidative stress induced by UUO. In the UUO kidney of the PrdxVsi mouse, knockdown of PrdxV increased Tyr1068-specific EGFR and Stat3 phosphorylation, whereas overexpression of WT PrdxV in 209/MDCT cells showed the opposite results. Immunoprecipitation revealed the specific interaction between WT PrdxV and Stat3 in the absence or presence of TGF-ß stimulation, whereas no PrdxV-EGFR or C48S PrdxV-Stat3 interactions were detected under any conditions. In conclusion, PrdxV is an antifibrotic effector that sustains renal physiology. Direct interaction between PrdxV and Stat3 through Cys48 is a major molecular mechanism.


Subject(s)
ErbB Receptors/metabolism , Kidney Diseases/metabolism , Peroxiredoxins/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , Animals , Cell Line , Cysteine/genetics , Cysteine/metabolism , ErbB Receptors/genetics , Fibrosis , Kidney Diseases/genetics , Kidney Diseases/pathology , Mice , Mice, Knockout , Peroxiredoxins/genetics , STAT3 Transcription Factor/genetics , Ureteral Obstruction/genetics , Ureteral Obstruction/metabolism , Ureteral Obstruction/pathology
9.
Int J Mol Sci ; 20(3)2019 Jan 25.
Article in English | MEDLINE | ID: mdl-30691015

ABSTRACT

The novel histone deacetylase inhibitor CG200745 was initially developed to treat various hematological and solid cancers. We investigated the molecular mechanisms associated with the renoprotective effects of CG200745 using deoxycorticosterone acetate (DOCA)-salt hypertensive (DSH) rats. DOCA strips (200 mg/kg) were implanted into rats one week after unilateral nephrectomy. Two weeks after DOCA implantation, DSH rats were randomly divided into two groups that received either physiological saline or CG200745 (5 mg/kg/day) for another two weeks. The extent of glomerulosclerosis and tubulointerstitial fibrosis was determined by Masson's trichrome staining. The renal expression of fibrosis and inflammatory markers was detected by semiquantitative immunoblotting, a polymerase chain reaction, and immunohistochemistry. Pathological signs such as glomerulosclerosis, tubulointerstitial fibrosis, increased systolic blood pressure, decreased creatinine clearance, and increased albumin-to-creatinine ratios in DSH rats were alleviated by CG200745 treatment compared to those manifestations in positive control animals. Furthermore, this treatment counteracted the increased expression of αSMA, TGF-ß1, and Bax, and the decreased expression of Bcl-2 in the kidneys of DSH rats. It also attenuated the increase in the number of apoptotic cells in DSH rats. Thus, CG200745 can effectively prevent the progression of renal injury in DSH rats by exerting anti-inflammatory, anti-fibrotic, and anti-apoptotic effects.


Subject(s)
Desoxycorticosterone Acetate/adverse effects , Histone Deacetylase Inhibitors/administration & dosage , Hydroxamic Acids/administration & dosage , Hypertension/drug therapy , Kidney Diseases/prevention & control , Naphthalenes/administration & dosage , Actins/metabolism , Albumins/metabolism , Animals , Apoptosis/drug effects , Biomarkers/metabolism , Creatinine/metabolism , Disease Models, Animal , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Hypertension/chemically induced , Hypertension/complications , Kidney Diseases/metabolism , Male , Naphthalenes/pharmacology , Random Allocation , Rats , Rats, Sprague-Dawley , Transforming Growth Factor beta1/metabolism
10.
Biol Cell ; 111(1): 18-27, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30304547

