Open Access

High glucose/lysophosphatidylcholine levels stimulate extracellular matrix deposition in diabetic nephropathy via platelet‑activating factor receptor

  • Authors:
    • Su‑Xian Zhou
    • Dong‑Mei Huo
    • Xiao‑Yun He
    • Ping Yu
    • Yan‑Hua Xiao
    • Chun‑Lin Ou
    • Ren-Mei Jiang
    • Dan Li
    • Hao Li
  • View Affiliations

  • Published online on: November 20, 2017     https://doi.org/10.3892/mmr.2017.8102
  • Pages: 2366-2372
  • Copyright: © Zhou et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Platelet-activating factor (PAF), protein kinase C (PKC)βI, transforming growth factor (TGF)‑β1 and aberrant extracellular matrix (ECM) deposition have been associated with diabetic nephropathy (DN). However, the mechanistic basis underlying this association remains to be elucidated. The present study investigated the association among the aforementioned factors in a DN model consisting of human mesangial cells (HMCs) exposed to high glucose (HG) and lysophosphatidylcholine (LPC) treatments. HMCs were divided into the following treatment groups: Control; PAF; PAF+PKCβI inhibitor LY333531; HG + LPC; PAF + HG + LPC; and PAF + HG + LPC + LY333531. Cells were cultured for 24 h, and PKCβI and TGF‑β1 expression was determined using the reverse transcription‑quantitative polymerase chain reaction and western blotting. The expression levels of the ECM‑associated molecules collagen IV and fibronectin in the supernatant were detected using ELISA analysis. Subcellular localization of PKCβI was assessed using immunocytochemistry. PKCβI and TGF‑β1 expression was increased in the PAF + HG + LPC group compared with the other groups (P<0.05); however, this effect was abolished in the presence of LY333531 (P<0.05). Supernatant fibronectin and collagen IV levels were increased in the PAF + HG + LPC group compared with the others (P<0.05); this was reversed by treatment with LY333531 (P<0.05). In cells treated with PAF, HG and LPC, PKCβI was translocated from the cytosol to the nucleus, an effect which was blocked when PKCβI expression was inhibited (P<0.05). The findings of the present study demonstrated that PAF stimulated ECM deposition in HMCs via activation of the PKC‑TGF‑β1 axis in a DN model.

Introduction

Diabetic nephropathy (DN) is the leading cause of end-stage renal disease (ESRD) in diabetes, with an incidence of 20–40% worldwide (1,2). DN is characterized by progressive renal interstitial fibrosis. A previous study reported that high glucose (HG) and lysophosphatidylcholine (LPC) levels were associated with the development and progression of DN (3); these two factors have been demonstrated to stimulate platelet-activating factor (PAF) expression and extracellular matrix (ECM) secretion by the mesangial cells (MCs) of the kidney (4).

Protein kinase C (PKC)βI is an isoenzyme in the PKC family and is involved in a number of biological processes, including cell proliferation, differentiation, apoptosis and angiogenesis (5), in addition to having a role in the pathogenesis of DN (6,7). PKC is aberrantly activated in the diabetic kidney, which leads to an increase in PKCβI activity and deposition of ECM proteins, including fibronectin (Fn) and collagen (Col) type IV (8,9). In addition, transforming growth factor (TGF)-β1 has an important role in ECM accumulation during renal fibrosis (10), and it has been implicated in the occurrence of DN (1113). However, the underlying molecular mechanism between PAF, PKC, TGF-β1 and the ECM in DN remains to be elucidated. The present study investigated the association among the aforementioned factors in a DN model consisting of human (H)MCs exposed to high HG) and LPC treatments. Reverse transcription-quantitative polymerase chain reaction and western blotting was used to detect PKCβI and TGF-β1 expression, and then an ELISA assay was used to detect the expression levels of the ECM-associated molecules collagen IV and fibronectin in the supernatant. To clarify the function of PKCβI, immunocytochemistry was used to demonstrated the subcellular localization of PKCβI. The results of the present study suggested that PAF stimulated ECM deposition in HMCs via activation of the PKC-TGF-β1 axis in a DN model.

