The Notch pathway mediates the angiotensin II-induced synthesis of extracellular matrix components in podocytes

  • Authors:
    • Min Yao
    • Xiaomei Wang
    • Xiaomeng Wang
    • Tao Zhang
    • Yanqing Chi
    • Feng Gao
  • View Affiliations

  • Published online on: April 22, 2015     https://doi.org/10.3892/ijmm.2015.2193
  • Pages: 294-300
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The Notch pathway is known to contribute to the development of glomerular disease. Angiotensin II (Ang II), an important member of the renin-angiotensin system, stimulates the accumulation of extracellular matrix components in glomerular disease; however, the exact mechanisms involved remain to be elucidated. In the present study, we aimed to investigate the effects of the Notch pathway on the synthesis of extracellular matrix components in Ang II-stimulated podocytes. Mouse podocytes were stimulated with Ang II (10-6 mol/l). The activation of the Notch pathway was inhibited by a vector carrying short hairpin RNA (shRNA) targeting Notch1 (sh-Notch1) or by γ-secretase inhibitor (GSI). The protein levels of Notch1, Notch intracellular domain 1 (NICD1), hairy and enhancer of split-1 (Hes1), matrix metalloproteinase (MMP)-2, MMP-9, transforming growth factor-β1 (TGF-β1), type IV collagen and laminin were determined by western blot analysis. The Notch1, Hes1, MMP-2, MMP-9, TGF-β1, type IV collagen and laminin mRNA levels were detected by RT-PCR. The MMP-2 and MMP-9 activity was measured using a cell active fluorescence assay kit. The levels of TGF-β1, type IV collagen and laminin were determined in the culture medium of the podocytes by enzyme-linked immunosorbent assay (ELISA). Our results revealed that Ang II upregulated Notch1, NICD1, Hes1, TGF-β1, type IV collagen and laminin expression and downregulated MMP-2 and MMP-9 expression in the cultured podocytes. The inhibition of the Notch pathway by sh-Notch1 or GSI increased MMP-2 and MMP-9 expression, decreased the TGF-β1 level and suppressed type IV collagen and laminin expression. The inhibition of the Notch pathway by sh-Notch1 or GSI also increased MMP-2 and MMP-9 activity, and decreased TGF-β1 levels, type IV collagen levels and laminin secretion. These findings indicate that the Notch pathway potentially mediates the Ang II-induced synthesis of extracellular matrix components in podocytes through the regulation of MMPs and TGF-β1.

Introduction

It is considered that the dysfunction of podocytes, which are a type of glomerular epithelial cell, plays an important role in progressive renal diseases, including focal segmental glomerulosclerosis (FSGS) and diabetic nephropathy (DN) (1,2). Podocyte injury is also associated with a higher degree of albuminuria and leads to the development of glomerulosclerosis. It has recently been demonstrated that the Notch pathway plays an important role in the onset and development of glomerular disease (3). Niranjan et al (4) reported that the Notch pathway was activated in podocytes in patients with FSGS and DN and in mouse models of these diseases. The Notch pathway is an evolutionarily conserved and widely used intercellular signaling pathway that influences cellular proliferation and differentiation (5,6). The binding of a ligand induces a conformational change in the Notch receptor. This allows an extracellular metalloprotease to cleave the receptor, which then allows the γ-secretase-mediated protease to release the Notch intracellular domain (NICD). NICD travels into the nucleus where it activates the transcription of downstream genes, such as the hairy and enhancer of split (Hes) gene.

It is known that alterations in hemodynamics contribute to the development of glomerular disease. Alterations in hemodynamics in glomerular disease activate angiotensin II (Ang II), an important member of the renin-angiotensin system, which damages podocytes and results in the accumulation of extracellular matrix (ECM) components and basement membrane thickening in the glomeruli (7,8). It is well recognized that in glomerular disease, Ang II increases the synthesis of ECM components, including collagens and laminin (9,10). Koshizaka et al (11) found that Ang II induced podocyte apoptosis through the activation of the Notch pathway in cultured podocytes. However, it is unclear as to whether the Notch pathway mediates the Ang II-induced ECM synthesis in podocytes.

