Excessive nNOS/NO/AMPK signaling activation mediated by the blockage of the CBS/H2S system contributes to oxygen‑glucose deprivation‑induced endoplasmic reticulum stress in PC12 cells

  • Authors:
    • Rui Zhang
    • Yong‑Quan Lin
    • Wei‑Sheng Wang
    • Xin‑Qiang Wang
  • View Affiliations

  • Published online on: June 21, 2017     https://doi.org/10.3892/ijmm.2017.3035
  • Pages: 549-557
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Hypoxic‑ischemia stress causes severe brain injury, leading to death and disability worldwide. Although it has been reported that endoplasmic reticulum (ER) stress is an essential step in the progression of hypoxia or ischemia‑induced brain injury, the underlying molecular mechanisms are and have not yet been fully elucidated. Accumulating evidence has indicated that both nitric oxide (NO) and hydrogen sulfide (H2S) play an important role in the development of cerebral ischemic injury. In the present study, we aimed to investigate the effect of the association between NO signaling and the cystathionine β‑synthase (CBS)/H2S system on ER stress in a cell model of cerebral hypoxia‑ischemia injury. We found that oxygen‑glucose deprivation (OGD) markedly increased the NO level and neuronal NO synthase (nNOS) activity. 3‑Bromo‑7‑nitroindazole (3‑Br‑7‑NI), a relatively selective nNOS inhibitor, abolished the OGD‑induced inhibition of cell viability and the increased expression of ER stress‑related proteins, including glucose‑regulated protein 78 (GRP78), C/EBP homologous protein (CHOP) and cleaved caspase‑12 in PC12 cells, indicating the contribution of excessive nNOS/NO signaling to OGD‑induced ER stress. Furthermore, we found that OGD increased the phosphorylated AMP‑activated protein kinase (p‑AMPK)/AMPK ratio, and the AMPK activator, 5‑aminoimidazole‑4‑carboxamide‑1‑β‑D‑ribofuranoside (AICAR), attenuated the effects on OGD‑induced ER stress, suggesting that OGD‑induced NO overproduction results in AMPK activation in PC12 cells. We also found that OGD induced the downregulation of the CBS/H2S system, as indicated by the decreased H2S level in the culture supernatant and CBS activity in PC12 cells. In addition, we found that treatment with NaHS (a H2S donor) or S‑adenosyl‑L‑methionine (SAM, a CBS agonist) mitigated OGD‑induced ER stress, as well as the NO level, nNOS activity and AMPK phosphorylation in PC12 cells. On the whole, these results suggest that the inhibition of the CBS/H2S system, which facilitated excessive nNOS/NO/AMPK activation, contributes to OGD‑induced ER stress.

Introduction

A number of empirical studies have demonstrated that hypoxic and ischemic stress play a role in multiple human central nervous system (CNS) diseases including ischemic stroke, which is becoming one of the leading causes of mortality worldwide (1). Although various pathogenic mechanisms associated with ischemic stroke, such as oxidative stress, apoptosis, inflammation, calcium overload, endoplasmic reticulum (ER) stress and the disruption of the blood-brain barrier (BBB) have been put forward (2,3), the underlying mechanisms of hypoxic/ischemic-induced cerebral injury are unclear and remain to be fully explored. According to recent studies, ER stress is an essential step in neuronal injury resulting from cerebral ischemia and the modulation of ER stress provides a remarkable protective function in the ischemic brain (4,5). Therefore, it is of realistic significance to further investigate the potential mechanisms associated with ER stress under conditions of cerebral ischemia. It is generally known that oxygen-glucose deprivation (OGD) induces neuronal cell injury, and this model is commonly used in research to examine cerebral ischemic injury (6). Thus, in this study, we used PC12 cells, which were induced to differentiate by nerve growth factor, and were then exposed to OGD to establish a cerebral hypoxia-ischemia model, in order to investigate the underlying mechanisms of OGD-induced ER stress.

It is well established that nitric oxide (NO) is a physiological intercellular messenger in the CNS, synthesized by the nitric oxide synthase (NOS)-catalyzed reaction (7,8). There are three isoforms of NOS characterized in brain cells, namely neuronal NOS (nNOS), inducible NOS (iNOS) and endothelial NOS (eNOS) (9). Increasing evidence has confirmed that particularly excessive NO production by nNOS mediates excitotoxicity by promoting a cascade reaction under energy depletion-induced neuronal brain injury (10,11). However, the mechanisms through which elevated levels of NO result in neuronal cell death in ischemic brain injury are still unclear. AMP-activated protein kinase (AMPK) is a serine threonine kinase, considered as a key metabolic and stress sensor/effector (12). AMPK is activated under pathological conditions, incuding nutrient deprivation, vigorous exercise, or heat shock. It has been demonstrated that AMPK plays an essential role in cerebral ischemia; however, its role remains controversial (13,14). It has been shown that AMPK activation is detrimental and the inhibition of AMPK activation is protective under conditions of cerebral ischemia (15). By contrast, others have suggested that the activation of AMPK leads to neuroprotection (16). Notably, increasing evidence indicates that NO is an important activator of AMPK, and the activation of AMPK is muted in mice lacking nNOS under conditions of cerebral ischemia (17,18). However, the association between AMPK and NOS/NO in OGD-induced PC12 cell injury remains unclear.