Materials and methods

Cell culture

HMCs donated by the Zhongda Hospital affiliated with Southeast University (Nanjing, China) were maintained in Dulbecco's modified Eagle's medium containing 10% fetal calf serum (Invitrogen; Thermo Fisher Scientific, Inc., Waltham, MA, USA) in an atmosphere containing 5% CO2 at 37°C.

The cells were divided into six groups: Control (5.5 mM D-glucose; Enzo Life Sciences, Inc., Farmingdale, NY, USA); PAF (2×10−8 M PAF C-16; Cayman Chemical Company, Ann Arbor, MI, USA); PAF + PKCβI inhibitor LY333531 (Enzo Life Sciences, Inc.; 2×10−8 M PAF and 2×10−7 M LY333531); HG + LPC (Sigma-Aldrich; Merck KGaA, Darmstadt, Germany; 30 mM D-glucose and 20 mg/l LPC); PAF + HG + LPC (2×10−8 PAF, 30 mM D-glucose and 20 mg/l LPC); and PAF + HG + LPC + LY333531 (2×10−8 PAF, 30 mM D-glucose, 20 mg/l LPC and 2×10−7 M LY333531) (4).

ELISA analysis

The expression levels of Fn and Col IV in the cell culture supernatants were detected using specific ELISA kits (cat nos. CSB-EL005745HU and CSB-E04551h) according to the manufacturer's protocol (JingMei Biotech, Shenzheng, China). Samples were analyzed in triplicate.

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

Total RNA was isolated from cells using TRIzol reagent (Invitrogen; Thermo Fisher Scientific, Inc.) and was reverse transcribed into cDNA using the Revert Aid First Strand cDNA Synthesis kit (Fermentas; Thermo Fisher Scientific, Inc.), according to the manufacturer's protocol. RT-qPCR assay was performed using a SYBR_Premix ExTaq II kit (Takara Biotechnology Co., Ltd., Dalian, China) was performed using in the CFX96 Real-Time PCR Detection system (Bio-Rad Laboratories, Inc., Hercules, CA, USA) to determine the relative expression levels of target genes. The sequences of forward and reverse primers: PKCβI, 5′-GGGGGCGACCTCATGTAT-3′ and 5′-GCAATTTCTGCAGCGTAAAA-3′; and GAPDH, 5′-ACACCCACTCCTCCACCTTT-3′ and 5′-TTACTCCTTGGAGGCCATGT-3′. Primers were designed using Premier Oligo version 5 and Primer version 6.22 (Premier Biosoft International, Palo Alto, CA, USA). The thermocycling program used was as follows: 95°C for 30 sec, followed by 40 cycles of 60°C for 30 sec and 72°C for 30 sec. Relative changes in expression level were calculated using the quantification cycle (2−ΔΔCq) method (14). Each sample was prepared in triplicate and the results are expressed as the mean of three independent experiments.

Western blotting

Cells were resuspended in lysis buffer (Beijing Solarbio Science & Technology Co., Ltd., Beijing, China) for 30 min and sonicated for 2 min at 20 W, followed by centrifugation at 12,000 × g for 10 min at 4°C. The supernatant was collected and 50 µg/lane protein (concentration determined using the bicinchoninic assay kit (Thermo Fisher Scientific, Inc.) was separated using SDS-PAGE on a 10% gel (Bio-Rad Laboratories, Inc.) and transferred to a nitrocellulose membrane (Bio-Rad Laboratories, Inc.), which was blocked in Tris-buffered saline/Tween-20 (TBST) with 5% non-fat milk for 1 h at 37°C. The membrane was subsequently incubated with primary antibodies against TGF-β1 (cat no. sc-146; 1:2,000), PKCβI (cat no. sc- 209; 1:1,000) and GAPDH (cat no. sc-25778; 1:500) (both from Santa Cruz Biotechnology, Inc., Dallas, TX, USA) overnight at 4°C. Following washing with TBST, the membranes were incubated with a horseradish peroxidase-conjugated labeled goat anti-rabbit secondary antibody (cat no. sc-2004; 1:500; Santa Cruz Biotechnology, Inc.) for 1 h at 4°C, followed by additional three washes with TBST. Protein bands were visualized by enhanced chemiluminescence (GE Healthcare, Chicago, IL, USA). The Scion Image system version 4.03 (National Institutes of Health, Bethesda, MD, USA) was used to quantify band intensity and data are expressed as the mean of three independent experiments.