Matrix metalloproteinases (MMPs) constitute a family of extracellular soluble or membrane-bound proteases that collectively degrade or proteolytically modify essentially all the main components of the ECM, including collagens, laminin and proteoglycans. Gelatinases (e.g., MMP-2 and MMP-9), which degrade type IV collagen and laminin, are important constitutional elements of the basement membrane of the glomeruli (12,13). It is well established that transforming growth factor-β1 (TGF-β1) is a potent stimulator of ECM production in glomerular injury and may be the most important growth factor in determining the extent of renal fibrosis following injury. The exposure of cultured podocytes and mesangial cells to TGF-β1 has been shown to increase the production of type IV collagen and laminin (1416). Previous studies have demonstrated that Ang II decreases matrix degradation due to a reduction in MMP activity and promotes the synthesis of ECM components through TGF-β1 (17,18).

In the present study, we aimed to investigate the hypothesis that Ang II may induce the activation of the Notch pathway in podocytes. The activation of the Notch pathway may alter the production of ECM through MMPs and TGF-β1 in Ang II-stimulated podocytes. The inhibition of the activation of the Notch pathway can prevent ECM accumulation and glomerulosclerosis.

Materials and methods

Cell culture

Conditionally immortalized mouse podocytes were purchased from the Cell Resource Center (Peking Union Medical College, Beijing, China). In this cell line, a temperature-sensitive mutant of the SV40 virus large T-cell antigen (tsA58 Tag) is controlled by a γ-interferon-inducible H-2Kb promoter. The podocytes were firstly cultured in RPMI-1640 medium supplemented with 10% fetal bovine serum (FBS) (both from Gibco-BRL, Gaithersburg, MD, USA) and 10 U/ml γ-interferon (PeproTech, Rochy Hill, NJ, USA) in a 33°C 5% CO2 atmosphere to induce proliferation, and were then incubated in RPMI-1640 medium supplemented with 10% FBS and deprived of γ-interferon in a 37°C 5% CO2 atmosphere for 14 days to induce quiescence and the differentiated phenotype, as previously described (19). The podocytes were grown to 75–85% confluence under growth restrictive conditions and growth-arrested in serum-free RPMI-1640 for 24 h to synchronize cell growth. After this time period, the medium was changed to fresh serum-free medium containing Ang II (10−6 mol/l; Sigma, St. Louis, MO, USA) at the indicated time points, as previously described (11). The transient transfection of the podocytes with a vector carrying short hairpin RNA (shRNA) targeting Notch1 (sh-Notch1) or a negative control vector (sh-Scramble) (Jingsai Corp., Wuhan, China) was carried out using Lipofectamine 2000 (Invitrogen, Carlsbad, CA, USA) according to the manufacturer’s instructions. For inhibition experiments, the cells were treated with γ-secretase inhibitor (GSI; Sigma) at 1 μmol/l for 30 min prior to stimulation with Ang II.

Western blot analysis

The cells were lysed in lysis buffer (20 mmol/l Tris·HCl, 2.5 mmol/l EDTA, 10% glycerol, 0.1% SDS, 1% Triton X-100, 1% sodium deoxycholate, 10 mmol/l sodium pyrophosphate, 50 mmol/l NaF, 1 mmol/l sodium vanadate and 1 mmol/l PMSF) and the protein concentrations were measured by Coomassie brilliant blue assay. Protein (40 μg) was loaded and separated on a SDS-polyacrylamide gel and then transferred onto a polyvinylidene fluoride (PVDF) membrane (Millipore Corp., Billerica, MA, USA). The membrane was blocked with 5% dry milk and incubated overnight at 4°C with rabbit anti-Notch1 (ab65297; 1:200 dilution), anti-NICD1 (ab52301; 1:200 dilution), anti-Hes1 (ab71599; 1:2,000 dilution) (all from Abcam, Cambridge, MA, USA), anti-MMP-2 (10737-2-AP; 1:500 dilution; Proteintech, Chicago, IL, USA), anti-MMP-9 (sc-10737; 1:400 dilution; Santa Cruz Biotechnology, Inc., Santa Cruz, CA, USA), anti-TGF-β1 (18978-1-AP; 1:1,000 dilution), anti-type IV collagen (19797-1-AP; 1:1,000 dilution), anti-laminin (19698-1-AP; 1:1,000 dilution) (all from Proteintech) and anti-β-actin (sc-130656; 1:1,000 dilution; Santa Cruz Biotechnology, Inc.) polyclonal antibodies. After washing, the membrane was incubated with goat anti-rabbit IgG horseradish peroxidase conjugate (SA00001-2; 1:5,000 dilution; Proteintech). Proteins in western blot analysis were quantified following acquisition and analysis of the image using the software of a UVP Image Station Lab Works version 4.5. Proteins expression was quantified by comparison with the internal control, β-actin.