Hydrogen sulfide (H2S) is a third signaling gaseous mediator followed by CO and NO, predominantly produced from L-cysteine in the CNS by cystathionine β-synthase (CBS), and has a variety of physiological and pathophysiological functions in the CNS (19). Emerging evidence indicates that H2S is considered as not only a neuromodulator, but also a neuroprotectant (20). It has also been shown that in vivo and in vitro models of cerebral ischemia injury, H2S treatment significantly reduces the infarct size and ameliorates neurological function via its antioxidant, anti-apoptotic and anti-inflammatory effects, implying the therapeutic role of H2S in cerebral ischemic stroke (21,22). In addition, H2S reduces ER stress induced by multiple neurotoxins, such as 6-hydroxy-dopamine (6-OHDA) (23) and homocysteine (24), resulting in neuroprotective effects. Thus, we wished to investigate whether the disruption of endogenous H2S generation is involved in OGD-induced ER stress. Furthermore, it has been shown that the complex interaction between CBS/H2S and NO signaling plays an important role in ischemia/reperfusion (I/R)-related brain damage (25). The activity of CBS can be suppressed by NO (26,27), while H2S increases eNOS activation and NO generation (28). Hence, the associatoin between these in cerebral ischemic injury is worthy of research.

In the present study, we observed that OGD induced ER stress accompanied by the upregulation of nNOS/NO/AMPK signaling and the downregulation of the CBS/H2S system in PC12 cells. Furthermore, NaHS or inhibitor of the nNOS pathway attenuated the ER stress induced by OGD. Simultaneously, the promotion of the CBS/H2S system attenuated OGD-induced nNOS/AMPK activation. These results demonstrated that OGD induced ER stress through the activation of nNOS/NO/AMPK signaling as a result of CBS/H2S system blockage.

Materials and methods

Reagents

3-Bromo-7-nitroindazole (3-Br-7-NI) was purchased from Tocris Cookson, Ltd. (Avonmouth, UK). 5-Aminoimid azole-4-carboxamide-1-β-d-ribofuranoside (AICAR) was obtained from Toronto Research Chemicals, Inc. (North York, ON, Canada). NaHS and S-adenosyl-L-methionine (SAM) were purchased from Sigma-Aldrich (St. Louis, MO, USA). Dulbecco's modified Eagle's medium (DMEM) and fetal bovine serum (FBS) were purchased from Gibco (Grand Island, NY, USA). Hoechst 33258, penicillin/streptomycin, RIPA lysis buffer and enhanced chemiluminescence (ECL) reagents were supplied by Beyotime Biotechnology (Shanghai, China). Cell counting kit-8 (CCK-8) was purchased from Dojindo Molecular Technologies (Rockville, MD, USA). The Nitric Oxide Assay kit and Total Nitric Oxide Synthase Assay kit were purchased from Nanjing Jiancheng Bioengineering Institute (Nanjing, China). The primary antibodies against glucose-regulated protein 78 (GRP78), C/EBP homologous protein (CHOP), cleaved caspase-12 and phosphorylated AMP-activated protein kinase (p-AMPK) were supplied by Cell Signaling Technology, Inc. (Beverly, MA, USA). Specific antibody to AMPK was obtained from Santa Cruz Biotechnology, Inc. (San Diego, CA, USA). All reagents were of the purest commercial grade.

Cell culture and model of OGD-induced cell injury and treatment

Highly differentiated rat adrenal pheochromocytoma (PC12) cells, obtained from the American Type Culture Collection (ATCC; Manassas, VA, USA), were maintained in DMEM supplemented with 10% FBS and 1% penicillin/streptomycin at 37°C in a humidified atmosphere with 5% CO2/95% air. To establish the model of OGD-induced cell injury, PC12 cells were exposed to OGD for 12 h by incubation in deoxygenated glucose-free and serum-free DMEM at 37°C in a humidified atmosphere with 5% CO2, 94% N2, and 1% O2 for 12 h. To investigate the effect of nNOS/NO signaling on OGD-induced injury, the PC12 cells were pre-treated with 3-Br-7-NI (a relatively selective nNOS inhibitor, 10 µM) for 30 min and then co-incubated with OGD for 12 h. To investigate the effect of the AMPK pathway on 3-Br-7-NI-caused the inhibition of ODG-induced injury in PC12 cells, PC12 cells ware pretreated with AICAR (20 µM) for 30 min and then incubated with 3-Br-7-NI (10 µM) for 30 min followed by exposure to OGD for 12 h.

Measurement of cell viability

The viability of the PC12 cells was evaluated by CCK-8 assay according to the manufacturer's instructions. In brief, the PC12 cells in the logarithmic phase were seeded into 96-well plate at a density of approximately 1×104 cells/well overnight. At the end of drug treatment, 10 µl of CCK-8 reagent were added to each well, followed by incubation for 4 h at 37°C. The absorbance at 450 nm was measured using a microplate reader (Epoch; BioTek, Winooski, VT, USA). The viability of the cells was expressed as a percentage of that of the control cells. The assays were performed in duplicate for 3 times.

Determination of intracellular NO levels and NOS activity

PC12 cells at the logarithmic growth phase were seeded in 6-well plates a density of approximately 1×106 cells/well overnight. Following the different interventions, total protein was extracted in PBS with Ultrasonic Cell Disruption System (5 sec, 15 times, 4°C) and quantified using the BCA Protein Assay kit. The NO level and total NOS activity were assessed using the Nitric Oxide Assay kit and Total Nitric Oxide Synthase Assay kit following the instructions provided by the respective manufacturers. Data related to the NO level are expressed as pmol/mg protein. Data related to total NOS activity are expressed as U/mg protein. Each independent experiment was repeated at least in triplicate.

