1,25-(OH)2D3 and its analogue BXL-628 inhibit high glucose-induced activation of RhoA/ROCK pathway in HK-2 cells

  • Authors:
    • Wei Zhang
    • Bin Yi
    • Ke Zhang
    • Aimei Li
    • Shikun Yang
    • Jing Huang
    • Jishi Liu
    • Hao Zhang
  • View Affiliations

  • Published online on: March 9, 2017     https://doi.org/10.3892/etm.2017.4211
  • Pages: 1969-1976
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

It has previously been reported that 1,25-(OH)2D3 inhibits high glucose-induced epithelial-to-mesenchymal transition (EMT) in HK-2 cells. However, the mechanism of this renoprotective action remains unclear. Elocalcitol (BXL‑628), a vitamin D analog, has been suggested to be effective on the RhoA/Rho associated protein kinase (ROCK) pathway, which serves a crucial role in high glucose‑induced EMT. The aim of the present study was to investigate the effect of 1,25‑(OH)2D3 and its analogue BXL‑628 on high glucose‑induced activation of the RhoA/ROCK pathway in human renal proximal tubular cells. HK‑2 cells were co‑treated with high glucose and either 1,25‑(OH)2D3 or BXL‑628. The RhoA expression levels and ROCK activity of the membrane were assessed via western blot analysis or immunofluorescence. α-smooth muscle actin (α‑SMA) and epithelial (E)‑cadherin were detected using western blotting and reverse transcription-quantitative polymerase chain reaction (RT‑qPCR), whereas collagen I and fibronectin levels were measured by ELISA and RT‑qPCR. The results demonstrated that 1,25‑(OH)2D3 and BXL‑628 both significantly downregulated the expression of active RhoA and ROCK activity induced by high glucose (P<0.05). Furthermore, the expressions of α‑SMA, collagen I, and fibronectin were significantly downregulated at both protein and mRNA (P<0.05) levels, whereas the expression of E‑cadherin was significantly increased (P<0.05) by 1,25‑(OH)2D3 or BXL‑628 treatment. In conclusion, the vitamin D receptor agonist 1,25‑(OH)2D3 and its analogue BXL‑628 were both able to attenuate high glucose‑induced EMT and extracellular matrix accumulation of HK‑2 cells by suppressing the RhoA/ROCK signaling pathway in vitro.

Introduction

Diabetic nephropathy (DN), a major microvascular complication of diabetes mellitus, is the leading cause of end-stage renal disease (ESRD) in the western world and the second most common cause of renal failure with increasing morbidity in China (1). Although maintenance of normoglycemia and therapeutic intervention in the renin-angiotensin-aldosterone system are effective treatments (2,3), these interventions only partially delay the progression of DN (4); therefore, identifying new therapeutic strategies remains of great importance. The pathogenesis of DN is complicated; renal fibrosis is the primary pathophysiologic process associated with chronic renal failure caused by DN (5). Epithelial-to-mesenchymal transition (EMT) and extracellular matrix (ECM) accumulation under pathological conditions also occur in mature tubular epithelial cells of the adult kidney, and have been implicated in renal interstitial fibrosis (6,7). It has previously been indicated that a large proportion of interstitial fibrosis cases originate from tubular epithelial cells via EMT in diseased kidneys (8). However, the underlying mechanisms for this remain to be elucidated.

RhoA, which is a member of the Rho guanosine triphosphatases (GTPases) family, is essential in the regulation of cellular functions (9). It cycles between an inactive GDP-bound and an active GTP-bound form, and its intrinsic hydrolytic activity is affected by various Rho regulators (10). Membrane localization via post-translational modification is required for RhoA activation (9). Previous studies have demonstrated that RhoA and its downstream kinase Rho-kinase (ROCK) are able to mediate matrix elaboration via mesangial cells (MCs) in hyperglycemic and DN conditions (4,11), and the RhoA/ROCK signaling pathway has been demonstrated to be associated with fibrosis progression in multiple organs including the kidneys, liver and lungs (4,9,12). Furthermore, RhoA/ROCK was previously reported to be associated with EMT (7,13), which was prevented by inhibition of RhoA activation in rat peritoneal cells (13). A previous study by the present authors also revealed that transforming growth factor (TGF)-β1-mediated RhoA/ROCK activation contributed to the dissolution of tight junctions during EMT in HK-2 cell in vitro (14).