Reverse transcription PCR (RT-PCR)

Total RNA was extracted from the podocytes using TRIzol reagent (Invitrogen) according to the instructions of the manufacturer and reverse transcribed using oligo(dT) primers in the presence of avian myeloblastosis virus reverse transcriptase to yield cDNA. The cDNA was amplified on a 7900HT Sequence Detection system (Applied Biosystems, Foster City, CA, USA) at default thermal cycling conditions: 2 min at 50°C, 10 min at 95°C for enzyme activation and then 40 cycles of 15 sec at 95°C for denaturation and 1 min at 60°C for annealing and extension. The results were analyzed using the relative standard curve method of analysis/ΔCt method of analysis. The primers used for PCR were as follows: 18S forward, 5′-CGC CGC TAG AGG TGA AAT TC-3′ and reverse, 5′-CCA GTC GGC ATC GTT TAT GG-3′ (149 bp); Notch1 forward, 5′-GTG GAT GAC CTA GGC AAG TCG-3′ and reverse, 5′-GTC TCC TCC TTG TTG TTC TGC A-3′ (118 bp); Hes1 forward, 5′-CAC GAC ACC GGA CAA ACC A-3′ and reverse, 5′-GCC GGG AGC TAT CTT TCT TAA GTG-3′ (148 bp); MMP-2 forward, 5′-CAG GGA ATG AGT ACT GGG TCT ATT-3′ and reverse, 5′-ACT CCA GTT AAA GGC AGC ATC TAC-3′ (118 bp); MMP-9 forward, 5′-CAA TCC TTG CAA TGT GGA TG-3′ and reverse, 5′-TAA GGA AGG GGC CCT GTA AT-3′ (128 bp); TGF-β1 forward, 5′-ACC GCA ACA ACG CAA TCT ATG-3′ and reverse, 5′-ATT CCG TCT CCT TGG TTC AG-3′ (196 bp); type IV collagen forward, 5′-GTC AAA CTA CTG CTA TCC CTC CGT GTC-3′ and reverse, 5′-CAT TCT ATA AAT GGA CTG GCT CGG AAT-3′ (162 bp); laminin forward, 5′-CCT GCC AAA TTC CTC GGT AAC-3′ and reverse, 5′-ACA TCG TAG GCA GAC GGC TG-3′ (101 bp).

Enzyme activity assay

The activity of MMP-2 and MMP-9 in the serum-free conditioned medium was assayed using a cell active MMP-2 and MMP-9 fluorescence assay kit (GenMed Scientifics, Arlington, MA, USA), according to the manufacturer’s instructions. The relative fluorescence units were determined with an excitation wavelength of 330 nm and an emission wavelength of 400 nm. The consistency of the fluorescent polypeptide segments was calculated on the basis of the relative fluorescence units. MMP-2 and MMP-9 activity was expressed as nmol/mg/min.

Enzyme-linked immunosorbent assay (ELISA)

After the cells were cultured in 6-well plates under the different experimental conditions, the supernatants were collected. The TGF-β1, type IV collagen and laminin protein levels were quantified using a commercial quantikine enzyme-linked immunosorbent assay kit (ELISA; R&D Systems, Minneapolis, MN, USA) according to the manufacturer’s instructions.

Statistical analysis

Data presented as bar graphs are the means ± standard deviation (SD) of at least 3 independent experiments. Statistical analysis was performed using the Student’s t-test with SPSS version 13.0 software (SPSS Inc., Chicago, IL, USA). The results were considered statistically significant at P<0.05.

Results

Ang II activates the Notch pathway in podocytes

To determine the effects of Ang II on the activation of the Notch pathway, we measured the Notch1, NICD1 and Hes1 levels in the podocytes stimulated with Ang II for 0, 12, 24, 48 and 72 h (Fig. 1). Notch1 protein and mRNA expression increased within 12 h of Ang II stimulation, peaked at 24 h, and then slightly decreased at 48 and 72 h (all P<0.01); the maximum protein expression levels of NICD1 were detected as early as 12 h and then the expression levels declined, but did not return to the basal levels (Ang II stimulation at 0 h) (all P<0.01; Fig. 1A and B); Hes1 protein and mRNA expression also peaked at 12 h and then decreased at 24 and 48 h (P<0.01; Fig. 1A and C). No changes in Hes1 protein and mRNA expression were detected in the cultured podocytes at 0 and 72 h (P>0.05; Fig. 1).