ABSTRACT

BACKGROUND INFORMATION: Tubulointerstitial fibrosis is the end-point of chronic kidney diseases. Tamoxifen, a selective oestrogen receptor (ER) modulator, attenuates renal fibrosis, by regulating the transforming growth factor (TGF)-ß/Smad signalling. Src and phosphoinositide 3-kinase (PI3K)/Akt pathways play critical roles in the pathogenesis of renal fibrosis. However, the activation of the non-canonical TGF-ß signalling in renal fibrosis after treatment with tamoxifen remains unclear. Renal fibrosis was induced by unilateral ureteral obstruction (UUO) in rats. Tamoxifen was orally administered after UUO. Additionally, HK-2 cells were treated with tamoxifen in the presence or absence of TGF-ß1. The selective ER down-regulator ICI and ER-α silencing were used to confirm the involvement of ER-α on the effect of tamoxifen on TGF-ß1-stimulated fibrosis in HK-2 cells. RESULTS: Tamoxifen treatment ameliorated UUO-induced renal fibrosis as shown by decreased expression of α-smooth muscle actin (SMA), fibronectin and connective tissue growth factor (CTGF). The phosphorylation of Src, PI3K, Akt, mammalian target of rapamycin (mTOR) and p70S6K significantly decreased in UUO kidneys from tamoxifen-treated animals. Tamoxifen dose-dependently suppressed the TGF-ß1-induced expression of α-SMA and CTGF, and phosphorylation of Src, PI3K, Akt, mTOR and p70S6K in HK-2 cells. These anti-fibrotic effects were reversed by treatment with ICI and silencing of ER-α. Moreover, inhibition of the PI3K/Akt and mTOR/p70S6K pathways was observed in HK-2 cells co-treated with PP1 (a Src kinase inhibitor) and tamoxifen. CONCLUSIONS: The anti-fibrotic effects of tamoxifen are associated with the suppression of Src kinase function via ER-α, followed by inhibition of the PI3K/Akt and mTOR/p70S6K signalling pathways. SIGNIFICANCE: Our findings suggest that tamoxifen is a novel therapeutic option for the prevention and treatment of renal fibrosis.


Subject(s)
Signal Transduction/drug effects , Tamoxifen/pharmacology , Animals , Male , Phosphatidylinositol 3-Kinases/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats, Sprague-Dawley , TOR Serine-Threonine Kinases/metabolism , src-Family Kinases/metabolism
11.
Sci Rep ; 8(1): 11546, 2018 08 01.
Article in English | MEDLINE | ID: mdl-30068917

ABSTRACT

Tubulointerstitial fibrosis is a common feature of kidney disease. Histone deacetylase (HDAC) inhibitors have been reported to attenuate renal fibrosis progression. Here, we investigated the effect of CG200745, a novel HDAC inhibitor, on renal fibrosis development in a mouse model of unilateral ureteral obstruction (UUO). To examine the effects of CG200745 on renal fibrosis in UUO, C57BL/6 J male mice were divided into three groups: control, UUO, and CG200745 (30 mg/kg/day)-treated UUO groups. CG 200745 was administered through drinking water for 1 week. Human proximal tubular epithelial (HK-2) cells were also treated with CG200745 (10 µM) with or without TGF-ß (2 ng/mL). Seven days after UUO, plasma creatinine did not differ among the groups. However, plasma neutrophil gelatinase-associated lipocalin (NGAL) levels were markedly increased in the UUO group, which were attenuated by CG200745 treatment. UUO kidneys developed marked fibrosis as indicated by collagen deposition and increased α-smooth muscle actin (SMA) and fibronectin expression. CG200745 treatment attenuated these fibrotic responses and suppressed UUO-induced production of transforming growth factor-beta1 (TGF-ß) and phosphorylation of Smad-2/3. CG200745 treatment also attenuated UUO-induced inflammation as indicated by the expression of inflammatory markers. Furthermore, CG200745 attenuated phosphorylation of p38 mitogen-activated protein kinase in UUO kidneys. In HK-2 cells, TGF-ß induced the expression of α-SMA and fibronectin, which were attenuated by CG200745 cotreatment. These results demonstrate that CG200745, a novel HDAC inhibitor, has a renoprotective effect by suppressing renal fibrosis and inflammation in a UUO mouse model.


Subject(s)
Fibrosis/drug therapy , Histone Deacetylase Inhibitors/administration & dosage , Hydroxamic Acids/administration & dosage , Naphthalenes/administration & dosage , Ureteral Obstruction/complications , Animals , Cell Line , Disease Models, Animal , Epithelial Cells/drug effects , Fibrosis/pathology , Humans , Kidney/pathology , Lipocalins/blood , Mice , Mice, Inbred C57BL , Phosphorylation , Protein Processing, Post-Translational , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Transforming Growth Factor beta/analysis , Treatment Outcome
12.
Korean J Physiol Pharmacol ; 22(2): 135-143, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29520166