Immunocytochemistry

Cells (2×104/ml) were cultured on coverslips in 24-well plates for 24 h, and subsequently fixed with 4% paraformaldehyde for 5 min at −20°C and blocked at room temperature for 30 min in 0.2% Triton X-100 in PBS. The cells were incubated with anti-PKCβI antibody (1:50) (cat no. 07-870; EMD Millipore, Billerica, MA, USA) overnight at 4°C, followed by fluorescein isothiocyanate-conjugated secondary antibody (1:400; cat no. K532511-8; Dako; Agilent Technologies, Inc., Santa Clara, CA, USA) for 1 h in the dark at room temperature. Following three washes in PBS, coverslips were placed on the slides and the cells were visualized using confocal microscopy. Fluorescence intensity (wavelength of 490 nm) was analyzed using Image J software (version number: 1.48u; (National Institutes of Health).

Statistical analysis

Data are expressed as the mean ± standard error of the mean. Data were analyzed using SPSS software version 13.0 (SPSS, Inc., Chicago, IL, USA). Differences between groups were assessed using the Student's t-test. P<0.05 was considered to indicate a statistically significant difference.

Results

PKCβI expression is upregulated in HMCs in the presence of PAF, HG and LPC

PKCβI mRNA expression level was increased in the PAF, HG + LPC, and PAF + HG + LPC groups compared with control group (P<0.05). The expression was increased in the PAF + HG + LPC group compared with cells treated with HG and LPC alone (P<0.05), this increase in PKCβI expression was reversed by treatment with the PKCβI inhibitor LY333531 (P<0.05; Table I; Fig. 1).

Table I.

PKCβI mRNA expression in each treatment group.

Table I.

PKCβI mRNA expression in each treatment group.

GroupExpression
Control 1.00±0.00
PAF 2.68±0.17a
PAF + LY333531 1.85±0.39a,b
HG + LPC 2.12±0.31a
PAF + HG + LPC 3.59±0.41a
PAF + HG + LPC + LY333531 2.76±0.57a,c

a P<0.05 vs. control group

b P<0.05 vs. PAF group

c P<0.05 vs. PAF + HG + LPC group. PKCβI, protein kinase C βI; PAF, platelet activating factor; LY333531, PKCβI inhibitor; HG, high glucose; LPC, lysophosphatidylcholine.

A similar trend was observed for PKCβI protein expression, which was increased in the PAF, HG + LPC and PAF + HG + LPC groups compared with control cells (P<0.05; Fig. 2). The observed upregulation in PKCβI expression levels was reduced following treatment with LY333531 (P<0.05).

TGF-β1 expression is upregulated in HMCs in the presence of PAF, HG and LPC

TGF-β1 mRNA (Table II; Fig. 3) and protein (Fig. 4) expression levels were upregulated in HMCs treated with PAF, HG and LPC, compared with the control (P<0.05). The increased expression was not observed in the presence of LY333531.

Table II.

TGF-β1 mRNA expression in each treatment group.

Table II.

TGF-β1 mRNA expression in each treatment group.

GroupExpression
Control 1.00±0.00
PAF 1.84±0.11a
PAF + LY333531 1.02±0.15b
HG + LPC 1.88±0.21a
PAF + HG + LPC 2.25±0.09a
PAF + HG + LPC + LY333531 1.95±0.11a,c

a P<0.05 vs. control group

b P<0.05 vs. PAF group

c P<0.05 vs. PAF + HG + LPC group. TGF-β1, transforming growth factor-β1; PAF, platelet activating factor; LY333531, PKCβI inhibitor; HG, high glucose; LPC, lysophosphatidylcholine.

ECM production is induced in HMCs in the presence of PAF, HG and LPC

The expression levels of two ECM proteins, Fn and Col IV, in the supernatant of cultured HMCs were significantly upregulated following treatment with PAF, HG and LPC, compared with the control group (P<0.05; Fig. 5), with increased levels observed in cells treated with all three factors compared with HG and LPC group (P<0.05). This effect was reduced following treatment with LY333531 (Table III).

Table III.