Measurement of H2S concentration

The level of H2S in the cell culture supernatant was measured using the methylene blue spectrophotometric method in that H2S and zinc acetate were co-incubated to form zinc sulfide which then dissolved in hydrochloric acid solution supplemented with N,N-dimethyl-p-phenylenediamine sulphate yielding and ferric chloride (FeCl3), resulting in the formation of methylene blue, which was quantified spectrophotometrically. Briefly, the culture supernatant of PC12 cells with different interventions was collected following centrifugation for 5 min at 1,000 rpm. A total of 500 µl of supernatant was combined with 250 µl of zinc acetate (1%), 250 µl of N,N-dimethyl-p-phenylenediamine sulfate (20 mmol/l) and 200 µl of FeCl3 (30 mmol/l) in 500 µl of hydrochloric acid solution (10%). Following incubation for 15 min at room temperature, the absorbance of at 670 nm was measured by spectrophotometry (LAS-3000; Fujifilm, Tokyo, Japan). The H2S concentration was calculated based on the standard curve which was generated by serial dilution of NaHS and was expressed in µmol/l.

Measurement of CBS activity

The PC12 cells subjected to the different treatments were collected and homogenized in potassium phosphate buffer (50 mmol/l, pH 6.8). Following centrifugation at 14,000 × g for 60 min at 4°C, the supernatant was collected for enzyme assays. A mixing system comprising L-cysteine (0.5 mol/l), enzyme protein (0–100 µg), 5-pyridoxal phosphate/potassium phosphate buffer solution (100 mmol/l) and potassium phosphate buffer (100 mM, pH 7.4) was used, transferring the Eppendorf tubes from ice to a shaking water bath at 37°C. Following incubation for 60 min, the reaction was terminated by the addition of w/v zinc acetate (1%) to trap H2S followed by v/v trichloroacetic acid (10%) to precipitate proteins. Subsequently, N,N-dimethyl-p-phenylenediamine-sulfate in HCl (7.2 M) was immediately added to the reaction system followed by addition of FeCl3 in HCl (1.2 M). The absorbance of at 670 nm was measured by spectrophotometry (LAS-3000; Fujifilm) and the H2S content was calculated against a calibration curve of standard NaHS solutions. The CBS activity was expressed as the amount of H2S generated per mg of reaction samples, with a unit of nmol/mg protein.

Measurement of caspase-3 activity

The activity of capsase-3 was detected using a commercial kit (Nanjing Jiancheng Bioengineering Institute) according to the manufacturer's instructions. The absorbance at 405 nm was detected using a microplate reader (Epoch; BioTek). Data were expressed as the fold of the control cells.

Western blot analysis

The PC12 cells were homogenized in RIPA lysis buffer at 4°C for 30 min and the supernatant was collected following centrifugation at 12,000 rpm for 10 min at 4°C. The protein concentration was measured using the BCA Protein Assay kit. Equal amounts of proteins were separated by 10–15% sodium dodecyl sulfate-polyacrylamide gel (SDS-PAGE), and then transferred onto polyvinylidene fluoride (PVDF) membranes (Millipore Corp., Bedford, MA, USA). After blocking for 2 h in TBS with 0.01% Tween-20 (TBST) containing 5% skim milk at room temperature, the membranes were incubated with primary antibodies against CBS (1:2,000; ab96252; Abcam, Cambridge, UK), AMPK (sc-19128; Santa Cruz Biotechnology, Inc., Santa Cruz, CA, USA), p-AMPK (1:2,000; cat. no. #2537), GRP78 (1:2000; cat. no. #3183) and CHOP (1:2,000; cat. no. #2895) (all from Cell Signaling Technology, Inc), cleaved caspase-12 (1:1,000; ab62484; Abcam), and β-actin (1:1,000; cat. no. #4970; Cell Signaling Technology, Inc.) overnight at 4°C, respectively. After washing with TBST for 3 times, the membranes were incubated with the appropriate diluted horseradish peroxidase (HRP)-conjugated secondary antibody for 2 h at room temperature. Subsequently, the membranes were washed again and the protein bands were detected using the ECL system (ZsBio, Beijing, China). The integrated optical density was calculated using ImageJ 1.4 6i software. The amount of protein was represented as a percentage of that of the control cells.

Statistical analysis

Data are expressed as the means ± SEM. The analysis of significant differences groups was performed by one-way analysis of variance (ANOVA) followed by the least significant difference (LSD) test. Differences were considered statistically significant at P<0.05.

Results

OGD induces cytotoxicity and ER stress by promoting nNOS/NO signaling in PC12 cells

As NOS/NO signaling is known to play a role in cerebral ischemic injury (10,29), in this study, we first investigated the alternations of intracellular NO generation in OGD-exposed PC12 cells. We found that the exposure of PC12 cells to OGD for 12 h caused an obvious increase in the level of NO in PC12 cells (Fig. 1A). To investigate the possible role of nNOS activity in mediating OGD-induced NO generation in PC12 cells, we detected the activity of nNOS and found an increase in the activity of nNOS following the exposure of the PC12 cells to OGD for 12 h (Fig. 1B). In addition, we examined the effect of the OGD-induced increase in nNOS/NO signaling on ER stress in PC12 cells. We found that incubation of the PC12 cells with 3-Br-7-NI (10 µM), a relatively selective nNOS inhibitor, for 30 min significantly abrogated the OGD-induced decrease in the viability of the PC12 cells (Fig. 1C). Simultaneously, pre-treatment with 3-Br-7-NI attenuated OGD-induced ER stress, as evidenced by the decreased expression of ER-related proteins, including GRP78 (Fig. 1D), CHOP (Fig. 1E) and cleaved caspase-12 (Fig. 1F) proteins in PC12 cells. Of note, 3-Br-7-NI treatment alone had no effect on cell viability and ER-related protein expression. Taken together, these results suggest that the OGD-mediated NO overproduction via the increased activity of nNOS, is in part, responsible for nerve cytotoxicity and ER stress during ischemia-induced cerebral injury.

