Acidosis promotes cell apoptosis through the G protein‑coupled receptor 4/CCAAT/enhancer‑binding protein homologous protein pathway

  • Authors:
    • Biao Dong
    • Xiaolu Zhang
    • Yafeng Fan
    • Songqiang Cao
    • Xuepei Zhang
  • View Affiliations

  • Published online on: September 21, 2018     https://doi.org/10.3892/ol.2018.9478
  • Pages: 6735-6741
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The aim of the present study was to investigate the effects of acidosis on the apoptosis of renal epithelial and endothelial cells, and the molecular pathways responsible for this. A human proximal tubular cell line, HK‑2, and human umbilical vein endothelial cells (HUVECs), were transfected with control or G protein‑coupled receptor 4 siRNA for 36 h. Cells were exposed to normal (pH 7.4) or acidic (pH 6.4) media. Western blot analysis was used to assess the protein expression levels of G protein‑coupled receptor 4 (GPR4), CCAAT/enhancer‑binding protein homologous protein (CHOP) and cleaved caspase‑3. Cell apoptosis was examined using the TUNEL assay and the lactate dehydrogenase (LDH) release assay. Using these techniques, it was demonstrated that acidosis increased the protein expression levels of GPR4, CHOP, cleaved caspase‑3 and intracellular cyclic adenosine monophosphate levels in hypoxia/reoxygenation (HR)‑treated cell lines. Knockdown of GPR4 in HK‑2 cells and HUVECs markedly reduced the protein expression levels of acidosis‑mediated GPR4, CHOP and cleaved caspase‑3, as well as the rate of cell apoptosis. Therefore, the results of the present study suggested that acidosis promotes the apoptosis of HK‑2 cells and HUVECs by regulating the GPR4/CHOP pathway.

Introduction

Acidosis is associated with various pathological conditions, including ischemia (restricted blood supply), diabetic ketoacidosis, defective blood flow, defective glycolytic metabolism, and lung and renal diseases (13). During acidosis, hydrogen ions accumulate in ischemic tissue, leading to intracellular acidosis (4). Within 5 min of the initiation of renal ischemia, the tissue pH drops from 7.2–6.5 and metabolic acidosis persists even with reperfusion (4). Acidosis causes tissue injury and promotes the progression of existing disease, such as stroke (5). However, the specific effects of acidosis on renal epithelial and endothelial cells, and the molecular pathways responsible for these effects, remain unknown.

A previous study suggested that G protein-coupled receptor 4 (GPR4) may be activated by a decrease in extracellular pH, which generates a second messenger protein (6). GPR4 acts as a pH sensor in the kidney, mediating the accumulation of intracellular cyclic adenosine monophosphate (cAMP), which regulates the activity of acid-base transport proteins in the kidney collecting duct (6,7). A previous study using GPR4-knockout mice has also indicated that GPR4 is involved in renal acid-base homeostasis (3).

CCAAT/enhancer-binding protein homologous protein (CHOP) is a transcriptional regulator and is an essential regulator of the endoplasmic reticulum stress (ERS)-mediated apoptosis pathway (8). CHOP is a pro-apoptotic protein and serves key roles in the pathology of liver fibrosis (9). It has been reported that the deletion of CHOP may protect mice from liver and kidney injury (10). A previous study reported that acidosis activates GPR4 in order to increase the expression of ERS-associated genes, including CHOP, in endothelial cells (11). Our previous study demonstrated that knockdown of GPR4 inhibited the expression of CHOP in hypoxia/reoxygenation (HR)-treated human umbilical vein endothelial cells (HUVECs) (12). However, to the best of our knowledge, the effect of GPR4 on the expression of CHOP in renal cells in response to acidosis remains uncharacterized. Therefore, the aim of the present study was to investigate the effects of acidosis on the apoptosis of renal epithelial cells and endothelial cells and the molecular pathways responsible for these effects.