Expression of the extracellular matrix components Fn and Col IV in the different treatment groups.

Table III.

Expression of the extracellular matrix components Fn and Col IV in the different treatment groups.

GroupFn, mg/lCol IV, µg/l
Control 3.90±0.43 4.54±0.74
PAF 7.05±0.05a 13.71±0.88a
PAF + LY333531 3.81±0.13b 5.31±0.81b
HG + LPC 7.89±0.34a,c 16.32±1.55a,c
PAF + HG + LPC 9.11±0.10a 22.89±0.34a
PAF + HG + LPC + LY333531 5.23±0.24a,c 11.40±0.72a,c

a P<0.05 vs. control group

b P<0.05 vs. PAF group

c P<0.05 vs. PAF + HG + LPC group. Fn, fibronectin; Col IV, collagen type IV; PAF, platelet activating factor; LY333531, PKCβI inhibitor; HG, high glucose; LPC, lysophosphatidylcholine.

PKCβI protein is translocated from the cytoplasm to the nucleus of HMCs following treatment with PAF, HG and LPC

In the control group, PKCβI was diffusely distributed throughout the cytoplasm, with no membrane or nuclear localization. Treatment with PAF, HG and LPC increased PKCβI protein levels, and induced the translocation of the protein from the cytoplasm to the nucleus(P<0.05). Treatment with LY333531 did not alter in the subcellular localization of PKCβI protein (Table IV; Figs. 6 and 7).

Table IV.

Mean fluorescence intensity of PKCβI in human mesangial cells under various treatment conditions.

Table IV.

Mean fluorescence intensity of PKCβI in human mesangial cells under various treatment conditions.

GroupMean fluorescence intensity
Control 11.80±2.57
PAF 41.14±7.21a
PAF + LY333531 20.19±3.60b
HG + LPC 48.92±7.70a
PAF + HG + LPC 54.45±3.57a
PAF + HG + LPC + LY333531 42.50±5.70a,c

a P<0.05 vs. control group

b P<0.05 vs. PAF group

c P<0.05 vs. PAF + HG + LPC group. PKCβI, protein kinase CβI; PAF, platelet activating factor; LY333531, PKCβI inhibitor; HG, high glucose; LPC, lysophosphatidylcholine.

Discussion

Diabetes mellitus is an important public health concern, especially in developed countries (15), with DN being the primary cause of ESRD worldwide (1619). DN is caused by nerve damage resulting from ECM deposition, mesangial expansion and basement membrane thickening (20). The accumulation of Fn and Col IV underlies chronic kidney diseases, including progressive renal interstitial fibrosis (21). Metabolic disorders, such as hyperlipidemia and hyperglycemia, are associated with the occurrence and development of DN, with increased glucose and fat levels having an adverse effect on glomerular capillary endothelial cells and MCs, in addition to podocytes in the kidney (22), via stimulation of ECM secretion (23) mediated by TGF-β/mothers against decapentaplegic homolog 3 signaling. A HG/high fat diet may upregulate Fn and Col IV expression, which may alter the structure and function of renal tubules and lead to renal tubulointerstitial fibrosis (24). PAF is a lipid polymer, involved in the metabolism of arachidonic acid, that has a role in DN by stimulating Fn secretion (25). The present study determined that Fn and Col IV secretion were stimulated by PAF, HG and LPC, consistent with previous studies (8,26,27). The findings of the present study supported the hypothesis that HG and LPC may be risk factors for renal fibrosis and DN.

PKC is a serine/threonine kinase expressed in various mammalian tissues, which regulates a number of signaling pathways (28,29). The present study revealed that PKC was diffusely distributed throughout the cytoplasm in untreated HMCs and translocated to the nucleus in the presence of PAF, HG and LPC. DN may be delayed or prevented by inhibiting PKC (30,31); enlargement of kidney volume and renal fibrosis were rescued by PKCβI-knockout in a mouse model of DN (8). LY333531 is a Food and Drug Administration-approved inhibitor of PKC-Β (32), which has been demonstrated to promote myocardial angiogenesis in diabetes (33) and improve albuminuria and other pathological features in DN rats via inhibition of PKC expression (34). Treatment with LY333531 was demonstrated to reduce mesangial matrix expansion and decrease the urinary protein excretion rate in diabetic mice (35). In the present study, LY333531 treatment prevented the nuclear localization of PKCβI protein in the presence of PAF, HG and LPC, which corresponded to the decrease in Fn and Col IV secretion. The findings of the present study suggested that PKCβI may have an important role in ECM deposition by HMCs in DN.