Transfection with sh-Notch1 or pre-treatment with GSI increases MMP and decreases TGF-β1 expression in the Ang II-stimulated podocytes

Compared with the cells stimulated with Ang II at 0 h, the protein levels of Notch1, NICD1 and Hes1 significantly increased at 12 h (Fig. 2A and B; all P<0.01). Transfection with sh-Notch1 decreased the Ang II-induced the protein overexpression of Notch1, NICD1 and Hes1 in the podocytes (all P<0.01; Fig. 2A and B). RT-PCR revealed similar changes in Notch1 and Hes1 mRNA expression following transfection (Fig. 2C). GSI decreased the NICD1 and Hes1 protein and the Hes1 mRNA expression in the podocytes stimulated with Ang II at 12 h (all P<0.01; Fig. 2). Western blot analysis and RT-PCR revealed that GSI did not inhibit Notch1 overexpression induced by Ang II at 12 h (all P>0.05). Ang II stimulation decreased MMP-2 and MMP-9 protein and mRNA expression at 12 h (all P<0.01; Fig. 3). Compared with the cells stimulated with Ang II, the MMP-2 and MMP-9 protein and mRNA levels significantly increased in the podocytes transfected with sh-Notch1 or pre-treated with GSI (all P<0.01). Incubation with Ang II for 12 h resulted in a significant upregulation in TGF-β1 protein and mRNA expression compared with the cells stimulated with Ang II at 0 h (all P<0.01). However, the changes observed in the TGF-β1 expression level in the Ang II-stimulated podocytes were reversed by transfection with sh-Notch1 or by the addition of GSI to the Ang II culture medium (all P<0.01).

Transfection with sh-Notch1 or pre-treatment with GSI increases MMP activity in the Ang II-stimulated podocytes

When the podocytes were incubated with Ang II for 12 h, Ang II markedly decreased MMP-2 and MMP-9 activity. However, transfection with sh-Notch1 or pre-treatment with GSI enhanced the Ang II-induced inhibition of MMP-2 and MMP-9 activity (all P<0.01; Fig. 4).

Transfection with sh-Notch1 or pre-treatment with GSI decreases type IV collagen and laminin expression in the Ang II-stimulated podocytes

As shown by western blot analysis (Fig. 5A and B), type IV collagen and laminin protein expression was induced by Ang II at 12 h compared with the podocytes treated with Ang II at 0 h; these expression levels were efficiently inhibited by transfection with sh-Notch1 or pre-treatment with GSI (P<0.05 or P<0.01). Compared with the podocytes stimulated with Ang II at 0 h, the mRNA levels of type IV collagen and laminin were increased in the Ang II-stimulated podocytes at 12 h. Their mRNA expression was inhibited following transfection with sh-Notch1 or pretreatment with GSI (all P<0.01; Fig. 5C).

Transfection with sh-Notch1 or pre-treatment with GSI decreases Ang II-induced TGF-β1, type IV collagen and laminin secretion

We also examined the concentrations of TGF-β1, type IV collagen and laminin in the culture medium of the podocytes using ELISA (Fig. 6). We found that the podocytes stimulated with Ang II for 12 h showed higher levels of TGF-β1, type IV collagen and laminin in the supernatants than those cultured with Ang II at 0 h (P<0.05 or P<0.01). Compared to the podocytes stimulated with Ang II, which induced the overexpression of TGF-β1, type IV collagen and laminin in the supernatants, transfection with sh-Notch1 or pre-treatment with GSI significantly decreased the expression of TGF-β1, type IV collagen and laminin (P<0.05 or P<0.01; Fig. 6).

Discussion

The Notch pathway is an evolutionarily conserved local cell-signaling mechanism that participates in a variety of cellular processes and is important in glomerular development (20,21). It has been found that the activation of the Notch pathway in podocytes plays a role in the development of glomerular disease (4). In our previous study, it was revealed that the activation of the Notch pathway in high glucose-stimulated podocytes altered the apoptotic pathway and induced apoptosis (22). Alterations in hemodynamics in glomerular disease induce higher levels of Ang II, which also interacts with the Notch pathway in podocytes (11). In this study, we also observed the activation of the Notch pathway in Ang II-stimulated podocytes. Notch1 seems to be a modulatory target of Ang II in podocytes. Ang II induces Notch1 cleavage in podocytes and the release of NICD1, which travels to the nucleus where it activates the Hes1 gene. The activation of the Notch pathway induced by Ang II was inhibited by Notch1 shRNA or GSI.