In recent years, the protective effect of vitamin D (VitD)/VitD receptor (VDR) in patients with DN has been illustrated (15,16). Clinical treatment with active VitD is typically accompanied by hypercalcemic side effects (17), therefore, VitD analogs with fewer side effects may provide therapeutic benefits. A previous clinical study demonstrated that treatment with paricalcitol, which is a VitD analog, was able to safely reduce residual albuminuria in patients with DN (18). Treatment with 1,25-(OH)2D3 (also known as calcitriol) or its analogs is able to repress the expression of α-SMA and collagen I in a unilateral ureteral occlusion model and cultured HK-2 cells (19,20). The renoprotective role of 1,25-(OH)2D3 and its analogs have been demonstrated to be associated with anti-inflammation, renin-angiotensin system (RAS) inhibition and prevention of EMT (17,2022). By binding to the VDR, 1,25-(OH)2D3 recruits cofactors to form a transcriptional complex which subsequently binds to VitD response elements in the promoter region of target genes, altering transcriptional events within target cells (17,20,23). However, the underlying mechanism of this renoprotective effect remains largely unknown at present.

Morelli et al (24) illustrated that treatment with elocalcitol (BXL-628), which is a VitD analog and VDR agonist, impaired RhoA membrane translocation and inhibited RhoA/ROCK activation in bladder smooth muscle cells, repressing RhoA-mediated cell migration and cytoskeleton remodeling (24,25). However, whether the anti-EMT and anti-fibrosis role of 1,25-(OH)2D3 and analogs is associated with the RhoA/ROCK signaling pathway remains to be elucidated.

The aim of the present study was to investigate the effect of the endogenous VDR ligand 1,25-(OH)2D3 and its analog BXL-628 on high glucose-induced activation of the RhoA/ROCK pathway in human renal proximal tubular cells. The results of the preset study may potentially elucidate a mechanism of the protective effect of VitD in DN.

Materials and methods

Cell culture and treatment

Human renal proximal tubular epithelial cells, HK-2 (ATCC, Manassas, VA, US), were cultured in Dulbecco's modified Eagle's medium supplemented with 10% fetal bovine serum (Thermo Fisher Scientific, Inc., Waltham, MA, USA), streptomycin (100 µg/ml) and penicillin (100 U/ml) in a humidified atmosphere containing 5% CO2 at 37°C. When cells reached 80% confluence, they were treated with high glucose (HG group; 30 mmol/l; Henyang Kaixin Chemical Reagent Company, Ltd., Hunan, China) for 0, 0.5, 1, 2, 4, 8 and 12 h to verify whether high glucose conditions activated RhoA. To determine the time at which high glucose induced EMT, HK-2 cells were treated with high glucose for 0, 12, 24, 48 and 72 h at 37°C respectively; mannitol (30 mmol/l; Henyang Kaixin Chemical Reagent Company, Ltd.) was used as negative control. For further analysis, HK-2 cells were cultured in high glucose medium (supplemented DMEM with 30 mmol/l glucose) at 37°C, and co-treated with 1,25-(OH)2D3 (VD3 group; 100 nM; Sigma-Aldrich; Merck KGaA, Darmstadt, Germany), or BXL-628 (BXL group; 10 nM; Molecular Devices, LLC, Sunnyvale, CA, USA) at 2 or 48 h, respectively, according to the results of the previous experiment. Low glucose (5.6 mmol/l) was used as the normal control (NC) group.