TGF-β1 is a TGF-β superfamily member which regulates a variety of cellular processes, including proliferation, differentiation and apoptosis (36,37). TGF-β1 has an important role in kidney hypertrophy (26), glomerular and renal tubular basement membrane thickening, and renal tubulointerstitial fibrosis (38,39), and previous studies have suggested that it may modulate ECM secretion in DN. For example, plasmacytoma variant translocation 1 was demonstrated to increase plasminogen TGF-β1 in addition to Fn expression in MCs (40), whereas TGF-β1 inhibited the expression of microRNA (miR)-26a to modulate DN progression in diabetic mice (41). ECM accumulation was increased via upregulation of miR-1207-5p in the presence of glucose and TGF-β1, which was implicated in DN pathogenesis (42). Additionally, Fn and Col IV levels were suppressed by the knockdown of TGF-β1 (43). The present study revealed that TGF-β1 mRNA and protein expression levels were upregulated in HMCs, following treatment with PAF, HG and LPC compared with the control group, which was accompanied by increased Fn and Col IV secretion; these effects were abolished by treatment with LY333531.

In conclusion, the findings of the present study suggested that ECM deposition by MCs may be induced by HG and LPC treatment and activation of PKCβI–TGF-β1 signaling via PAF. Increased ECM deposition increases the risk of glomerular fibrosis and DN in individuals with disorders of glucose and lipid metabolism. The present findings reveal novel strategies for managing DN by targeting the PKC-TGF-β1 signaling pathway in MCs.

Acknowledgements

The authors of the present study would like to thank the Research Center of Guilin Medical University, and the laboratory staff for their assistance. The present study was supported by the National Natural Science Foundation of China (grant nos. 81260134 and 81560148).

References

1 

Schernthaner G, Mogensen CE and Schernthaner GH: The effects of GLP-1 analogues, DPP-4 inhibitors and SGLT2 inhibitors on the renal system. Diab Vasc Dis Res. 11:306–323. 2014. View Article : Google Scholar : PubMed/NCBI

2 

Zhang MH, Feng L, Zhu MM, Gu JF, Jiang J, Cheng XD, Ding SM, Wu C and Jia XB: The anti-inflammation effect of Moutan Cortex on advanced glycation end products-induced rat mesangial cells dysfunction and High-glucose-fat diet and streptozotocin-induced diabetic nephropathy rats. J Ethnopharmacol. 151:591–600. 2014. View Article : Google Scholar : PubMed/NCBI

3 

Xie S, Lu K, Zhang Y, Song X, Tan M and Wang C: Effects of Jiangya Xiaoke prescription on TGF-beta1 in diabetic nephropathy rats with hypertension and its mechanisms. Int J Clin Exp Med. 8:5129–5136. 2015.PubMed/NCBI

4 

Zhou SX, Lei MX and Zhao JJ: The study of the effects of platelet activating factor (PAF) on the relation between the endothelial cell and mesangial cells exposed to high glucose and high lysophosphatidylcholine. Chin J Diabetes. 18:591–593. 2010.(In Chinese).

5 

Al-Khodor S and Abu KY: Triggering Ras signalling by intracellular Francisella tularensis through recruitment of PKCalpha and betaI to the SOS2/GrB2 complex is essential for bacterial proliferation in the cytosol. Cell Microbiol. 12:1604–1621. 2010. View Article : Google Scholar : PubMed/NCBI

6 

Noh H and King GL: The role of protein kinase C activation in diabetic nephropathy. Kidney Int Suppl. S49–S53. 2007. View Article : Google Scholar : PubMed/NCBI

7 

Bryant DM, Roignot J, Datta A, Overeem AW, Kim M, Yu W, Peng X, Eastburn DJ, Ewal AJ, Werb Z and Mostov KE: A molecular switch for the orientation of epithelial cell polarization. Dev Cell. 31:171–187. 2014. View Article : Google Scholar : PubMed/NCBI