ECM accumulation in glomeruli is considered the most common destructive pathway associated with chronic glomerular disease, which is characterized by the remodeling of the interstitial ECM, resulting in the excessive deposition of ECM components, including type IV collagen and laminin (23). MMPs are zinc-containing endopeptidases that are involved in the remodeling of the ECM and are crucial for tissue development and homeostasis. MMP-2 and MMP-9 cleave the denatured collagens and laminin, as well as some chemokines. In view of their matrix-degrading capacity, MMP-2 and MMP-9 were originally considered to be beneficial to chronic renal fibrogenesis due to their perceived potential to lessen interstitial matrix accumulation and deposition (12,13,24). Studies on renal fibrosis have demonstrated that a decrease in MMP levels promotes the deposition of ECM in the context of renal fibrosis (11,25). Ang II has been shown to inhibit the angiotensin converting enzyme II (ACE2)-induced MMP-2 activity through the Ang II type-1 receptor (AT1R) and extracellular signal-regulated kinase 1 and 2 (ERK1/2) signaling pathway in human cardiofibroblasts (26). Under normal glucose conditions, Ang II induces a dose-dependent downregulation in MMP-2; on the contrary, MMP-9 is upregulated by Ang II (27). However, we found that Ang II inhibited the protein and mRNA expression, as well as the activity of MMP-2 and MMP-9 in podocytes. An enhanced MMP activity has been shown to be responsible for the development of disease due to their ability to degrade type IV collagen and laminin (28,29). Zhou et al (30) found that the downregulation of Notch1 decreased the migration and invasion capacities of hepatocellular carcinoma cells by regulating CD44v6, E-cadherin, MMP-2, MMP-9 and urokinase-type plasminogen activator (uPA). In this study, after the activation of the Notch pathway was inhibited by Notch1 shRNA or GSI in Ang II-stimulated podocytes, MMP-2 and MMP-9 expression and activity increased. These results indicated that Ang II inhibited MMP-2 and MMP-9 through the Notch pathway in podocytes.

It is well established that TGF-β1 is a potent stimulator of ECM production in glomerular injury and may be the most important growth factor in determining the extent of renal fibrosis following injury. It has been demonstrated that TGF-β1 mediates the production of type IV collagen and laminin in mesangial cells under conditions of high glucose in vitro (31). The exposure of cultured conditionally immortalized human podocytes to TGF-β1 has also been shown to increase the production of the basement membrane components (32). In the present study, we found that the stimulation of podocytes with Ang II induced type IV collagen and laminin accumulation by increasing TGF-β1 mRNA and protein synthesis. Ang II has been shown to induce the expression and production of TGF-β1 through a mechanism dependent on reactive oxygen species production in mouse skeletal muscle cells (33). In another study, Ang II-induced cardiac hypertrophy and fibrosis were significantly enhanced by the increase in TGF-β1 expression in fibroblast growth factor 16 (Fgf16) knockout mice (34). Furthermore, our finding that the inhibition of the Notch pathway by Notch1 shRNA or GSI markedly prevented the Ang II-induced upregulation of TGF-β1 suggests that Ang II induces TGF-β1 expression through the Notch pathway. Aoyagi-Ikeda et al (35) found that Notch induced myofibro-blast differentiation through a TGF-β-Smad3 pathway that activated SMA gene transcription in alveolar epithelial cells, and increased migratory behavior in pulmonary fibrosis. In a previous study, blocking Notch signaling by the γ-secretase inhibitor, DAPT, was shown to significantly attenuate liver fibrosis, decrease the expression of TGF-β1 and suppress the endothelial-to-mesenchymal transition (EMT) process in a rat hepatic stellate cell line (36). In this study, we found that the activity of MMP-2 and MMP-9 was reduced, and TGF-β1 expression was increased in Ang II-stimulated podocytes. Imbalances between the synthesis and degradation of ECM proteins are considered to play important roles in the progression of glomerular sclerosis in glomerular disease (37).

In conclusion, our data demonstrate that the Ang II-induced activation of the Notch pathway, which inhibited MMP activity, increased TGF-β1 expression and increased the synthesis of ECM components in podocytes. Thus, the activation of the Notch pathway plays a role in Ang II-induced glomerular injury. The blockade of the Notch pathway may thus be an effective method for the treatment of glomerular disease.