Western blotting

Total protein was extracted from indicated cells using radioimmunoprecipitation assay lysis buffer (Beyotime Institute of Biotechnology, Haimen, China), and membrane protein was extracted using a Plasma Membrane Protein Extraction kit (BioVision, Inc., Milpitas, CA, USA) according to the manufacturer's protocol. A total of 50 µg total protein or membrane protein was separated by 10% SDS-PAGE and transferred onto polyvinylidene fluoride membranes. Membranes were blocked for 1 h at room temperature with 5% non-fat milk, subsequently rinsed and incubated overnight at 4°C with anti-RhoA (1:1,000; #2117; Cell Signaling Technology, Inc., Danvers, MA, USA), anti-α-SMA (1:3,000; sc-32251; Santa Cruz Biotechnology, Inc., Dallas, TX, USA), anti-epithelial (E)-cadherin (1:3,000; #3195; Cell Signaling Technology, Inc.), anti-ATPase Na+/K+ (1:4,000; ab7671; Abcam, Cambridge, MA, USA), anti-phosphorylated-myosin-binding subunit (MBS) (1:1,000; sc-514261; Santa Cruz Biotechnology, Inc.), anti-ROCK (1:1,000; sc-17794; Santa Cruz Biotechnology, Inc.) and anti-β-actin antibodies (1:5,000; PA1-183; Invitrogen; Thermo Fisher Scientific, Inc.). Membranes were subsequently washed with TBST and incubated with the appropriate secondary antibody (goat anti-rabbit or goat anti-mouse; CW0114S and CW0102S, respectively; 1:3,000; Kangwei Biotech Company, Beijing, China) for 2 h at room temperature. Enhanced chemiluminescence reagent (Pierce; Thermo Fisher Scientific, Inc.) was used to detect the signal on the membrane. Data was analyzed using densitometry with Image-Pro plus software 6.0 (Media Cybernetics, Inc., Rockville, MD, USA) and normalized to β-actin or ATPase Na+/K+ expression.

Immunofluorescence

Cells were fixed with 4% paraformaldehyde (pH 7.4) at room temperature for 30 min, permeabilized for 10 min with PBS containing 0.1% Triton X-100, rinsed with PBS and subsequently incubated with blocking buffer (normal goat serum; CW0130S; Kangwei Biotech Company) at room temperature for 30 min. Immunostaining was performed as previously described (24) using anti-pan-cadherin (1:100; ab195203; Abcam) and anti-RhoA antibodies (1:100; ab54835; Abcam), followed by tetramethylrhodamine-conjugated AffiniPure (red) goat anti-mouse (1:200; SA00007-1; Proteintech Group, Inc., Chicago, IL, USA) and fluorescein isothiocyanate-conjugated AffiniPure (green) goat anti-rabbit (1:200; SA00003-2; Proteintech Group, Inc.) antibodies, respectively. A fluorescent microscope (BX61 Automated Fluorescent Microscope; Olympus Corporation, Tokyo, Japan) was use to observe the cells.

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

Total mRNA was extracted using TRIzol reagent (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's protocol. A total of 2 µg RNA was used for cDNA synthesis with a ReverTra Ace qPCR RT kit (Toyobo Co., Ltd., Osaka, Japan) according to the manufacturer's protocol. Total RNA was treated with DNase I prior to transcription according the kit manual. A total reaction volume of 20 µl was used for PCR with a Taq DNA polymerase-based 2x master mix for real-time PCR (SYBR®-Green Real-time PCR Master Mix, Toyobo Co., Ltd.) using the iQ5 real-time PCR system (Bio-Rad Laboratories, Inc., Hercules, CA, USA). The sequence detector was programmed for the following PCR conditions: 50°C for 2 min, 95°C for 10 min, 40 cycles at 95°C for 15 sec and 60°C for 1 min. The relative expressions of α-SMA, collagen I, fibronectin and E-cadherin mRNA were expressed relative to β-actin as an internal control, using the 2−ΔΔCq method (26). Oligo 6.0 software (Molecular Biology Insights, Inc., Cascade, CO, USA) was used to design PCR primers (Table I), which were synthesized by Shanghai Sangong Pharmaceutical Co., Ltd. (Shanghai, China).

Table I.

Primers used for reverse transcription-quantitative polymerase chain reaction.

Table I.

Primers used for reverse transcription-quantitative polymerase chain reaction.

mRNAPrimer sequencesPrimer length (bp)
α-SMAForward: 5′-TTCAATGTCCCAGCCATGTA-3′125
Reverse: 5′-GAAGGAATAGCCACGCTCAG-3′
E-cadherinForward: 5′-TGCCCAGAAAATGAAAAAGG-3′120
Reverse: 5′-GTGTATGTGGCAATGCGTTC-3′
Collagen IForward: 5′-CCAAATCTGTCTCCCCAGAA-3′136
Reverse: 5′-TCAAAAACGAAGGGGAGATG-3′
FibronectinForward: 5′-GCTTCCTGGCACTTCTGGTC-3′136
Reverse: 5′-CTACATTCGGCGGGTATGGT-3′
β-actinForward: 5′-CATCCTGCGTCTGGACCTGG-3′107
Reverse: 5′-TAATGTCACGCACGATTTCC-3′

[i] SMA, smooth muscle actin; E, epithelial.