nNOS/NO signaling is involved in OGD-induced AMPK activation in PC12 cells

A previous demonstrated the interaction between NO and AMPK activation in stroke (17). Thus, we wished to determine whether this interaction exists in OGD-exposed PC12 cells. We found that exposure to OGD for 12 h led to a marked increase in the expression of phosphorylated AMPK (p-AMPK Thr-172) and in the ratio of p-AMPK/AMPK in the PC12 cells (Fig. 2A), indicating that OGD induced AMPK activation. Subsequently, in order to determine whether the OGD-induced AMPK activation is dependent on the enhancement of nNOS/NO signaling, the PC12 cells were pre-treated with 3-Br-7-NI (a nNOS inhibitor, 10 µM) for 30 min and then co-exposed to OGD for 12 h. We found that 3-Br-7-NI markedly abolished the OGD-induced increase in the phosphorylation levels of AMPK (Fig. 2B) in the PC12 cells. These results indicated that the OGD-induced activation of AMPK was dependent on nNOS/NO signaling; this in turn, may potentially promote neuronal injury during cerebral ischemia and anoxia.

Activation of AMPK signaling attenuates the inhibitory effects of 3-Br-7-NI on OGD-induced excessive ER stress in PC12 cells

To further demonstrate whether AMPK activation is involved in OGD-induced ER stress mediated by the nNOS/NO system, the effect of AICAR, an AMPK activator, on ER stress was investigated. As shown in Fig. 3A, pre-treatment with AICAR (20 µM) attenuated the promoting effects of 3-Br-7-NI on the viability of the OGD-exposed PC12 cells, indicating that OGD induced cytotoxicity via NOS-activated AMPK. In addition, we found that pre-treatment with AICAR markedly abolished the inhibitory effects of 3-Br-7-NI on OGD-induced ER stress, as evidenced by the upregulated expression of GRP-78 (Fig. 3B), CHOP (Fig. 3C) and cleaved caspase-12 (Fig. 3D). These results indicated that the OGD-induced increase in nNOS/NO/AMPK signaling contributed to OGD-induced ER stress.

Blockage of the CBS/H2S system contributes to OGD-induced ER stress in PC12 cells

We then investigated the role of the CBS/H2S system in OGD-treated PC12 cells. As shown in Fig. 4, we found that exposure of the PC12 cells to OGD for 12 h markedly reduced the level of H2S in the culture supernatant (Fig. 4A). In addition, the activity of CBS was also attenuated by exposure to OGD (Fig. 4B). These results suggest that exposure to OGD results in the downregulation of the CBS/H2S system. In order to further confirm the role of the CBS/H2S system in OGD-induced neuronal injury, NaHS (a donor of H2S) and SAM (a CBS agonist) were used. We found that pre-treatment with NaHS (200 µM) for 30 min and SAM (100 µM) for 1 h markedly attenuated the OGD-induced decrease in cell viability, while NaHS or SAM treatment alone had no effect on the viability of PC12 cells (Fig. 4C). At the same time, both NaHS and SAM abolished the OGD-induced increase in the expression of ER stress-related marker proteins, including GRP78 (Fig. 4D), CHOP (Fig. 4E) and cleaved caspase-12 (Fig. 4F) in PC12 cells. These results indicate that OGD causes cytotoxicity and ER stress via the inhibition of the CBS/H2S system.

Enhancement of CBS/H2S by NaHS and SAM mitigates the OGD-induced activation of nNOS/NO and AMPK in PC12 cells

Finally, we further investigated the association of CBS/H2S, nNOS/NO and AMPK in the PC12 cells exposed to OGD. We found that the increased levels of H2S induced by NaHS (200 µM) and the enhanced activity of CBS induced by SAM (100 µM) distinctly reversed the OGD-induced NO overproduction, as evidenced by a decrease in the levels of NO (Fig. 5A) and in the activity of nNOS (Fig. 5B), indicating that the inhibition of the CBS/H2S system mediates the OGD-induced upregulation of the nNOS/NO system. Simultaneously, NaHS or SAM treatment also reduced the ratio of p-AMPK (Thr-172)/AMPK in the PC12 cells exposed to OGD (Fig. 5C), suggesting that OGD induced AMPK activation by suppressing the CBS/H2S system.

Discussion

In the present study, we explored the roles of nNOS/NO/AMPK signaling and the CBS/H2S system in OGD-induced ER stress in PC12 cells. In this study, to the best of our knowledge, we demonstrate for the first time that OGD causes ER stress through the activation of the nNOS/NO/AMPK pathway resulting from the inhibition of the CBS/H2S system.

According to the World Health Organization, approximately 15 million individuals each year suffer from cerebral ischemic injury, such as stroke (30). Despite this, the understanding of the mechanisms underlying cerebral ischemic injury remains extremely limited. Accumulating evidence has indicated that ER stress plays an important role in the progression of brain neuronal injury resulting from I/R (3133). Certain stimuli, such as ischemia and hypoxia may trigger the accumulation of unfolded proteins in the ER, leading to excessive or aberrant ER stress and accelerated nerve damage (34). These findings suggest that the elucidation of mechamisms responsible for ER stress signaling may provide a novel target for effective therapeutic approaches for cerebral ischemia.