Acknowledgments

This study was supported by grants from the Hebei Natural Science Foundation of China (H2014206294) and the Department of Health of Hebei Province of China (ZL20140030).

References

1 

Ikezumi Y, Suzuki T, Karasawa T, Yamada T, Hasegawa H, Nishimura H and Uchiyama M: Low birthweight and premature birth are risk factors for podocytopenia and focal segmental glomerulosclerosis. Am J Nephrol. 38:149–157. 2013. View Article : Google Scholar : PubMed/NCBI

2 

Ma R, Liu L, Liu X, Wang Y, Jiang W and Xu L: Triptolide markedly attenuates albuminuria and podocyte injury in an animal model of diabetic nephropathy. Exp Ther Med. 6:649–656. 2013.PubMed/NCBI

3 

Sirin Y and Susztak K: Notch in the kidney: development and disease. J Pathol. 226:394–403. 2012. View Article : Google Scholar

4 

Niranjan T, Bielesz B, Gruenwald A, Ponda MP, Kopp JB, Thomas DB and Susztak K: The Notch pathway in podocytes plays a role in the development of glomerular disease. Nat Med. 14:290–298. 2008. View Article : Google Scholar : PubMed/NCBI

5 

Zhou W, Wang G and Guo S: Regulation of angiogenesis via Notch signaling in breast cancer and cancer stem cells. Biochim Biophys Acta. 1836:304–320. 2013.PubMed/NCBI

6 

Ji X, Wang Z, Geamanu A, Sarkar FH and Gupta SV: Inhibition of cell growth and induction of apoptosis in non-small cell lung cancer cells by delta-tocotrienol is associated with notch-1 down-regulation. J Cell Biochem. 112:2773–2783. 2011. View Article : Google Scholar : PubMed/NCBI

7 

Wang W, Li L, Zhou Z, Gao J and Sun Y: Effect of spironolactone combined with angiotensin-converting enzyme inhibitors and/or angiotensin II receptor blockers on chronic glomerular disease. Exp Ther Med. 6:1527–1531. 2013.PubMed/NCBI

8 

Ding Y, Stidham RD, Bumeister R, Trevino I, Winters A, Sprouse M, Ding M, Ferguson DA, Meyer CJ, Wigley WC and Ma R: The synthetic triterpenoid, RTA 405, increases the glomerular filtration rate and reduces angiotensin II-induced contraction of glomerular mesangial cells. Kidney Int. 83:845–854. 2013. View Article : Google Scholar :

9 

Siragy HM and Carey RM: Role of the intrarenal renin-angiotensin-aldosterone system in chronic kidney disease. Am J Nephrol. 31:541–550. 2010. View Article : Google Scholar : PubMed/NCBI

10 

Yang S, Yao B, Zhou Y, Yin H, Zhang MZ and Harris RC: Intrarenal dopamine modulates progressive angiotensin II-mediated renal injury. Am J Physiol Renal Physiol. 302:F742–F749. 2012. View Article : Google Scholar :

11 

Koshizaka M, Takemoto M, Sato S, Tokuyama H, Fujimoto M, Okabe E, Ishibashi R, Ishikawa T, Tsurutani Y, Onishi S, Mezawa M, He P, Honjo S, Ueda S, Saito Y and Yokote K: An angiotensin II type 1 receptor blocker prevents renal injury via inhibition of the Notch pathway in Ins2 Akita diabetic mice. Exp Diabetes Res. 2012:1598742012. View Article : Google Scholar : PubMed/NCBI

12 

Zhou TB, Qin YH, Lei FY, Su LN, Zhao YJ and Huang WF: Less gelatinases is associated with apolipoprotein E accumulation in glomerulosclerosis rats. Histol Histopathol. 27:249–256. 2012.

13 

Ji L, Yin XX, Wu ZM, Wang JY, Lu Q and Gao YY: Ginkgo biloba extract prevents glucose-induced accumulation of ECM in rat mesangial cells. Phytother Res. 23:477–485. 2009. View Article : Google Scholar

14 

Wang B, Komers R, Carew R, Winbanks CE, Xu B, Herman-Edelstein M, Koh P, Thomas M, Jandeleit-Dahm K, Gregorevic P, Cooper ME and Kantharidis P: Suppression of microRNA-29 expression by TGF-β1 promotes collagen expression and renal fibrosis. J Am Soc Nephrol. 23:252–265. 2012. View Article : Google Scholar :