ELISA assay

The concentrations of collagen I and fibronectin secreted from cultured cells in medium were determined using an ELISA kits (CSB-E13445 h and CSB-EL005721HU; Cusabio Biotech Co., Ltd., Wuhan, China) according to the manufacturer's protocol. The final absorbance at 450 nm was measured using TECAN Infinite M200 microplate reader (Thermo Fisher Scientific, Inc.). Concentrations of collagen I and fibronectin were calculated using a standard curve constructed in the same plate, and expressed relative to the cell protein concentration.

Statistical analysis

All experiments were performed at least 3 times. GraphPad Prism 5.0 software (GraphPad Software, Inc., La Jolla, CA, USA) was used for the analysis and graphics presentation. One-way analysis of variance followed by Tukey's multiple comparisons test or Student's t-test was used depending on the experimental condition. The data were expressed as mean ± standard error of the mean. P<0.05 was considered to indicate a statistically significant difference.

Results

1,25-(OH)2D3 and BXL-628 decreases RhoA activation and ROCK activity in HK-2 cells

Using western blot analysis, it was demonstrated that high glucose conditions induced RhoA activation in a time-dependent manner, which was significantly increased from 1 to 8 h (P<0.01) compared with the control group, reaching a peak at 2 h and returning to the control level at 12 h post-administration (Fig. 1A). The effects of 1,25-(OH)2D3 and its analog BXL-628 on HK-2 cells treated with high glucose for 2 h were also investigated. As illustrated in Fig. 1B, active RhoA protein expression in the HG group was significantly upregulated compared with the NC group (P<0.05), and treatment with 1,25-(OH)2D3 or BXL-628 significantly decreased the expression of active RhoA protein (both P<0.05). Additionally, ROCK activity was evaluated by measuring the phospho-MBS level (Fig. 1C). It was revealed that ROCK activity was significantly lower in the VD3 and BXL groups compared with the high glucose group (P<0.01).

Indirect immunofluorescence also revealed that expression of active RhoA protein was markedly reduced in the VD3 and BXL groups compared with the NC group (Fig. 2). The intracellular localization of RhoA was monitored by staining with a specific monoclonal antibody and compared with pan-cadherin immune reactivity as a cell membrane marker. The merged images obtained by dual labeling of RhoA and pan-cadherin immune staining confirmed that high glucose stimulation increased RhoA localization at the plasma membrane, whereas treatment with 1,25-(OH)2D3 or BXL-628 treatment reduced RhoA membrane expression.

Effects of 1,25-(OH)2D3 and BXL-628 on the expression of E-cadherin and α-SMA

As shown in Fig. 3, high glucose stimulation resulted in a significant time-dependent decrease in E-cadherin protein levels (P<0.05 at 12 h, P<0.01 from 24–72 h; Fig. 3A) and mRNA (P<0.01; Fig. 3B) compared with the NC group, whereas high glucose stimulation resulted in a significant time-dependent increase in α-SMA at both the protein and mRNA levels (both P<0.05 at 12 h, P<0.01 from 24–72 h; Fig. 3C-D). Treatment with either 1,25-(OH)2D3 or BXL-628 attenuated the downregulation in E-cadherin expression at the protein (P<0.05; Fig. 4A) and mRNA levels (P<0.01; Fig. 4B) compared with the HG group. Both 1,25-(OH)2D3 and BXL-628 treatments significantly inhibited the expression of α-SMA at the protein (P<0.05; Fig. 4C) and mRNA (P<0.01; Fig. 4D) levels compared with the HG group (Fig. 4B and D).