NO is a physiological mediator generated from L-arginine and oxygen by various forms of NOS, including eNOS, nNOS and iNOS in the brain (35). Increasing evidence reveals a wide range of roles for NOS/NO signaling involved in the occurrence and development of ischemic brain or brain ischemic injury (36). However, NO acts in a protective or deleterious manner depended upon the NOS isoform (37). It has been reported that hypoxic-ischemic injury is attenuated in mice deficient in nNOS, but is exacerbated in eNOS-deficient mice (38,39), indicating that NO overproduction released from nNOS contributes to brain damage. Other studies have further demonstrated that NO is a mediator of neuronal damage following cerebral ischemia (40,41) and the inhibition of NOS decreases BBB disruption, leading to neuroprotective effects under conditions of I/R injury during acute hypertension in rats (42). Furthermore, Nadjafi et al found that NO production was increased during OGD/reperfusion in OLN-93 oligodendrocytes (43). Consistent with these studies, our study demonstrated that exposure to OGD markedly increased the level of NO and the activity of nNOS in PC12 cells. Notably, an experimental study detected that 3-Br-7-NI, a potent and selective nNOS inhibitor, attenuated brain ischemic injury in diabetic stroke via the inhibition of the ER stress pathway (44), implying the mediation of NOS/NO signaling in ER stress under conditions of cerebral ischemia. Similar to this, the current study demonstrated that 3-Br-7-NI pre-treatment also mitigated OGD-induced ER stress, as evidenced by the down-regulated expression of ER-related proteins, such as GRP78, CHOP and cleaved caspase-12 in PC12 cells. Thus, these results suggest that OGD induces ER stress through the enhancement of nNOS/NO signaling.

AMPK has been reported to be present in most mammalian tissues, including the brain (45). It is increasingly becoming recognized that alternations in AMPK activation are not only related to metabolic needs, but are also related to sensing and responding to ʻvarious cell stressʼ, such as ischemia, hypoxia and energy depletion (46). However, little is known regarding the physiological and pathological functions of AMPK and the mechanisms through which AMPK activation occurs in the brain during ischemia. Of note, some studies have shown that cerebral ischemia increases the activation of AMPK in the brain in an NO-dependent manner, which is muted in mice lacking nNOS (17,18), indicating the important role of NOS/NO signaling in AMPK activation. Nevertheless, the interactions between AMPK, NO and NOS warrant further investigation, particularly under the condition of cerebral ischemic injury. Our finds indicated that exposure to OGD increased the phosphorylation of AMPK in PC12 cells, while this effect was abolished by pre-treatment with 3-Br-7-NI, indicating that the OGD-induced AMPK activation is dependent on nNOS/NO signaling. In addition, we found that the AMPK activator, AICAR, attenuated the protective effects of 3-Br-7-NI against OGD-induced cytotoxicity and ER stress in PC12 cells. These results suggest that OGD causes ER stress by increasing the activity of AMPK, as a result of nNOS/NO signaling promotion.

Emerging evidence indicates that H2S plays a broad range of roles in cerebral ischemic injury (20,47). In in vivo model of cerebral I/R injury, H2S pre-conditioning improved neurological function and decreased the infarct size, implying that H2S plays a therapeutic role in cerebral ischemic stroke (21). In addition, a number of studies have confirmed that H2S attenuates ER stress induced by multiple stresses and neurotoxins, including chronic unpredictable mild stress, homocysteine and 6-OHDA (23,24,48), exerting neuroprotective effects. However, the association of H2S and ER stress in cerebral ischemic injury is not yet understood. Thus, we hypothesized that the disruption of H2S may be also involved in ER stress in PC12 cells exposed to OGD. In the current study, we found that exposure to OGD markedly reduced the level of H2S in the culture supernatant, as well as the activity and expression of CBS, indicating that the CBS/H2S system was inhibited by OGD. These findings are consistent with those of the study by Shen et al, who demonstrated that in acute ischemic conditions, CBS is upregulated and activated followed by causing an increased production of H2S (49). In addition, we found that NaHS abolished the OGD-induced decrease in the viability of the PC12 cells and the increase in ER stress, as evidenced by the increased expression of GRP78, CHOP and cleaved caspase-12 in the PC12 cells following treatment with NaHS. These results indicated that blocking the CBS/H2S system contributes to OGD-induced ER stress.

An increasing number of studies have revealed the complex association between H2S and NO, which has an indispensable effect in multiple diseases (50,51). In another study, Grossi proved that H2S is regarded as a co-factor responsible for the generation of NO (52). In addition, another finding is that H2S directly inhibits eNOS, as well as nNOS, contributing to the dual modulation of vascular tension (53). In the present study, we further investigated the association between the CBS/H2S system and nNOS/NO signaling in OGD-exposed PC12 cells. We found that pre-treatment with NaHS markedly attenuated OGD-induced NO overproduction by the excessive activation of nNOS and AMPK activation in PC12 cells. These results indicated that OGD induced the activation of nNOS/NO/AMPK signaling activation through the inhibition of CBS/H2S, leading to ER stress.