15 

Schena FP and Gesualdo L: Pathogenetic mechanisms of diabetic nephropathy. J Am Soc Nephrol. 16(Suppl 1): S30–S33. 2005. View Article : Google Scholar : PubMed/NCBI

16 

Singh LP, Green K, Alexander M, Bassly S and Crook ED: Hexosamines and TGF-beta1 use similar signaling pathways to mediate matrix protein synthesis in mesangial cells. Am J Physiol Renal Physiol. 286:F409–F416. 2004. View Article : Google Scholar

17 

Han SY, Jee YH, Han KH, Kang YS, Kim HK, Han JY, Kim YS and Cha DR: An imbalance between matrix metalloproteinase-2 and tissue inhibitor of matrix metalloproteinase-2 contributes to the development of early diabetic nephropathy. Nephrol Dial Transplant. 21:2406–2416. 2006. View Article : Google Scholar : PubMed/NCBI

18 

Liu GX, Li YQ, Huang XR, Wei L, Chen HY, Shi YJ, Heuchel RL and Lan HY: Disruption of Smad7 promotes Ang II-mediated renal inflammation and fibrosis via Sp1-TGF-β/Smad3-NF-κB-dependent mechanisms in mice. PLoS One. 8:e535732013. View Article : Google Scholar

19 

Mundel P, Reiser J, Zúñiga Mejía Borja A, Pavenstädt H, Davidson GR, Kriz W and Zeller R: Rearrangements of the cytoskeleton and cell contacts induce process formation during differentiation of conditionally immortalized mouse podocyte cell lines. Exp Cell Res. 236:248–258. 1997. View Article : Google Scholar : PubMed/NCBI

20 

Cheng HT, Kim M, Valerius MT, Surendran K, Schuster-Gossler K, Gossler A, McMahon AP and Kopan R: Notch2, but not Notch1, is required for proximal fate acquisition in the mammalian nephron. Development. 134:801–811. 2007. View Article : Google Scholar : PubMed/NCBI

21 

Arumugam TV, Chan SL, Jo DG, Yilmaz G, Tang SC, Cheng A, Gleichmann M, Okun E, Dixit VD, Chigurupati S, Mughal MR, Ouyang X, Miele L, Magnus T, Poosala S, Granger DN and Mattson MP: Gamma secretase-mediated Notch signaling worsens braindamage and functional outcome in ischemic stroke. Nat Med. 12:621–623. 2006. View Article : Google Scholar : PubMed/NCBI

22 

Gao F, Yao M, Shi Y, Hao J, Ren Y, Liu Q, Wang X and Duan H: Notch pathway is involved in high glucose-induced apoptosis in podocytes via Bcl-2 and p53 pathways. J Cell Biochem. 114:1029–1038. 2013. View Article : Google Scholar

23 

Lennon R, Byron A, Humphries JD, Randles MJ, Carisey A, Murphy S, Knight D, Brenchley PE, Zent R and Humphries MJ: Global analysis reveals the complexity of the human glomerular extracellular matrix. J Am Soc Nephrol. 25:939–951. 2014. View Article : Google Scholar : PubMed/NCBI

24 

Cheng S, Pollock AS, Mahimkar R, Olson JL and Lovett DH: Matrix metalloproteinase 2 and basement membrane integrity: a unifying mechanism for progressive renal injury. FASEB J. 20:1898–1900. 2006. View Article : Google Scholar : PubMed/NCBI

25 

Tsai JP, Liou JH, Kao WT, Wang SC, Lian JD and Chang HR: Increased expression of intranuclear matrix metalloproteinase 9 in atrophic renal tubules is associated with renal fibrosis. PLoS One. 7:e481642012. View Article : Google Scholar : PubMed/NCBI

26 

Kuan TC, Chen MY, Liao YC, Ko L, Hong YH, Yen CY, Hsieh WY, Cheng KS, Wu CL and Lin CS: Angiotensin II down-regulates ACE2-mediated enhancement of MMP-2 activity in human cardiofibroblasts. Biochem Cell Biol. 91:435–442. 2013. View Article : Google Scholar : PubMed/NCBI

27 

Solini A, Rossi C, Santini E, Madec S, Salvati A and Ferrannini E: Angiotensin-II and rosuvastatin influence matrix remodeling in human mesangial cells via metalloproteinase modulation. J Hypertens. 29:1930–1939. 2011. View Article : Google Scholar : PubMed/NCBI