Effects of 1,25-(OH)2D3 and BXL-628 on the expression of ECM in HK-2 cells

To further analyze the effects of 1,25-(OH)2D3 and its analog BXL-628 on ECM accumulation induced by high glucose, the expressions of collagen I and fibronectin in the culture supernatant were detected using ELISA. Cellular mRNA expression was evaluated using qPCR. The results indicated that high glucose stimulation significantly increased the expression of collagen I protein (P<0.05; Fig. 5A) and mRNA (P<0.01; Fig. 5B) compared with the NC group. High glucose stimulation also induced a significant increase in fibronectin protein (P<0.05; Fig. 5C) and mRNA (P<0.01; Fig. 5D) compared with the NC groups. Notably, both 1,25-(OH)2D3 and BXL-628 significantly decreased the expression of collagen I and fibronectin compared with the HG group (P<0.01 for collagen protein, collagen mRNA and fibronectin mRNA; P<0.05 for fibronectin protein; Fig. 5A-D).

Discussion

The present study demonstrated that 1,25-(OH)2D3 and its analog BXL-628 contribute to renoprotection by preventing high glucose-induced EMT and ECM accumulation via inactivating the RhoA/ROCK signaling pathway in vitro. Hyperglycemia is one of the most important risk factors in diabetes and its complications, including DN (1,5). Hyperglycemic conditions activate a number of important intracellular signaling pathways, including the TGF-β/SMAD, nuclear factor-κB and Rho/ROCK pathways (12,15,27,28), which serve crucial roles in the diabetic renal inflammation response, RAS activation, EMT and fibrosis (3,8,19,29). VitD and VDR have previously been demonstrated to have renal protective functions via their regulative role in pathways associated with the prevention of EMT and fibrosis (17,20,30,31). Peng et al (4) illustrated that high glucose was able to activate RhoA/Rho-kinase in MCs, leading to downstream activator protein-1 activation and fibronectin induction (4). In accordance with the studies mentioned above, the results of the present study also demonstrated that high glucose-induced activation of the RhoA/ROCK pathway subsequently resulted in EMT and ECM accumulation in HK-2 cells. Furthermore, treatment with 1,25-(OH)2D3 or BXL-628 significantly inactivated the RhoA/ROCK pathway and attenuated high glucose-induced EMT and ECM accumulation in HK-2 cells. The present study found that 1,25-(OH)2D3 and BXL-628 treatment was able to decrease the high glucose-stimulated upregulation of α-SMA and downregulation of E-cadherin. Additionally, fibronectin and collagen I accumulation were significantly suppressed in both the VD3 and BXL groups compared with the HG group.

VDR acts as a transcriptional factor and a nongenomic activator of the RhoA/ROCK and p38MAPK/mitogen and stress activated kinase (MSK)1 pathways, which are required for the biofunction of 1,25-(OH)2D3 in colon carcinoma cells (32). By binding to VDR, 1,25-(OH)2D3 induces an influx of Ca2+ into cytoplasm and subsequently activates the RhoA/ROCK and p38MAPK-MSK1 kinase pathways (32). This indicates that the rapid modulation of ion content and cytosolic GTPases and kinases is associated with EMT and ECM accumulation (33,34). However, the present study demonstrated that 1,25-(OH)2D3 acts as an inhibitor of RhoA/ROCK and its role may depend on cell types. In accordance with the results of the present study, it has previously been demonstrated that BXL-628 is able to inhibit RhoA activation in a dose-dependent manner without altering RhoA expression, and ameliorates excessive hyperplasia and aberrant differentiation in bladder smooth muscle cells (24,25). In the present study, 1,25-(OH)2D3 exerted a similar effect to BXL-628 at a much higher concentration. This suggests that BXL-626 may be a more efficient pharmaceutical choice, although further beneficial value must be investigated through more clinical studies and fundamental research. 1,25-(OH)2D3 and BXL-628 do not alter the expression of RhoA and ROCK, however they do increase their membrane translocation and binding to GTP (24,35).

In conclusion, the results of the present study demonstrate that 1,25-(OH)2D3 and BXL-628 treatment repress the high glucose-activated RhoA/ROCK pathway in HK-2 cells. Additionally, activation of the VitD/VDR pathway by treatment with 1,25-(OH)2D3 or BXL-628 decreased the expression of α-SMA, collagen I and fibronectin, and increased E-cadherin expression; this is suggestive of an anti-EMT and anti-fibrosis role of 1,25-(OH)2D3 and BXL-628. These results indicate that the renal protective effects of 1,25-(OH)2D3 and its analog are mediated, at least in part, by the RhoA/ROCK signaling pathway.