In conclusion, our findings demonstrate that exposure to OGD induces ER stress, and the activation of the nNOS/NO/AMPK pathway results from the attenuation of the CBS/H2S system. Our findings provide a better understanding of the signal transduction mechanisms involved in the pathophysiological process of cerebral ischemia-induced ER stress, which provides novel targets and guidance for the development of neuroprotective agents.

References

1 

Adeoye O, Hornung R, Khatri P and Kleindorfer D: Recombinant tissue-type plasminogen activator use for ischemic stroke in the United States: A doubling of treatment rates over the course of 5 years. Stroke. 42:1952–1955. 2011. View Article : Google Scholar : PubMed/NCBI

2 

Manzanero S, Santro T and Arumugam TV: Neuronal oxidative stress in acute ischemic stroke: Sources and contribution to cell injury. Neurochem Int. 62:712–718. 2013. View Article : Google Scholar

3 

Sierra C, Coca A and Schiffrin EL: Vascular mechanisms in the pathogenesis of stroke. Curr Hypertens Rep. 13:200–207. 2011. View Article : Google Scholar : PubMed/NCBI

4 

Xin Q, Ji B, Cheng B, Wang C, Liu H, Chen X, Chen J and Bai B: Endoplasmic reticulum stress in cerebral ischemia. Neurochem Int. 68:18–27. 2014. View Article : Google Scholar : PubMed/NCBI

5 

DeGracia DJ and Montie HL: Cerebral ischemia and the unfolded protein response. J Neurochem. 91:1–8. 2004. View Article : Google Scholar : PubMed/NCBI

6 

Wang CH, Lee WJ, Ghanta VK, Wang WT, Cheng SY and Hsueh CM: Molecules involve in the self-protection of neurons against glucose-oxygen-serum deprivation (GOSD)-induced cell damage. Brain Res Bull. 79:169–176. 2009. View Article : Google Scholar : PubMed/NCBI

7 

Vincent SR: Nitric oxide: A radical neurotransmitter in the central nervous system. Prog Neurobiol. 42:129–160. 1994. View Article : Google Scholar : PubMed/NCBI

8 

Garthwaite J, Charles SL and Chess-Williams R: Endothelium-derived relaxing factor release on activation of NMDA receptors suggests role as intercellular messenger in the brain. Nature. 336:385–388. 1988. View Article : Google Scholar : PubMed/NCBI

9 

Qi SH, Hao LY, Yue J, Zong YY and Zhang GY: Exogenous nitric oxide negatively regulates the S-nitrosylation p38 mitogen-activated protein kinase activation during cerebral ischaemia and reperfusion. Neuropathol Appl Neurobiol. 39:284–297. 2013. View Article : Google Scholar

10 

Liu H, Li J, Zhao F, Wang H, Qu Y and Mu D: Nitric oxide synthase in hypoxic or ischemic brain injury. Rev Neurosci. 26:105–117. 2015. View Article : Google Scholar : PubMed/NCBI

11 

Brown GC: Nitric oxide and neuronal death. Nitric Oxide. 23:153–165. 2010. View Article : Google Scholar : PubMed/NCBI

12 

Tzatsos A and Tsichlis PN: Energy depletion inhibits phosphatidylinositol 3-kinase/Akt signaling and induces apoptosis via AMP-activated protein kinase-dependent phosphorylation of IRS-1 at Ser-794. J Biol Chem. 282:18069–18082. 2007. View Article : Google Scholar : PubMed/NCBI

13 

Li J and McCullough LD: Effects of AMP-activated protein kinase in cerebral ischemia. J Cereb Blood Flow Metab. 30:480–492. 2010. View Article : Google Scholar :

14 

Hardie DG and Frenguelli BG: A neural protection racket: AMPK and the GABA(B) receptor. Neuron. 53:159–162. 2007. View Article : Google Scholar : PubMed/NCBI

15 

Culmsee C, Monnig J, Kemp BE and Mattson MP: AMP-activated protein kinase is highly expressed in neurons in the developing rat brain and promotes neuronal survival following glucose deprivation. J Mol Neurosci. 17:45–58. 2001. View Article : Google Scholar : PubMed/NCBI

16 

Kuramoto N, Wilkins ME, Fairfax BP, Revilla-Sanchez R, Terunuma M, Tamaki K, Iemata M, Warren N, Couve A, Calver A, et al: Phospho-dependent functional modulation of GABA(B) receptors by the metabolic sensor AMP-dependent protein kinase. Neuron. 53:233–247. 2007. View Article : Google Scholar : PubMed/NCBI

17 

McCullough LD, Zeng Z, Li H, Landree LE, McFadden J and Ronnett GV: Pharmacological inhibition of AMP-activated protein kinase provides neuroprotection in stroke. J Biol Chem. 280:20493–20502. 2005. View Article : Google Scholar : PubMed/NCBI

18 

Heales SJ, Bolaños JP, Stewart VC, Brookes PS, Land JM and Clark JB: Nitric oxide, mitochondria and neurological disease. Biochim Biophys Acta. 1410:215–228. 1999. View Article : Google Scholar : PubMed/NCBI

19 

Stein A and Bailey SM: Redox biology of hydrogen sulfide: Implications for physiology, pathophysiology, and pharmacology. Redox Biol. 1:32–39. 2013. View Article : Google Scholar : PubMed/NCBI

20 

Zhang X and Bian JS: Hydrogen sulfide: A neuromodulator and neuroprotectant in the central nervous system. ACS Chem Neurosci. 5:876–883. 2014. View Article : Google Scholar : PubMed/NCBI