ABSTRACT

Tumor necrosis factor-α (TNFα) and the angiotensin system are involved in inflammatory diseases and may contribute to acute kidney injury. We investigated the mechanisms by which TNFα-converting enzyme (TACE) contributes to lipopolysaccharide (LPS)-induced renal inflammation and the effect of TACE inhibitor treatment on LPS-induced cellular injury in human renal proximal tubule epithelial (HK-2) cells. Mice were treated with LPS (10 mg/kg, i.p.) and HK-2 cells were cultured with or without LPS (10 µg/ml) in the presence or absence of a type 1 TACE inhibitor (1 µM) or type 2 TACE inhibitor (10 µM). LPS treatment induced increased serum creatinine, TNFα, and urinary neutrophil gelatinase-associated lipocalin. Angiotensin II type 1 receptor, mitogen activated protein kinase (MAPK), and TACE increased, while angiotensin-converting enzyme-2 (ACE2) expression decreased in LPS-induced acute kidney injury and LPS-treated HK-2 cells. LPS induced reactive oxygen species and the down-regulation of ACE2, and these responses were prevented by TACE inhibitors in HK-2 cells. TACE inhibitors increased cell viability in LPS-treated HK-2 cells and attenuated oxidative stress and inflammatory cytokines. Our findings indicate that LPS activates renin angiotensin system components via the activation of TACE. Furthermore, inhibitors of TACE are potential therapeutic agents for kidney injury.

13.
Sci Rep ; 7(1): 4319, 2017 06 28.
Article in English | MEDLINE | ID: mdl-28659586

ABSTRACT

Ischemia/reperfusion injury triggers acute kidney injury (AKI) by aggravating oxidative stress mediated mitochondria dysfunction. The peroxisome proliferator-activated receptor gamma coactivator 1α (PGC-1α) is a master player that regulates mitochondrial biogenesis and the antioxidant response. We postulated that PGC-1α functions as cytoprotective effector in renal cells and that its regulation mechanism is coordinated by nuclear factor erythroid 2-related factor 2 (Nrf-2). In this study, to understand the effect and molecular mechanisms of PGC-1α, we developed an empty vector or PGC-1α-overexpressing stable cell lines in HK-2 cells (Mock or PGC-1α stable cells). PGC-1α overexpression increased the viability of cells affected by H2O2 mediated injury, protected against H2O2-mediated apoptotic events and inhibited reactive oxygen species accumulation in the cytosol and mitochondria as compared to that in Mock cells. The cytoprotective effect of PGC-1α was related to Nrf-2 upregulation, which was counteracted by Nrf-2-specific knockdown. Using inhibitor of p38, we found that regulation of the p38/glycogen synthase kinase 3ß (GSK3ß)/Nrf-2 axis was involved in the protective effects of PGC-1α. Taken together, we suggest that PGC-1α protects human renal tubule cells from H2O2-mediated apoptotic injury by upregulating Nrf-2 via GSK3ß inactivation mediated by activated p38.


Subject(s)
Glycogen Synthase Kinase 3 beta/metabolism , Hydrogen Peroxide/pharmacology , NF-E2-Related Factor 2/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Antioxidants/metabolism , Apoptosis/drug effects , Apoptosis/genetics , Cell Line , Gene Expression , Gene Expression Regulation , Kidney Diseases/etiology , Kidney Diseases/metabolism , Kidney Diseases/pathology , Male , Mice , NF-E2-Related Factor 2/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , RNA, Small Interfering/genetics , Reactive Oxygen Species , Reperfusion Injury/genetics , Reperfusion Injury/metabolism , Reperfusion Injury/pathology
14.
PLoS One ; 10(11): e0142664, 2015.
Article in English | MEDLINE | ID: mdl-26556707