Materials and methods

Cell culture and treatment

The human umbilical vein endothelial cell line, HUVEC, was obtained from All Cells LLC. (Alameda, CA, USA), and the human proximal tubular HK-2 cell line was obtained from the American Type Culture Collection (Manassas, VA, USA). Cells were cultured in Dulbecco's modified Eagle medium (DMEM; Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA) containing 10% fetal bovine serum (Gibco; Thermo Fisher Scientific, Inc.) in 5% CO2 at 37°C.

For hypoxia/reoxygenation (HR) treatment, 5×105 cells in 2 ml media were placed into each well of 6-well flat-bottomed plates at 37°C. Cells were exposed to hypoxia (1% O2) for 4 h, followed by a 6 h re-oxygenation (95% O2) period. Subsequently, HUVECs and HK-2 cells were subjected to acidosis by adjusting the media to pH 6.4 by applying 0.1 M HCl for 12 h prior to harvesting.

siRNA transfection

Cell transfection was performed as previously described (13). In brief, cells were seeded onto 24-well pates at 1×105 cells/well. Cells were then transfected with 80 nM siRNA using Lipofectamine® 2000 (Thermo Fisher Scientific, Inc.), according to the manufacturer's protocol. The non-target scrambled siRNA duplexes (Shanghai GenePharma Co., Ltd., Shanghai, China; cat. no. A06001) and GPR4 siRNA (siGPR4) duplexes were purchased from Invitrogen; Thermo Fisher Scientific, Inc. The sequences of si-GPR4 were as follows: Forward, 3′-CCCUCUACAUCUUUGUCAUTT-5′ and reverse, 3′-AUGACAAAGAUGUAGAGGGTT-5′ (12). The cells were harvested after 36 h of transfection.

Western blot analysis

Total cell protein was extracted from HUVECs and HK-2 cells using a Nuclear Extract kit (Active Motif, Carlsbad, CA, USA), according to the manufacturer's protocols. Equal amounts of protein (60 µg) were separated by 10% SDS-PAGE, prior to being transferred onto Whatman nitrocellulose membranes (Sigma-Aldrich; Merck KGaA, Darmstadt, Germany). Subsequent to blocking in 5% skimmed milk for 1 h at 37°C, the membranes were incubated with primary antibodies against the following overnight at 4°C: GPR4 (dilution, 1:1,000; R&D Systems, Inc., Minneapolis, MN, USA; cat. no. RDC0299), CHOP (dilution, 1:1,000; Cell Signaling Technology, Inc., Danvers, MA, USA; cat. no. 2895), cleaved caspase-3 (dilution, 1:1,000; Cell Signaling Technology, Inc.; cat. no. 9661) and GAPDH (dilution, 1:2,000; Santa Cruz Biotechnology, Inc., Dallas, TX, USA; cat. no. sc-32233). Following 3 washes in Tris-buffered saline with 0.1% Tween-20 (TBST), the membranes were incubated with a horseradish peroxidase-conjugated mouse anti-rabbit secondary antibody (dilution, 1:10,000; Cell Signaling Technology, Inc.; cat. no. 5127) for 1 h at 37°C. The blot was visualized using an enhanced chemiluminescence kit (GE Healthcare, Chicago, IL, USA). Densitometry was performed using ImageJ software (version 1.43a; National Institutes of Health, Bethesda, MD, USA) and GAPDH was used as a loading control.

Measurement of cAMP levels

Cells were treated with 50 mM 3-isobutyl-1-methylxanthine (Sigma-Aldrich; Merck KGaA) for 30 min, followed by HR treatment, performed as aforementioned. Subsequently, 200 µl 0.1 M HCl was added to lyse the cells for 12 h at 37°C. The lysate was boiled at 90°C for 5 min and centrifuged at 10,000 × g at 4°C for 5 min prior to the supernatant being collected. The cAMP levels were measured using a cAMP ELISA kit (cat no. KGE002; R&D Systems Inc.), according to the manufacturer's protocol.