8 

Meier M, Park JK, Overheu D, Kirsch T, Lindschau C, Gueler F, Leitges M, Menne J and Haller H: Deletion of protein kinase C-beta isoform in vivo reduces renal hypertrophy but not albuminuria in the streptozotocin-induced diabetic mouse model. Diabetes. 56:346–354. 2007. View Article : Google Scholar : PubMed/NCBI

9 

Idris I and Donnelly R: Protein kinase C beta inhibition: A novel therapeutic strategy for diabetic microangiopathy. Diab Vasc Dis Res. 3:172–178. 2006. View Article : Google Scholar : PubMed/NCBI

10 

Muñoz-Felix JM, Oujo B and Lopez-Novoa JM: The role of endoglin in kidney fibrosis. Expert Rev Mol Med. 16:e182014. View Article : Google Scholar : PubMed/NCBI

11 

Wang T, Chen SS, Chen R, Yu DM and Yu P: Reduced beta 2 glycoprotein I improves diabetic nephropathy via inhibiting TGF-β1-p38 MAPK pathway. Int J Clin Exp Pathol. 8:2321–2333. 2015.PubMed/NCBI

12 

Hathaway CK, Gasim AM, Grant R, Chang AS, Kim HS, Madden VJ, Bagnell CR Jr, Jennette JC, Smithies O and Kakoki M: Low TGFβ1 expression prevents and high expression exacerbates diabetic nephropathy in mice. Proc Natl Acad Sci USA. 112:pp. 5815–5820. 2015; View Article : Google Scholar : PubMed/NCBI

13 

Gao P, Li L, Ji L, Wei Y, Li H, Shang G, Zhao Z, Chen Q, Jiang T and Zhang N: Nrf2 ameliorates diabetic nephropathy progression by transcriptional repression of TGFβ1 through interactions with c-Jun and SP1. Biochim Biophys Acta. 1839:1110–1120. 2014. View Article : Google Scholar : PubMed/NCBI

14 

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

15 

Duan JG, Chen XY, Wang L, Lau A, Wong A, Thomas GN, Tomlinson B, Liu R, Chan JC, Leung TW, et al: Sex differences in epidemiology and risk factors of acute coronary syndrome in Chinese patients with type 2 diabetes: A long-term prospective cohort study. PLoS One. 10:e1220312015. View Article : Google Scholar

16 

Bakris GL, Pitt B, Weir MR, Freeman MW, Mayo MR, Garza D, Stasiv Y, Zawadzki R, Berman L and Bushinsky DA: AMETHYST-DN Investigators: Effect of patiromer on serum potassium level in patients with hyperkalemia and diabetic kidney disease: The AMETHYST-DN randomized clinical trial. JAMA. 314:151–161. 2015. View Article : Google Scholar : PubMed/NCBI

17 

Panduru NM, Saraheimo M, Forsblom C, Thorn LM, Gordin D, Wadén J, Tolonen N, Bierhaus A, Humpert PM and Groop PH; FinnDiane Study Group, : Urinary adiponectin is an independent predictor of progression to end-stage renal disease in patients with type 1 diabetes and diabetic nephropathy. Diabetes Care. 38:883–890. 2015. View Article : Google Scholar : PubMed/NCBI

18 

De Nicola L, Provenzano M, Chiodini P, Borrelli S, Garofalo C, Pacilio M, Liberti ME, Sagliocca A, Conte G and Minutolo R: Independent role of underlying kidney disease on renal prognosis of patients with chronic kidney disease under nephrology care. PLoS One. 10:e1270712015. View Article : Google Scholar

19 

Liu X, Yang G, Fan Q and Wang L: Proteomic profile in glomeruli of type-2 diabetic KKAy mice using 2-dimensional differential gel electrophoresis. Med Sci Monit. 20:2705–2713. 2014. View Article : Google Scholar : PubMed/NCBI

20 

Abe H: Recent progress in understanding the molecular pathogenesis of diabetic nephropathy. Rinsho Byori. 59:179–186. 2011.(In Japanese). PubMed/NCBI

21 

Rossert J, Terraz-Durasnel C and Brideau G: Growth factors, cytokines, and renal fibrosis duringthe course of diabetic nephropathy. Diabetes Metab. 26 Suppl 4:S16–S24. 2000.