ABSTRACT

BACKGROUND: Angiotensin-(1-7) [Ang-(1-7)] counteracts many actions of the renin-angiotensin-aldosterone system. Despite its renoprotective effects, extensive controversy exists regarding the role of Ang-(1-7) in obstructive nephropathy, which is characterized by renal tubulointerstitial fibrosis and apoptosis. METHODS: To examine the effects of Ang-(1-7) in unilateral ureteral obstruction (UUO), male Sprague-Dawley rats were divided into three groups: control, UUO, and Ang-(1-7)-treated UUO rats. Ang-(1-7) was continuously infused (24 µg/[kg·h]) using osmotic pumps. We also treated NRK-52E cells in vitro with Ang II (1 µM) in the presence or absence of Ang-(1-7) (1 µM), Mas receptor antagonist A779 (1 µM), and Mas receptor siRNA (50 nM) to examine the effects of Ang-(1-7) treatment on Ang II-stimulated renal injury via Mas receptor. RESULTS: Angiotensin II (Ang II) and angiotensin type 1 receptor (AT1R) protein expression was higher in UUO kidneys than in controls. Ang-(1-7) treatment also decreased proapoptotic protein expression in UUO kidneys. Ang-(1-7) also significantly ameliorated TUNEL positive cells in UUO kidneys. Additionally, Ang-(1-7) reduced profibrotic protein expression and decreased the increased tumor growth factor (TGF)-ß1/Smad signaling present in UUO kidneys. In NRK-52E cells, Ang II induced the expression of TGF-ß1/Smad signaling effectors and proapoptotic and fibrotic proteins, as well as cell cycle arrest, which were attenuated by Ang-(1-7) pretreatment. However, treatment with A779 and Mas receptor siRNA enhanced Ang II-induced apoptosis and fibrosis. Moreover, Ang II increased tumor necrosis factor-α converting enzyme (TACE) and decreased angiotensin-converting enzyme 2 (ACE2) expression in NRK-52E cells, while pretreatment with Ang-(1-7) or A779 significantly inhibited or enhanced these effects, respectively. CONCLUSION: Ang-(1-7) prevents obstructive nephropathy by suppressing renal apoptosis and fibrosis, possibly by regulating TGF-ß1/Smad signaling and cell cycle arrest via suppression of AT1R expression. In addition, Ang-(1-7) increased and decreased ACE2 and TACE expression, respectively, which could potentially mediate a positive feedback mechanism via the Mas receptor.


Subject(s)
Angiotensin I/therapeutic use , Kidney Diseases/drug therapy , Kidney/drug effects , Peptide Fragments/therapeutic use , Renin-Angiotensin System/drug effects , Angiotensin I/pharmacology , Angiotensin II/analogs & derivatives , Angiotensin II/pharmacology , Animals , Apoptosis/drug effects , Cell Line , Kidney/metabolism , Kidney/pathology , Kidney Diseases/metabolism , Kidney Diseases/pathology , Male , Peptide Fragments/pharmacology , Rats , Rats, Sprague-Dawley , Receptor, Angiotensin, Type 1/metabolism , Signal Transduction/drug effects , Transforming Growth Factor beta1/metabolism
15.
Clin Exp Nephrol ; 19(1): 82-91, 2015 Feb.
Article in English | MEDLINE | ID: mdl-24643788

ABSTRACT

BACKGROUND: Kidney is one of the major target organs in sepsis, while effective prevention of septic acute kidney injury has not yet been established. α-Lipoic acid (LA) has been known to exert beneficial effects against lipopolysaccharide (LPS)-induced damages in various organs such as heart, lung, and liver. We investigated the protective effect of LA on LPS-induced kidney injury. METHODS: Two groups of rats were treated with LPS (20 mg/kg, i.p.), one of which being co-treated with LA (50 mg/kg), while the control group was treated with vehicle alone. Human renal proximal tubular epithelial cells (HK-2 cells) were cultured with or without LPS (10 µg/ml) in the presence or absence of LA (100 µg/ml) for 3 h prior to LPS treatment. RESULTS: Serum creatinine level was increased in LPS-treated rats, which was attenuated by LA co-treatment. LPS treatment induced cleaved caspase-3 expression in the kidney, which was counteracted by LA. Terminal deoxynucleotidyl transferase dUTP nick-end labeling-positive cells increased in the kidneys of LPS-treated rats compared with controls, which was counteracted by LA treatment. Protein expression of inducible nitric oxide synthase and cyclooxygenase-2 detected by immunoblotting and/or immunohistochemical staining, along with mRNA levels of pro-inflammatory cytokines detected by real-time polymerase chain reaction, was increased in the kidney with LPS administration, which was ameliorated with LA treatment. LA also protected LPS-induced tubular dysfunction, preserving type 3 Na(+)/H(+) exchanger and aquaporin 2 expressions in the kidney. Suppression of LPS-induced expression of cleaved caspase-3 by LA was also observed in HK-2 cells. Increased protein expression of phospho-extracellular signal-regulated kinases 1/2 and c-Jun N-terminal kinases by LPS treatment was attenuated by LA pretreatment, while p38 was not affected by either LPS or LA treatment. MitoTracker Red demonstrated LA prevented LPS-induced increment of mitochondrial oxidative stress, where concurrent 4',6-diamidino-2-phenylindole staining also revealed marked fragmentation and condensation of nuclei in HK-2 cells treated with LPS, which was prevented by LA. CONCLUSION: LA treatment attenuates LPS-induced kidney injury, such as renal tubular dysfunction, by suppression of apoptosis, and inflammation.