TUNEL assay

A TUNEL assay was performed using the In Situ Cell Death Detection kit (Roche Diagnostics, Basel, Switzerland). In brief, cells were fixed with 4% paraformaldehyde for 1 h at room temperature and were permeabilized with 0.1% Triton X-100 in PBS for 2 min on ice. Subsequent to rinsing with PBS, the cells were incubated with the TUNEL reaction mixture for 60 min at 37°C. Cell nuclei were counterstained with 10 mg/ml 4′,6-diamidino-2-phenylindole for 1 h at 37°C. Finally, the slides were mounted with synthetic resin and observed under a fluorescence microscope (original magnification, ×400; BX-53; Olympus Corporation, Tokyo, Japan). The total number of TUNEL positive cells was calculated in ≥3 fields of view.

Measurement of lactate dehydrogenase (LDH) release

At 36 h after cell transfection, the supernatant was collected from both HK-2 and HUVEC cells after centrifugation at 3,000 × g for 5 min at room temperature and was measured using the CytoTox 96 Non-Radioactive Cytotoxicity assay (Promega Corporation, Madison, WI, USA), according to the manufacturer's protocol. The results are expressed as the percentage of maximal LDH released into the culture medium.

Statistical analysis

All results are expressed as the mean ± standard deviation. The statistical analyses were performed using GraphPad Prism 6.0 software (GraphPad Software, Inc., La Jolla, CA, USA). Comparisons between 2 groups were performed using unpaired Student's t-tests, while comparisons between ≥3 groups were performed using one-way analysis of variance, following by Bonferroni's post-hoc test. P<0.05 was considered to indicate a statistically significant difference.

Results

Acidosis enhances HR-induced GPR4 and CHOP expression, as well as apoptosis

Renal epithelial and endothelial cells are susceptible to ischemic injury, which may lead to renal injury (14). In order to investigate the effect of HR on the expression of GPR4 and CHOP in the two cell types, HK-2 cells were confronted with HR injury and were experimentally assessed. As demonstrated in Fig. 1A and B, the expression levels of GPR4 and CHOP proteins were significantly increased in both HK-2 cells and HUVECs following HR treatment or HCl treatment alone, compared with the control group with a normal pH. The expression levels of GPR4 and CHOP were upregulated in the HR group treated with HCl compared with the cells treated with HR alone. Furthermore, the expression of cleaved caspase-3 was also increased by HCl treatment in the HR group compared with the HR group with a normal pH.

Intracellular cAMP is a downstream effector of GPR4 (15). The expression level of cAMP was increased in the cells exposed to HR and subsequent HCl treatment, compared with the cells treated with HR alone (Fig. 1C). The same changes of gene expressions (Fig. 2A and B) and cAMP levels (Fig. 2C) existed in HR-treated HUVECs. These results suggested that the HR-induced increase in GPR4 and CHOP expression and the apoptosis of HK-2 cells and HUVECs may result from acidosis.

GPR4-knockdown attenuates the GPR4 and CHOP expression, and the cell apoptosis induced by acidosis

To determine whether GPR4 function is associated with acidosis-induced GPR4/CHOP expression or cell apoptosis, GPR4 expression was knocked down by si-GPR4 in HK-2 cells and HUVECs. Acidosis resulted in an increase in the protein expression levels of GPR4, CHOP and cleaved caspase-3 in the HK-2 cells and HUVECs transfected with scrambled siRNA. The expression of these proteins and the acidosis-induced increase in intracellular cAMP expression levels in HK-2 cells and HUVECs were inhibited by knockdown of GPR4 (Figs. 3A-C; 4A-C).

GPR4-knockdown leads to a decrease in acidosis-induced apoptosis

To confirm the role of GPR4 in acidosis-induced cell apoptosis, a TUNEL assay was performed. TUNEL staining indicated an increased number of TUNEL positive HK-2/vector cells in response to acidic pH compared with the control group under normal pH conditions (Fig. 5A), whilst the increase in TUNEL positive cells induced by acidosis was attenuated by si-GPR4 treatment. These results were confirmed by measuring the release of LDH, which is a marker of cellular injury. Under acidic conditions, LDH release was significantly reduced in cells transfected with si-GPR4 compared with cells transfected with scrambled siRNA (Fig. 5B). Similarly, GPR4 interference led to a decrease in the number of TUNEL positive HUVEC/vector cells (Fig. 5C) and LDH release from HUVECs (Fig. 5D). The results indicated that GPR4 expression was required for acidosis-induced apoptosis of the HK-2 cells and HUVECs.