22 

Zhou L: Research progress in impact of high glucose and hyperlipidemia on glomerular cells. New Med. 286–289. 2014.(In Chinese).

23 

Li L, Yin Q, Tang X, Bai L, Zhang J, Gou S, Zhu H, Cheng J, Fu P and Liu F: C3a receptor antagonist ameliorates inflammatory and fibrotic signals in type 2 diabetic nephropathy by suppressing the activation of TGF-β/smad3 and IKBα pathway. PLoS One. 9:e1136392014. View Article : Google Scholar : PubMed/NCBI

24 

Li HG, Cai YJ and Zou JH: The effects of high-gucrose and high-fat diet on tubulointerstitial fibrosis in New Zanland white rabbits. Chinese J Zoology. 145–150. 2010.(In Chinese).

25 

Yoshikawa M, Matsumoto K, Iida M, Akasawa A, Moriyama H and Saito H: Effect of extracellular matrix proteins on platelet-activating factor-induced eosinophil chemotaxis. Int Arch Allergy Immunol. 128 Suppl 1:S3–S11. 2002. View Article : Google Scholar

26 

Yao LJ, Wang JQ, Zhao H, Liu JS and Deng AG: Effect of telmisartan on expression of protein kinase C-alpha in kidneys of diabetic mice. Acta Pharmacol Sin. 28:829–838. 2007. View Article : Google Scholar : PubMed/NCBI

27 

Wogensen L, Krag S, Chai Q and Ledet T: The use of transgenic animals in the study of diabetic kidney disease. Horm Metab Res. 37 Suppl 1:S17–S25. 2005. View Article : Google Scholar

28 

Mishra S and Vinayak M: Ellagic acid checks lymphoma promotion via regulation of PKC signaling pathway. Mol Biol Rep. 40:1417–1428. 2013. View Article : Google Scholar : PubMed/NCBI

29 

do Carmo A, Balça-Silva J, Matias D and Lopes MC: PKC signaling in glioblastoma. Cancer Biol Ther. 14:287–294. 2013. View Article : Google Scholar : PubMed/NCBI

30 

Manabe E, Handa O, Naito Y, Mizushima K, Akagiri S, Adachi S, Takagi T, Kokura S, Maoka T and Yoshikawa T: Astaxanthin protects mesangial cells from hyperglycemia-induced oxidative signaling. J Cell Biochem. 103:1925–1937. 2008. View Article : Google Scholar : PubMed/NCBI

31 

Ochi S, Harigai M, Mizoguchi F, Iwai H, Hagiyama H, Oka T and Miyasaka N: Leflunomide-related acute interstitial pneumonia in two patients with rheumatoid arthritis: Autopsy findings with a mosaic pattern of acute and organizing diffuse alveolar damage. Mod Rheumatol. 16:316–320. 2006. View Article : Google Scholar : PubMed/NCBI

32 

Schwartz SG, Flynn HW Jr and Aiello LP: Ruboxistaurin mesilate hydrate for diabetic retinopathy. Drugs Today (Barc). 45:269–274. 2009. View Article : Google Scholar : PubMed/NCBI

33 

Wang F, Huang D, Zhu W, Li S, Yan M, Wei M and Li J: Selective inhibition of PKCbeta2 preserves cardiac function after myocardial infarction and is associated with improved angiogenesis of ischemic myocardium in diabetic rats. Int J Mol Med. 32:1037–1046. 2013. View Article : Google Scholar : PubMed/NCBI

34 

Kelly DJ, Zhang Y, Hepper C, Gow RM, Jaworski K, Kemp BE, Wilkinson-Berka JL and Gilbert RE: Protein kinase C beta inhibition attenuates the progression of experimental diabetic nephropathy in the presence of continued hypertension. Diabetes. 52:512–518. 2003. View Article : Google Scholar : PubMed/NCBI

35 

Koya D, Haneda M, Nakagawa H, Isshiki K, Sato H, Maeda S, Sugimoto T, Yasuda H, Kashiwagi A, Ways DK, et al: Amelioration of accelerated diabetic mesangial expansion by treatment with a PKC beta inhibitor in diabetic db/db mice, a rodent model for type 2 diabetes. FASEB J. 14:439–447. 2000.PubMed/NCBI