Subject(s)
Acute Kidney Injury/chemically induced , Acute Kidney Injury/prevention & control , Antioxidants/pharmacology , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/toxicity , Thioctic Acid/pharmacology , Acute Kidney Injury/pathology , Animals , Apoptosis/drug effects , Caspase 3/biosynthesis , Cell Line , Creatinine/blood , Humans , Ion Channels/drug effects , Kidney Function Tests , Mitogen-Activated Protein Kinases/metabolism , Oxidative Stress/drug effects , Rats
16.
PLoS One ; 9(1): e86553, 2014.
Article in English | MEDLINE | ID: mdl-24475141

ABSTRACT

The farnesoid X receptor (FXR) is mainly expressed in liver, intestine and kidney. We investigated whether 6-ethyl chenodeoxycholic acid (6ECDCA), a semisynthetic derivative of chenodeoxycholic aicd (CDCA, an FXR ligand), protects against kidney injury and modulates small heterodimer partner (SHP) in cisplatin-induced kidney injury. Cisplatin inhibited SHP protein expression in the kidney of cisplatin-treated mice and human proximal tubular (HK2) cells; this effect was counteracted by FXR ligand. Hematoxylin and eosin staining revealed the presence of tubular casts, obstructions and dilatations in cisplatin-induced kidney injury, which was attenuated by FXR ligand. FXR ligand also attenuated protein expression of transforming growth factor-ß1 (TGF-ß1), Smad signaling, and the epithelial-to-mesenchymal transition process, inflammatory markers and cytokines, and apoptotic markers in cisplatin-treated mice. Cisplatin induced NF-κB activation in HK2 cell; this effect was attenuated by pretreatment with FXR ligand. In SHP knockdown by small interfering RNA, cisplatin-induced activation of TGF-ß1, p-JNK and Bax/Bcl-2 ratio was not attenuated, while SHP overexpression and FXR ligand inhibited expression of these proteins in cisplatin-pretreated HK2 cells. In conclusion, FXR ligand, 6ECDCA prevents cisplatin-induced kidney injury, the underlying mechanism of which may be associated with anti-fibrotic, anti-inflammatory, and anti-apoptotic effects through SHP induction.


Subject(s)
Acute Kidney Injury/chemically induced , Acute Kidney Injury/prevention & control , Chenodeoxycholic Acid/analogs & derivatives , Cisplatin/adverse effects , Receptors, Cytoplasmic and Nuclear/metabolism , Analysis of Variance , Animals , Chenodeoxycholic Acid/metabolism , Chenodeoxycholic Acid/pharmacology , DNA Primers/genetics , Epithelial-Mesenchymal Transition/drug effects , Immunoblotting , Immunoenzyme Techniques , In Situ Nick-End Labeling , Male , Mice , Mice, Inbred C57BL , RNA Interference , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Signal Transduction/drug effects , Transforming Growth Factor beta1/metabolism
17.
J Renin Angiotensin Aldosterone Syst ; 15(4): 348-61, 2014 Dec.
Article in English | MEDLINE | ID: mdl-23420778