Discussion

Acidosis is a pathological condition whereby acidity over-accumulates in the body fluid (16). The condition aggravates ischemia-associated tissue injury (17), and metabolic acidosis has been demonstrated to exacerbate the renal injury induced by ischemia/reperfusion (IR) (18). In addition, acidosis has been demonstrated to induce cell apoptosis and inhibit cell proliferation (19). It has been demonstrated that during stroke, the pH of brain tissue may drop to 6.0–6.5 and that this acidotic stress increases neural cell apoptosis (20). Our previous study indicated that HR-induced apoptosis was alleviated by neutralization of acidity (12). The results of the present study demonstrated that acidosis promoted HR-induced apoptosis. Taken together, these results suggest that HR-induced cell apoptosis in HK-2 cells and HUVECs may result from acidosis.

It has been reported that GPR4 is involved in the regulation of acid-base balance and kidney acid secretion (6). Acidosis may activate the expression of GPR4 in order to regulate the expression of the genes involved in different biological pathways, including the ERS response and cell metabolism (3,21). A previous study demonstrated that acidosis/GPR4 regulated endothelial cell adhesion through cAMP production and served a role in the inflammatory response of vascular endothelial cells (3). In the present study, it was demonstrated that acidosis enhanced HR-induced GPR4 protein expression in HK-2 cells and HUVECs. CHOP is the key inducer of apoptosis in the ERS response (22), and is induced in hypoxia-treated renal tubular epithelial cells and promotes apoptosis (23). Our previous study indicated that hypoxia-induced CHOP expression is mediated by GPR4 (12). This result is supported by those of the present study, in which acidosis was demonstrated to induce the expression of CHOP. However, this effect was diminished by GPR4 knockdown in HK-2 cells and HUVECs. Therefore, acidosis-induced CHOP expression was regulated by GPR4 in HK-2 cells and HUVECs.

An association was identified between HR and cell apoptosis in vitro. Furthermore, the observation that inhibition of GRP4 expression reduces the cell apoptosis induced by acidosis contributes to the characterization of the role of the GPR4/CHOP pathway in the pathogenesis of IR injury, and suggests a potential molecular candidate for IR injury therapeutics. cAMP is a ubiquitous second messenger protein that regulates numerous cellular processes (24). Previous studies have demonstrated that an acidic pH activates GPR4 in order to induce the production of cAMP (15). Chen et al (3) reported that activation of GPR4 by acidosis increased endothelial cell adhesion via the cAMP pathway. Our previous study demonstrated that a cAMP analog, 8-bromo-cAMP, induced CHOP expression in HUVECs (18). The present study demonstrated that GPR4 expression reduced acidosis-induced cAMP and CHOP expression levels in HK-2 cells and HUVECs. Taken together, these results suggested that acidosis-induced CHOP expression may result from GPR4 activation and cAMP accumulation.

In conclusion, the present study demonstrated that HR-induced cell apoptosis in HK-2 cells and HUVECs may result from acidosis. The CHOP pathway serves a key role in acidosis-induced cell apoptosis. These results indicated that GPR4 inhibitors may be promising and effective therapeutic tools for the treatment of ischemic renal injury.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

All data that were generated or analyzed in this study are included in this manuscript.