36 

Hinz B: The extracellular matrix and transforming growth factor-β1: Tale of a strained relationship. Matrix Biol. 47:54–65. 2015. View Article : Google Scholar : PubMed/NCBI

37 

Kajdaniuk D, Marek B, Borgiel-Marek H and Kos-Kudła B: Transforming growth factor β1 (TGFβ1) in physiology and pathology. Endokrynol Pol. 64:384–396. 2013. View Article : Google Scholar : PubMed/NCBI

38 

Huang W, Xu C, Kahng KW, Noble NA, Border WA and Huang Y: Aldosterone and TGF-beta1 synergistically increase PAI-1 and decrease matrix degradation in rat renal mesangial and fibroblast cells. Am J Physiol Renal Physiol. 294:F1287–F1295. 2008. View Article : Google Scholar : PubMed/NCBI

39 

Sam R, Wanna L, Gudehithlu KP, Garber SL, Dunea G, Arruda JA and Singh AK: Glomerular epithelial cells transform to myofibroblasts: Early but not late removal of TGF-beta1 reverses transformation. Transl Res. 148:142–148. 2006. View Article : Google Scholar : PubMed/NCBI

40 

Alvarez ML and DiStefano JK: Functional characterization of the plasmacytoma variant translocation 1 gene (PVT1) in diabetic nephropathy. PLoS One. 6:e186712011. View Article : Google Scholar : PubMed/NCBI

41 

Koga K, Yokoi H, Mori K, Kasahara M, Kuwabara T, Imamaki H, Ishii A, Mori KP, Kato Y, Ohno S, et al: MicroRNA-26a inhibits TGF-β-induced extracellular matrix protein expression in podocytes by targeting CTGF and is downregulated in diabetic nephropathy. Diabetologia. 58:2169–2180. 2015. View Article : Google Scholar : PubMed/NCBI

42 

Alvarez ML, Khosroheidari M, Eddy E and Kiefer J: Role of microRNA 1207-5P and its host gene, the long non-coding RNA Pvt1, as mediators of extracellular matrix accumulation in the kidney: Implications for diabetic nephropathy. PLoS One. 8:e774682013. View Article : Google Scholar : PubMed/NCBI

43 

Hwang M, Kim HJ, Noh HJ, Chang YC, Chae YM, Kim KH, Jeon JP, Lee TS, Oh HK, Lee YS and Park KK: TGF-beta1 siRNA suppresses the tubulointerstitial fibrosis in the kidney of ureteral obstruction. Exp Mol Pathol. 81:48–54. 2006. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

February-2018
Volume 17 Issue 2

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhou SX, Huo DM, He XY, Yu P, Xiao YH, Ou CL, Jiang R, Li D and Li H: High glucose/lysophosphatidylcholine levels stimulate extracellular matrix deposition in diabetic nephropathy via platelet‑activating factor receptor. Mol Med Rep 17: 2366-2372, 2018
APA
Zhou, S., Huo, D., He, X., Yu, P., Xiao, Y., Ou, C. ... Li, H. (2018). High glucose/lysophosphatidylcholine levels stimulate extracellular matrix deposition in diabetic nephropathy via platelet‑activating factor receptor. Molecular Medicine Reports, 17, 2366-2372. https://doi.org/10.3892/mmr.2017.8102
MLA
Zhou, S., Huo, D., He, X., Yu, P., Xiao, Y., Ou, C., Jiang, R., Li, D., Li, H."High glucose/lysophosphatidylcholine levels stimulate extracellular matrix deposition in diabetic nephropathy via platelet‑activating factor receptor". Molecular Medicine Reports 17.2 (2018): 2366-2372.
Chicago
Zhou, S., Huo, D., He, X., Yu, P., Xiao, Y., Ou, C., Jiang, R., Li, D., Li, H."High glucose/lysophosphatidylcholine levels stimulate extracellular matrix deposition in diabetic nephropathy via platelet‑activating factor receptor". Molecular Medicine Reports 17, no. 2 (2018): 2366-2372. https://doi.org/10.3892/mmr.2017.8102