ABSTRACT

This study aimed to examine the protective effects of aliskiren on gentamicin-induced nephropathy. Rats were injected with gentamicin (100 mg/kg per day) for 14 days. Aliskiren was infused for two weeks. Human proximal tubular epithelial cell lines (HK-2) were cultured with gentamicin in the absence or presence of aliskiren. Inflammatory profibrotic and apoptotic markers were evaluated in vivo and in vitro. Aliskiren treatment attenuated the decreased creatinine clearance, increased fractional sodium excretion, glomerulosclerosis and tubulointerstitial fibrosis and counteracted the increased ED-1 expression in gentamicin-treated rats. The levels of inflammatory cytokines (TNF-α, IL-1ß and IFN-γ) and adhesion molecules (MCP-1, ICAM-1 and VCAM-1) increased in the gentamicin-treated kidneys. These changes were restored by aliskiren co-treatment. Aliskiren effectively reversed transforming growth factor-ß-induced fibrotic responses such as induction of α-smooth muscle actin in gentamicin-treated rat kidneys. Along with these changes, aliskiren also attenuated the increase in nuclear factor κB and phosphorylated extracellular signal-regulated kinase (pERK 1/2) levels in HK-2 cells cultured with gentamicin. In addition, aliskiren decreased the number of TUNEL-positive nuclei and reduced the expression of proapoptotic markers in gentamicin-treated HK-2 cells. These findings suggest that aliskiren attenuates gentamicin-induced nephropathy by suppression of inflammatory, profibrotic and apoptotic factors through inhibition of the nuclear factor κB, Smads and mitogen-activated protein kinase signaling pathways.


Subject(s)
Amides/therapeutic use , Fumarates/therapeutic use , Gentamicins/adverse effects , Kidney Diseases/chemically induced , Kidney Diseases/drug therapy , Protective Agents/therapeutic use , Renin/antagonists & inhibitors , Amides/pharmacology , Animals , Apoptosis/drug effects , Cell Adhesion Molecules/metabolism , Cytokines/genetics , Cytokines/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibrosis , Fumarates/pharmacology , Humans , Immunoblotting , Inflammation Mediators/metabolism , Kidney/drug effects , Kidney/enzymology , Kidney/pathology , Kidney/physiopathology , Kidney Diseases/pathology , Kidney Diseases/physiopathology , Kidney Function Tests , Kidney Tubules/drug effects , Kidney Tubules/pathology , Male , NF-kappa B/metabolism , Protective Agents/pharmacology , Proton-Translocating ATPases/metabolism , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Receptors, Cell Surface/metabolism , Renin/metabolism , Smad Proteins/metabolism , Transforming Growth Factor beta1/metabolism , Vacuolar Proton-Translocating ATPases , bcl-2-Associated X Protein/metabolism
18.
Korean J Physiol Pharmacol ; 17(5): 435-40, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24227945

ABSTRACT

While the anti-apoptotic effect of paricalcitol has been demonstrated in various animal models, it is not yet clear whether paricalcitol attenuates the apoptosis in gentamicin (GM)-induced kidney injury. We investigated the effect of paricalcitol on apoptotic pathways in rat kidneys damaged by GM. Rats were randomly divided into three groups: 1) Control group (n=8), where only vehicle was delivered, 2) GM group (n=10), where rats were treated with GM (150 mg/kg/day) for 7 days, 3) PARI group (n=10), where rats were co-treated with paricalcitol (0.2 µg/kg/day) and GM for 7 days. Paricalcitol attenuated renal dysfunction by GM administration in biochemical profiles. In terminal deoxynucleotidyl transferase dUTP nick end labeling staining, increased apoptosis was observed in GM group, which was reversed by paricalcitol co-treatment. Immunoblotting using protein samples from rat cortex/outer stripe of outer medulla showed increased Bax/Bcl-2 ratio and cleaved form of caspase-3 in GM group, both of which were reversed by paricalcitol. The phosphorylated Jun-N-terminal kinase (JNK) expression was increase in GM, which was counteracted by paricalcitol. The protein expression of p-Akt and nitro-tyrosine was also enhanced in GM-treated rats compared with control rats, which was reversed by paricalcitol co-treatment. Paricalcitol protects GM-induced renal injury by antiapoptotic mechanisms, including inhibition of intrinsic apoptosis pathway and JNK.