Authors' contributions

BD designed the present study. XLZ and YF conducted the experiments. SC performed the statistical analysis. XPZ interpreted the statistical analysis, and reviewed and approved the final version to be published. All authors read and approved the manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Koul PB: Diabetic ketoacidosis: A current appraisal of pathophysiology and management. Clin Pediatr (Phila). 48:135–144. 2009. View Article : Google Scholar : PubMed/NCBI

2 

Poschet J, Perkett E and Deretic V: Hyperacidification in cystic fibrosis: Links with lung disease and new prospects for treatment. Trends Mol Med. 8:512–519. 2002. View Article : Google Scholar : PubMed/NCBI

3 

Chen A, Dong L, Leffler NR, Asch AS, Witte ON and Yang LV: Activation of GPR4 by acidosis increases endothelial cell adhesion through the cAMP/Epac pathway. PLoS One. 6:e275862011. View Article : Google Scholar : PubMed/NCBI

4 

Bonventre JV: Mediators of ischemic renal injury. Annu Rev Med. 39:531–544. 1988. View Article : Google Scholar : PubMed/NCBI

5 

Xiong ZG, Zhu XM, Chu XP, Minami M, Hey J, Wei WL, MacDonald JF, Wemmie JA, Price MP, Welsh MJ and Simon RP: Neuroprotection in Ischemia: Blocking calcium-permeable acid-sensing ion channels. Cell. 118:687–698. 2004. View Article : Google Scholar : PubMed/NCBI

6 

Sun X, Yang LV, Tiegs BC, Arend LJ, McGraw DW, Penn RB and Petrovic S: Deletion of the pH sensor GPR4 decreases renal acid excretion. J Am Soc Nephrol. 21:1745–1755. 2010. View Article : Google Scholar : PubMed/NCBI

7 

Brown D, Breton S, Ausiello DA and Marshansky V: Sensing, signaling and sorting events in kidney epithelial cell physiology. Traffic. 10:275–284. 2009. View Article : Google Scholar : PubMed/NCBI

8 

Nishitoh H: CHOP is a multifunctional transcription factor in the ER stress response. J Biochem. 151:217–219. 2012. View Article : Google Scholar : PubMed/NCBI

9 

Tamaki N, Hatano E, Taura K, Tada M, Kodama Y, Nitta T, Iwaisako K, Seo S, Nakajima A, Ikai I and Uemoto S: CHOP deficiency attenuates cholestasis-induced liver fibrosis by reduction of hepatocyte injury. Am J Physiol Gastrointest Liver Physiol. 294:G498–G505. 2008. View Article : Google Scholar : PubMed/NCBI

10 

Prachasilchai W, Sonoda H, Yokota-Ikeda N, Ito K, Kudo T, Imaizumi K and Ikeda M: The protective effect of a newly developed molecular chaperone-inducer against mouse ischemic acute kidney injury. J Pharmacol Sci. 109:311–314. 2009. View Article : Google Scholar : PubMed/NCBI

11 

Dong L, Li Z, Leffler NR, Asch AS, Chi JT and Yang LV: Acidosis activation of the proton-sensing GPR4 receptor stimulates vascular endothelial cell inflammatory responses revealed by transcriptome analysis. PLoS One. 8:e619912013. View Article : Google Scholar : PubMed/NCBI

12 

Dong B, Zhou H, Han C, Yao J, Xu L, Zhang M, Fu Y and Xia Q: Ischemia/reperfusion-induced CHOP expression promotes apoptosis and impairs renal function recovery: The role of acidosis and GPR4. PLoS One. 9:e1109442014. View Article : Google Scholar : PubMed/NCBI

13 

He K, Chen X, Han C, Xu L, Zhang J, Zhang M and Xia Q: Lipopolysaccharide-induced cross-tolerance against renal ischemia-reperfusion injury is mediated by hypoxia-inducible factor-2α-regulated nitric oxide production. Kidney Int. 85:276–288. 2014. View Article : Google Scholar : PubMed/NCBI

14 

Miao J, Lesher AM, Miwa T, Sato S, Gullipalli D and Song WC: Tissue-specific deletion of Crry from mouse proximal tubular epithelial cells increases susceptibility to renal ischemia-reperfusion injury. Kidney Int. 86:726–737. 2014. View Article : Google Scholar : PubMed/NCBI