Acknowledgements

The present study was supported by the National Natural Science Foundation of China (grant no. 81470961), the Natural Science Foundation of Hunan Province (grant no. 14JJ2037) and the New Xiangya Talent Project of the Third Xiangya Hospital of Central South University (grant no. 20150313).

References

1 

Xu Y, Wang L, He J, Bi Y, Li M, Wang T, Wang L, Jiang Y, Dai M, Lu J, et al: Prevalence and control of diabetes in Chinese adults. JAMA. 310:948–959. 2013. View Article : Google Scholar : PubMed/NCBI

2 

No authors listed: Retinopathy and nephropathy in patients with type 1 diabetes four years after a trial of intensive therapy. The diabetes control and complications trial/epidemiology of diabetes interventions and complications research group. N Engl J Med. 342:381–389. 2000. View Article : Google Scholar : PubMed/NCBI

3 

Lewis EJ, Hunsicker LG, Bain RP and Rohde RD: The effect of angiotensin-converting-enzyme inhibition on diabetic nephropathy. The collaborative study group. N Engl J Med. 329:1456–1462. 1993. View Article : Google Scholar : PubMed/NCBI

4 

Peng F, Wu D, Gao B, Ingram AJ, Zhang B, Chorneyko K, McKenzie R and Krepinsky JC: RhoA/Rho-kinase contribute to the pathogenesis of diabetic renal disease. Diabetes. 57:1683–1692. 2008. View Article : Google Scholar : PubMed/NCBI

5 

Afkarian M, Sachs MC, Kestenbaum B, Hirsch IB, Tuttle KR, Himmelfarb J and de Boer IH: Kidney disease and increased mortality risk in type 2 diabetes. J Am Soc Nephrol. 24:302–308. 2013. View Article : Google Scholar : PubMed/NCBI

6 

Xu Y, Wan J, Jiang D and Wu X: BMP-7 counteracts TGF-beta1-induced epithelial-to-mesenchymal transition in human renal proximal tubular epithelial cells. J Nephrol. 22:403–410. 2009.PubMed/NCBI

7 

Bhowmick NA, Ghiassi M, Bakin A, Aakre M, Lundquist CA, Engel ME, Arteaga CL and Moses HL: Transforming growth factor-beta1 mediates epithelial to mesenchymal transdifferentiation through a RhoA-dependent mechanism. Mol Biol Cell. 12:27–36. 2001. View Article : Google Scholar : PubMed/NCBI

8 

Liu Y: Epithelial to mesenchymal transition in renal fibrogenesis: Pathologic significance, molecular mechanism, and therapeutic intervention. J Am Soc Nephrol. 15:1–12. 2004. View Article : Google Scholar : PubMed/NCBI

9 

Etienne-Manneville S and Hall A: Rho GTPases in cell biology. Nature. 420:629–635. 2002. View Article : Google Scholar : PubMed/NCBI

10 

Burridge K and Wennerberg K: Rho and Rac take center stage. Cell. 116:167–179. 2004. View Article : Google Scholar : PubMed/NCBI

11 

Kolavennu V, Zeng L, Peng H, Wang Y and Danesh FR: Targeting of RhoA/ROCK signaling ameliorates progression of diabetic nephropathy independent of glucose control. Diabetes. 57:714–723. 2008. View Article : Google Scholar : PubMed/NCBI

12 

Manickam N, Patel M, Griendling KK, Gorin Y and Barnes JL: RhoA/Rho kinase mediates TGF-β1-induced kidney myofibroblast activation through Poldip2/Nox4-derived reactive oxygen species. Am J Physiol Renal Physiol. 307:F159–F171. 2014. View Article : Google Scholar : PubMed/NCBI

13 

Zhang H, Liu X, Liu Y, Yi B and Yu X: Epithelial-mesenchymal transition of rat peritoneal mesothelial cells via Rhoa/Rock pathway. In Vitro Cell Dev Biol Anim. 47:165–172. 2011. View Article : Google Scholar : PubMed/NCBI

14 

Zhang K, Zhang H, Xiang H, Liu J, Liu Y, Zhang X, Wang J and Tang Y: TGF-β1 induces the dissolution of tight junctions in human renal proximal tubular cells: Role of the RhoA/ROCK signaling pathway. Int J Mol Med. 32:464–468. 2013.PubMed/NCBI