28 

Czech KA, Bennett M and Devarajan P: Distinct metalloproteinase excretion patterns in focal segmental glomerulosclerosis. Pediatr Nephrol. 26:2179–2184. 2011. View Article : Google Scholar : PubMed/NCBI

29 

Sun H, Ge N, Shao M, Cheng X, Li Y, Li S and Shen J: Lumbrokinase attenuates diabetic nephropathy through regulating extracellular matrix degradation in Streptozotocin-induced diabetic rats. Diabetes Res Clin Pract. 100:85–95. 2013. View Article : Google Scholar : PubMed/NCBI

30 

Zhou L, Zhang N, Song W, You N, Li Q, Sun W, Zhang Y, Wang D and Dou K: The significance of Notch1 compared with Notch3 in high metastasis and poor overall survival in hepatocellular carcinoma. PLoS One. 8:e573822013. View Article : Google Scholar : PubMed/NCBI

31 

Tang DQ, Wei YQ, Yin XX, Lu Q, Hao HH, Zhai YP, Wang JY and Ren J: In vitro suppression of quercetin on hypertrophy and extracellular matrix accumulation in rat glomerular mesangial cells cultured by high glucose. Fitoterapia. 82:920–926. 2011. View Article : Google Scholar : PubMed/NCBI

32 

Herman-Edelstein M, Thomas MC, Thallas-Bonke V, Saleem M, Cooper ME and Kantharidis P: Dedifferentiation of immortalized human podocytes in response to transforming growth factor-β: a model for diabetic podocytopathy. Diabetes. 60:1779–1788. 2011. View Article : Google Scholar : PubMed/NCBI

33 

Morales MG, Abrigo J, Meneses C, Simon F, Cisternas F, Rivera JC, Vazquez Y and Cabello-Verrugio C: The Ang-(1–7)/Mas-1 axis attenuates the expression and signalling of TGF-β1 induced by AngII in mouse skeletal muscle. Clin Sci (Lond). 127:251–264. 2014. View Article : Google Scholar

34 

Matsumoto E, Sasaki S, Kinoshita H, Kito T, Ohta H, Konishi M, Kuwahara K, Nakao K and Itoh N: Angiotensin II-induced cardiac hypertrophy and fibrosis are promoted in mice lacking Fgf16. Genes Cells. 18:544–553. 2013. View Article : Google Scholar : PubMed/NCBI

35 

Aoyagi-Ikeda K, Maeno T, Matsui H, Ueno M, Hara K, Aoki Y, Aoki F, Shimizu T, Doi H, Kawai-Kowase K, Iso T, Suga T, Arai M and Kurabayashi M: Notch induces myofibroblast differentiation of alveolar epithelial cells via transforming growthfactor-{beta}-Smad3 pathway. Am J Respir Cell Mol Biol. 45:136–144. 2011.PubMed/NCBI

36 

Chen Y, Zheng S, Qi D, Zheng S, Guo J, Zhang S and Weng Z: Inhibition of Notch signaling by a γ-secretase inhibitor attenuates hepatic fibrosis in rats. PLoS One. 7:e465122012. View Article : Google Scholar

37 

Tsioufis C, Bafakis I, Kasiakogias A and Stefanadis C: The role of matrix metalloproteinases in diabetes mellitus. Curr Top Med Chem. 12:1159–1165. 2012. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2015
Volume 36 Issue 1

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yao M, Wang X, Wang X, Zhang T, Chi Y and Gao F: The Notch pathway mediates the angiotensin II-induced synthesis of extracellular matrix components in podocytes. Int J Mol Med 36: 294-300, 2015
APA
Yao, M., Wang, X., Wang, X., Zhang, T., Chi, Y., & Gao, F. (2015). The Notch pathway mediates the angiotensin II-induced synthesis of extracellular matrix components in podocytes. International Journal of Molecular Medicine, 36, 294-300. https://doi.org/10.3892/ijmm.2015.2193
MLA
Yao, M., Wang, X., Wang, X., Zhang, T., Chi, Y., Gao, F."The Notch pathway mediates the angiotensin II-induced synthesis of extracellular matrix components in podocytes". International Journal of Molecular Medicine 36.1 (2015): 294-300.
Chicago
Yao, M., Wang, X., Wang, X., Zhang, T., Chi, Y., Gao, F."The Notch pathway mediates the angiotensin II-induced synthesis of extracellular matrix components in podocytes". International Journal of Molecular Medicine 36, no. 1 (2015): 294-300. https://doi.org/10.3892/ijmm.2015.2193