21 

Gheibi S, Aboutaleb N, Khaksari M, Kalalian-Moghaddam H, Vakili A, Asadi Y, Mehrjerdi FZ and Gheibi A: Hydrogen sulfide protects the brain against ischemic reperfusion injury in a transient model of focal cerebral ischemia. J Mol Neurosci. 54:264–270. 2014. View Article : Google Scholar : PubMed/NCBI

22 

Yin J, Tu C, Zhao J, Ou D, Chen G, Liu Y and Xiao X: Exogenous hydrogen sulfide protects against global cerebral ischemia/reperfusion injury via its anti-oxidative, anti-inflammatory and anti-apoptotic effects in rats. Brain Res. 1491:188–196. 2013. View Article : Google Scholar

23 

Xie L, Tiong CX and Bian JS: Hydrogen sulfide protects SH-SY5Y cells against 6-hydroxydopamine-induced endoplasmic reticulum stress. Am J Physiol Cell Physiol. 303:C81–C91. 2012. View Article : Google Scholar : PubMed/NCBI

24 

Wei HJ, Xu JH, Li MH, Tang JP, Zou W, Zhang P, Wang L, Wang CY and Tang XQ: Hydrogen sulfide inhibits homocysteine-induced endoplasmic reticulum stress and neuronal apoptosis in rat hippocampus via upregulation of the BDNF-TrkB pathway. Acta Pharmacol Sin. 35:707–715. 2014. View Article : Google Scholar : PubMed/NCBI

25 

Yin W, Lan L, Huang Z, Ji J, Fang J, Wang X, Ji H, Peng S, Xu J and Zhang Y: Discovery of a ring-opened derivative of 3-n-butylphthalide bearing NO/H2S-donating moieties as a potential anti-ischemic stroke agent. Eur J Med Chem. 115:369–380. 2016. View Article : Google Scholar : PubMed/NCBI

26 

Morikawa T, Kajimura M, Nakamura T, Hishiki T, Nakanishi T, Yukutake Y, Nagahata Y, Ishikawa M, Hattori K, Takenouchi T, et al: Hypoxic regulation of the cerebral microcirculation is mediated by a carbon monoxide-sensitive hydrogen sulfide pathway. Proc Natl Acad Sci USA. 109:1293–1298. 2012. View Article : Google Scholar : PubMed/NCBI

27 

Taoka S and Banerjee R: Characterization of NO binding to human cystathionine beta-synthase: Possible implications of the effects of CO and NO binding to the human enzyme. J Inorg Biochem. 87:245–251. 2001. View Article : Google Scholar : PubMed/NCBI

28 

Zhao W, Zhang J, Lu Y and Wang R: The vasorelaxant effect of H(2)S as a novel endogenous gaseous K(ATP) channel opener. EMBO J. 20:6008–6016. 2001. View Article : Google Scholar : PubMed/NCBI

29 

Liu HT and Mu DZ: Inducible nitric oxide synthase and brain hypoxic-ischemic brain damage. Zhongguo Dang dai er ke za zhi. 16:962–967. 2014.In Chinese. PubMed/NCBI

30 

Moskowitz MA, Lo EH and Iadecola C: The science of stroke: Mechanisms in search of treatments. Neuron. 67:181–198. 2010. View Article : Google Scholar : PubMed/NCBI

31 

Bai X, Liu S, Yuan L, Xie Y, Li T, Wang L, Wang X, Zhang T, Qin S, Song G, et al: Hydrogen-rich saline mediates neuroprotection through the regulation of endoplasmic reticulum stress and autophagy under hypoxia-ischemia neonatal brain injury in mice. Brain Res. 1646:410–417. 2016. View Article : Google Scholar : PubMed/NCBI

32 

Cao G, Zhou H, Jiang N, Han Y, Hu Y, Zhang Y, Qi J, Kou J and Yu B: YiQiFuMai powder injection ameliorates cerebral ischemia by inhibiting endoplasmic reticulum stress-mediated neuronal apoptosis. Oxid Med Cell Longev. 2016:54932792016. View Article : Google Scholar : PubMed/NCBI

33 

Osada N, Kosuge Y, Ishige K and Ito Y: Characterization of neuronal and astroglial responses to ER stress in the hippocampal CA1 area in mice following transient forebrain ischemia. Neurochem Int. 57:1–7. 2010. View Article : Google Scholar : PubMed/NCBI

34 

Herrmann AG, Deighton RF, Le Bihan T, McCulloch MC, Searcy JL, Kerr LE and McCulloch J: Adaptive changes in the neuronal proteome: Mitochondrial energy production, endoplasmic reticulum stress, and ribosomal dysfunction in the cellular response to metabolic stress. J Cereb Blood Flow Metab. 33:673–683. 2013. View Article : Google Scholar : PubMed/NCBI

35 

Edelman GM and Gally JA: Nitric oxide: Linking space and time in the brain. Proc Natl Acad Sci USA. 89:11651–11652. 1992. View Article : Google Scholar : PubMed/NCBI

36 

Lu Q, Harris VA, Rafikov R, Sun X, Kumar S and Black SM: Nitric oxide induces hypoxia ischemic injury in the neonatal brain via the disruption of neuronal iron metabolism. Redox Biol. 6:112–121. 2015. View Article : Google Scholar : PubMed/NCBI

37 

Pei DS, Song YJ, Yu HM, Hu WW, Du Y and Zhang GY: Exogenous nitric oxide negatively regulates c-Jun N-terminal kinase activation via inhibiting endogenous NO-induced S-nitrosylation during cerebral ischemia and reperfusion in rat hippocampus. J Neurochem. 106:1952–1963. 2008. View Article : Google Scholar : PubMed/NCBI