15 

Al-Rubeaan K, Youssef AM, Subhani SN, Ahmad NA, Al-Sharqawi AH, Al-Mutlaq HM, David SK and AlNaqeb D: Diabetic nephropathy and its risk factors in a society with a type 2 diabetes epidemic: A Saudi national diabetes registry-based study. PLoS One. 9:e889562014. View Article : Google Scholar : PubMed/NCBI

16 

Guo J, Xia N, Yang L, Zhou S, Zhang Q, Qiao Y and Liu Z: GSK-3β and vitamin D receptor are involved in β-catenin and snail signaling in high glucose-induced epithelial-mesenchymal transition of mouse podocytes. Cell Physiol Biochem. 33:1087–1096. 2014. View Article : Google Scholar : PubMed/NCBI

17 

Meems LM, Cannon MV, Mahmud H, Voors AA, van Gilst WH, Silljé HH, Ruifrok WP and de Boer RA: The vitamin D receptor activator paricalcitol prevents fibrosis and diastolic dysfunction in a murine model of pressure overload. J Steroid Biochem Mol Biol. 132:282–289. 2012. View Article : Google Scholar : PubMed/NCBI

18 

de Zeeuw D, Agarwal R, Amdahl M, Audhya P, Coyne D, Garimella T, Parving HH, Pritchett Y, Remuzzi G, Ritz E and Andress D: Selective vitamin D receptor activation with paricalcitol for reduction of albuminuria in patients with type 2 diabetes (VITAL study): A randomised controlled trial. Lancet. 376:1543–1551. 2010. View Article : Google Scholar : PubMed/NCBI

19 

Tan X, Li Y and Liu Y: Paricalcitol attenuates renal interstitial fibrosis in obstructive nephropathy. J Am Soc Nephrol. 17:3382–3393. 2006. View Article : Google Scholar : PubMed/NCBI

20 

Xiong M, Gong J, Liu Y, Xiang R and Tan X: Loss of vitamin D receptor in chronic kidney disease: A potential mechanism linking inflammation to epithelial-to-mesenchymal transition. Am J Physiol Renal Physiol. 303:F1107–F1115. 2012. View Article : Google Scholar : PubMed/NCBI

21 

Sanchez-Niño MD, Bozic M, Córdoba-Lanús E, Valcheva P, Gracia O, Ibarz M, Fernandez E, Navarro-Gonzalez JF, Ortiz A and Valdivielso JM: Beyond proteinuria: VDR activation reduces renal inflammation in experimental diabetic nephropathy. Am J Physiol Renal Physiol. 302:F647–F657. 2012. View Article : Google Scholar : PubMed/NCBI

22 

Duffy MM, McNicholas BA, Monaghan DA, Hanley SA, McMahon JM, Pindjakova J, Alagesan S, Fearnhead HO and Griffin MD: Mesenchymal stem cells and a vitamin D receptor agonist additively suppress T helper 17 cells and the related inflammatory response in the kidney. Am J Physiol Renal Physiol. 307:F1412–F1426. 2014. View Article : Google Scholar : PubMed/NCBI

23 

Ding N, Yu RT, Subramaniam N, Sherman MH, Wilson C, Rao R, Leblanc M, Coulter S, He M, Scott C, et al: A vitamin D receptor/SMAD genomic circuit gates hepatic fibrotic response. Cell. 153:601–613. 2013. View Article : Google Scholar : PubMed/NCBI

24 

Morelli A, Vignozzi L, Filippi S, Vannelli GB, Ambrosini S, Mancina R, Crescioli C, Donati S, Fibbi B, Colli E, et al: BXL-628, a vitamin D receptor agonist effective in benign prostatic hyperplasia treatment, prevents RhoA activation and inhibits RhoA/Rho kinase signaling in rat and human bladder. Prostate. 67:234–247. 2007. View Article : Google Scholar : PubMed/NCBI

25 

Manchanda PK, Kibler AJ, Zhang M, Ravi J and Bid HK: Vitamin D receptor as a therapeutic target for benign prostatic hyperplasia. Indian J Urol. 28:377–381. 2012. View Article : Google Scholar : PubMed/NCBI

26 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T)) Method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

27 

Tang SC, Leung JC and Lai KN: Diabetic tubulopathy: An emerging entity. Contrib Nephrol. 170:124–134. 2011. View Article : Google Scholar : PubMed/NCBI