15 

Yang LV, Radu CG, Roy M, Lee S, McLaughlin J, Teitell MA, Iruela-Arispe ML and Witte ON: Vascular abnormalities in mice deficient for the G protein-coupled receptor GPR4 that functions as a pH sensor. Mol Cell Biol. 27:1334–1347. 2007. View Article : Google Scholar : PubMed/NCBI

16 

Wang YC, Li WZ, Wu Y, Yin YY, Dong LY, Chen ZW and Wu WN: Acid-sensing ion channel 1a contributes to the effect of extracellular acidosis on NLRP1 inflammasome activation in cortical neurons. J Neuroinflammation. 12:2462015. View Article : Google Scholar : PubMed/NCBI

17 

Cronberg T, Rytter A, Asztély F, Söder A and Wieloch T: Glucose but not lactate in combination with acidosis aggravates ischemic neuronal death in vitro. Stroke. 35:753–757. 2004. View Article : Google Scholar : PubMed/NCBI

18 

Magalhães PA, Magalhães PA, de Brito TS, Freire RS, da Silva MT, dos Santos AA, Vale ML, de Menezes DB, Martins AM and Libório AB: Metabolic acidosis aggravates experimental acute kidney injury. Life Sci. 146:58–65. 2016. View Article : Google Scholar : PubMed/NCBI

19 

Sanderlin E, Justus C, Krewson E and Yang L: Emerging roles for the pH-sensing G protein-coupled receptors in response to acidotic stress. Cell Health Cytoskelet. 2015:99–109. 2015.

20 

Huang Y and McNamara JO: Ischemic stroke: ‘Acidotoxicity’ is a perpetrator. Cell. 118:665–666. 2004. View Article : Google Scholar : PubMed/NCBI

21 

Dong L, Krewson EA and Yang LV: Acidosis activates endoplasmic reticulum stress pathways through GPR4 in human vascular endothelial cells. Int J Mol Sci. 18:pii: E2782017. View Article : Google Scholar

22 

Ding X, Ma M, Teng J, Shao F, Wu E and Wang X: Numb protects human renal tubular epithelial cells from bovine serum albumin-induced apoptosis through antagonizing CHOP/PERK pathway. J Cell Biochem. 117:163–171. 2016. View Article : Google Scholar : PubMed/NCBI

23 

Yang JR, Yao FH, Zhang JG, Ji ZY, Li KL, Zhan J, Tong YN, Lin LR and He YN: Ischemia-reperfusion induces renal tubule pyroptosis via the CHOP-caspase-11 pathway. Am J Physiol Renal Physiol. 306:F75–F84. 2014. View Article : Google Scholar : PubMed/NCBI

24 

Nicol X, Hong KP and Spitzer NC: Spatial and temporal second messenger codes for growth cone turning. Proc Natl Acad Sci USA. 108:13776–13781. 2011. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2018
Volume 16 Issue 5

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Dong B, Zhang X, Fan Y, Cao S and Zhang X: Acidosis promotes cell apoptosis through the G protein‑coupled receptor 4/CCAAT/enhancer‑binding protein homologous protein pathway. Oncol Lett 16: 6735-6741, 2018
APA
Dong, B., Zhang, X., Fan, Y., Cao, S., & Zhang, X. (2018). Acidosis promotes cell apoptosis through the G protein‑coupled receptor 4/CCAAT/enhancer‑binding protein homologous protein pathway. Oncology Letters, 16, 6735-6741. https://doi.org/10.3892/ol.2018.9478
MLA
Dong, B., Zhang, X., Fan, Y., Cao, S., Zhang, X."Acidosis promotes cell apoptosis through the G protein‑coupled receptor 4/CCAAT/enhancer‑binding protein homologous protein pathway". Oncology Letters 16.5 (2018): 6735-6741.
Chicago
Dong, B., Zhang, X., Fan, Y., Cao, S., Zhang, X."Acidosis promotes cell apoptosis through the G protein‑coupled receptor 4/CCAAT/enhancer‑binding protein homologous protein pathway". Oncology Letters 16, no. 5 (2018): 6735-6741. https://doi.org/10.3892/ol.2018.9478