19.
Kidney Blood Press Res ; 36(1): 248-57, 2012.
Article in English | MEDLINE | ID: mdl-23171857

ABSTRACT

BACKGROUND/AIMS: Sildenafil, the first selective phosphodiesterase-5 (PDE5) inhibitor to be widely used for treating erectile dysfunction, has been investigated with regard to its cardioand renoprotective effects in animal models. This study further investigated the renoprotective effects of sildenafil and their molecular mechanisms in deoxycorticosterone acetate (DOCA)-salt hypertensive (DSH) rats. METHODS: DOCA strips (200 mg/kg) were implanted in rats 1 week after unilateral nephrectomy. These rats were fed on a control diet, with or without sildenafil (50 mg·kg(-1)day(-1)), for 2 weeks. Systolic blood pressure (SBP) was measured by the tail cuff method, and the urinary albumin-to-creatinine ratio (ACR) was calculated. The extent of glomerulosclerosis and tubulointerstitial fibrosis was determined by Masson's trichrome stain. Renal expression of ED-1, transforming growth factor-ß1 (TGF-ß1), Bax, and Bcl-2 were determined by semiquantitative immunoblotting, polymerase chain reaction (PCR), and immunohistochemistry. TUNEL staining was used for detecting apoptotic cells. RESULTS: The increased SBP in DSH rats was not attenuated by sildenafil treatment. The decreased creatinine clearance and increased ACR in DSH rats, compared with control animals, were attenuated by sildenafil treatment. Further, sildenafil treatment attenuated glomerulosclerosis and tubulointerstitial fibrosis in DSH rats and counteracted the increased expression of ED-1, TGF-ß1, and Bax and the decreased expression of Bcl-2 in the kidneys of these rats. The increase in the number of apoptotic cells in DSH rats was attenuated by sildenafil treatment. CONCLUSION: Sildenafil effectively prevented the progression of renal injury in DSH rats via its anti-inflammatory, antifibrotic, and antiapoptotic effects.


Subject(s)
Desoxycorticosterone/adverse effects , Hypertension/chemically induced , Hypertension/drug therapy , Kidney Diseases/prevention & control , Kidney/metabolism , Kidney/pathology , Piperazines/therapeutic use , Sulfones/therapeutic use , Animals , Apoptosis/drug effects , Apoptosis/physiology , Blood Pressure/drug effects , Blood Pressure/physiology , Disease Models, Animal , Disease Progression , Hypertension/complications , Kidney/drug effects , Kidney Diseases/metabolism , Kidney Diseases/pathology , Male , Phosphodiesterase 5 Inhibitors/pharmacology , Phosphodiesterase 5 Inhibitors/therapeutic use , Piperazines/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Purines/pharmacology , Purines/therapeutic use , Rats , Rats, Sprague-Dawley , Sildenafil Citrate , Sulfones/pharmacology , Transforming Growth Factor beta1/metabolism , bcl-2-Associated X Protein/metabolism
20.
Korean J Physiol Pharmacol ; 15(5): 273-7, 2011 Oct.
Article in English | MEDLINE | ID: mdl-22128259

ABSTRACT

Nitric oxide (NO) and atrial natriuretic peptide (ANP) may induce vascular relaxation by increasing the production of cyclic guanosine monophosphate (cGMP), an important mediator of vascular tone during sepsis. This study aimed to determine whether regulation of NO and the ANP system is altered in lipopolysaccharide (LPS)-induced kidney injury. LPS (10 mg.kg(-1)) was injected in the tail veins of male Sprague-Dawley rats; 12 hours later, the kidneys were removed. Protein expression of NO synthase (NOS) and neutral endopeptidase (NEP) was determined by semiquantitative immunoblotting. As an index of synthesis of NO, its stable metabolites (nitrite/nitrate, NOx) were measured using colorimetric assays. mRNA expression of the ANP system was determined by real-time polymerase chain reaction. To determine the activity of guanylyl cyclase (GC), the amount of cGMP generated in response to sodium nitroprusside (SNP) and ANP was calculated. Creatinine clearance decreased and fractional excretion of sodium increased in LPS-treated rats compared with the controls. Inducible NOS protein expression increased in LPS-treated rats, while that of endothelial NOS, neuronal NOS, and NEP remained unchanged. Additionally, urinary and plasma NOx levels increased in LPS-treated rats. SNP-stimulated GC activity remained unchanged in the glomerulus and papilla in the LPS-treated rats. mRNA expression of natriuretic peptide receptor (NPR)-C decreased in LPS-treated rats, while that of ANP and NPR-A did not change. ANP-stimulated GC activity reduced in the glomerulus and papilla. In conclusion, enhancement of the NO/cGMP pathway and decrease in ANP clearance were found play a role in the pathogenesis of LPS-induced kidney injury.

SELECTION OF CITATIONS
SEARCH DETAIL
...