28 

Kanlaya R, Sintiprungrat K and Thongboonkerd V: Secreted products of macrophages exposed to calcium oxalate crystals induce epithelial mesenchymal transition of renal tubular cells via RhoA-dependent TGF-β1 pathway. Cell Biochem Biophys. 67:1207–1215. 2013. View Article : Google Scholar : PubMed/NCBI

29 

Mezzano S, Droguett A, Burgos ME, Ardiles LG, Flores CA, Aros CA, Caorsi I, Vío CP, Ruiz-Ortega M and Egido J: Renin-angiotensin system activation and interstitial inflammation in human diabetic nephropathy. Kidney Int Suppl. S64–S70. 2003. View Article : Google Scholar : PubMed/NCBI

30 

Kim CS, Joo SY, Lee KE, Choi JS, Bae EH, Ma SK, Kim SH, Lee J and Kim SW: Paricalcitol attenuates 4-hydroxy-2-hexenal-induced inflammation and epithelial-mesenchymal transition in human renal proximal tubular epithelial cells. PLoS One. 8:e631862013. View Article : Google Scholar : PubMed/NCBI

31 

Gonçalves JG, de Bragança AC, Canale D, Shimizu MH, Sanches TR, Moysés RM, Andrade L, Seguro AC and Volpini RA: Vitamin D deficiency aggravates chronic kidney disease progression after ischemic acute kidney injury. PLoS One. 9:e1072282014. View Article : Google Scholar : PubMed/NCBI

32 

Ordonez-Mórán P, Larriba MJ, Pálmer HG, Valero RA, Barbáchano A, Duñach M, de Herreros AG, Villalobos C, Berciano MT, Lafarga M and Muñoz A: RhoA-ROCK and p38MAPK-MSK1 mediate vitamin D effects on gene expression, phenotype, and Wnt pathway in colon cancer cells. J Cell Biol. 183:697–710. 2008. View Article : Google Scholar : PubMed/NCBI

33 

Pertz O, Hodgson L, Klemke RL and Hahn KM: Spatiotemporal dynamics of RhoA activity in migrating cells. Nature. 440:1069–1072. 2006. View Article : Google Scholar : PubMed/NCBI

34 

Wildenberg GA, Dohn MR, Carnahan RH, Davis MA, Lobdell NA, Settleman J and Reynolds AB: p120-catenin and p190RhoGAP regulate cell-cell adhesion by coordinating antagonism between Rac and Rho. Cell. 127:1027–1039. 2006. View Article : Google Scholar : PubMed/NCBI

35 

Amuchastegui S, Daniel KC and Adorini L: Inhibition of acute and chronic allograft rejection in mouse models by BXL-628, a nonhypercalcemic vitamin D receptor agonist. Transplantation. 80:81–87. 2005. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May-2017
Volume 13 Issue 5

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhang W, Yi B, Zhang K, Li A, Yang S, Huang J, Liu J and Zhang H: 1,25-(OH)2D3 and its analogue BXL-628 inhibit high glucose-induced activation of RhoA/ROCK pathway in HK-2 cells. Exp Ther Med 13: 1969-1976, 2017
APA
Zhang, W., Yi, B., Zhang, K., Li, A., Yang, S., Huang, J. ... Zhang, H. (2017). 1,25-(OH)2D3 and its analogue BXL-628 inhibit high glucose-induced activation of RhoA/ROCK pathway in HK-2 cells. Experimental and Therapeutic Medicine, 13, 1969-1976. https://doi.org/10.3892/etm.2017.4211
MLA
Zhang, W., Yi, B., Zhang, K., Li, A., Yang, S., Huang, J., Liu, J., Zhang, H."1,25-(OH)2D3 and its analogue BXL-628 inhibit high glucose-induced activation of RhoA/ROCK pathway in HK-2 cells". Experimental and Therapeutic Medicine 13.5 (2017): 1969-1976.
Chicago
Zhang, W., Yi, B., Zhang, K., Li, A., Yang, S., Huang, J., Liu, J., Zhang, H."1,25-(OH)2D3 and its analogue BXL-628 inhibit high glucose-induced activation of RhoA/ROCK pathway in HK-2 cells". Experimental and Therapeutic Medicine 13, no. 5 (2017): 1969-1976. https://doi.org/10.3892/etm.2017.4211