38 

Huang Z, Huang PL, Ma J, Meng W, Ayata C, Fishman MC and Moskowitz MA: Enlarged infarcts in endothelial nitric oxide synthase knockout mice are attenuated by nitro-L-arginine. J Cereb Blood Flow Metab. 16:981–987. 1996. View Article : Google Scholar : PubMed/NCBI

39 

Huang Z, Huang PL, Panahian N, Dalkara T, Fishman MC and Moskowitz MA: Effects of cerebral ischemia in mice deficient in neuronal nitric oxide synthase. Science. 265:1883–1885. 1994. View Article : Google Scholar : PubMed/NCBI

40 

Ferriero DM, Holtzman DM, Black SM and Sheldon RA: Neonatal mice lacking neuronal nitric oxide synthase are less vulnerable to hypoxic-ischemic injury. Neurobiol Dis. 3:64–71. 1996. View Article : Google Scholar : PubMed/NCBI

41 

Nowicki JP, Duval D, Poignet H and Scatton B: Nitric oxide mediates neuronal death after focal cerebral ischemia in the mouse. Eur J Pharmacol. 204:339–340. 1991. View Article : Google Scholar : PubMed/NCBI

42 

Mohammadi MT, Shid Moosavi SM and Dehghani GA: Contribution of nitric oxide synthase (NOS) activity in blood-brain barrier disruption and edema after acute ischemia/reperfusion in aortic coarctation-induced hypertensive rats. Iran Biomed J. 15:22–30. 2011.PubMed/NCBI

43 

Nadjafi S, Ebrahimi SA and Rahbar-Roshandel N: Effect of berberine on nitric oxide production during oxygen-glucose deprivation/reperfusion in OLN-93 oligodendrocytes. Pak J Biol Sci. 17:1185–1189. 2014. View Article : Google Scholar

44 

Srinivasan K and Sharma SS: 3-Bromo-7-nitroindazole attenuates brain ischemic injury in diabetic stroke via inhibition of endoplasmic reticulum stress pathway involving CHOP. Life Sci. 90:154–160. 2012. View Article : Google Scholar

45 

Gao G, Widmer J, Stapleton D, Teh T, Cox T, Kemp BE and Witters LA: Catalytic subunits of the porcine and rat 5′-AMP-activated protein kinase are members of the SNF1 protein kinase family. Biochim Biophys Acta. 1266:73–82. 1995. View Article : Google Scholar : PubMed/NCBI

46 

Ramamurthy S and Ronnett GV: Developing a head for energy sensing: AMP-activated protein kinase as a multifunctional metabolic sensor in the brain. J Physiol. 574:85–93. 2006. View Article : Google Scholar : PubMed/NCBI

47 

Ji K, Xue L, Cheng J and Bai Y: Preconditioning of H2S inha-lation protects against cerebral ischemia/reperfusion injury by induction of HSP70 through PI3K/Akt/Nrf2 pathway. Brain Res Bull. 121:68–74. 2016. View Article : Google Scholar : PubMed/NCBI

48 

Tan H, Zou W, Jiang J, Tian Y, Xiao Z, Bi L, Zeng H and Tang X: Disturbance of hippocampal H2S generation contributes to CUMS-induced depression-like behavior: Involvement in endoplasmic reticulum stress of hippocampus. Acta Biochim Biophys Sin (Shanghai). 47:285–291. 2015. View Article : Google Scholar

49 

Shen Y, Shen Z, Miao L, Xin X, Lin S, Zhu Y, Guo W and Zhu YZ: miRNA-30 family inhibition protects against cardiac ischemic injury by regulating cystathionine-gamma-lyase expression. Antioxid Redox Signal. 22:224–240. 2015. View Article : Google Scholar :

50 

Chatzianastasiou A, Bibli SI, Andreadou I, Efentakis P, Kaludercic N, Wood ME, Whiteman M, Di Lisa F, Daiber A, Manolopoulos VG, et al: Cardioprotection by H2S donors: Nitric oxide-dependent and -independent mechanisms. J Pharmacol Exp Ther. 358:431–440. 2016. View Article : Google Scholar : PubMed/NCBI

51 

Duan XC, Liu SY, Guo R, Xiao L, Xue HM, Guo Q, Jin S and Wu YM: Cystathionine-β-synthase gene transfer into rostral ventrolateral medulla exacerbates hypertension via nitric oxide in spontaneously hypertensive rats. Am J Hypertens. 28:1106–1113. 2015. View Article : Google Scholar : PubMed/NCBI

52 

Grossi L: Hydrogen sulfide induces nitric oxide release from nitrite. Bioorg Med Chem Lett. 19:6092–6094. 2009. View Article : Google Scholar : PubMed/NCBI

53 

Kubo S, Doe I, Kurokawa Y, Nishikawa H and Kawabata A: Direct inhibition of endothelial nitric oxide synthase by hydrogen sulfide: Contribution to dual modulation of vascular tension. Toxicology. 232:138–146. 2007. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2017
Volume 40 Issue 2

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
Zhang R, Lin YQ, Wang WS and Wang XQ: Excessive nNOS/NO/AMPK signaling activation mediated by the blockage of the CBS/H2S system contributes to oxygen‑glucose deprivation‑induced endoplasmic reticulum stress in PC12 cells. Int J Mol Med 40: 549-557, 2017
APA
Zhang, R., Lin, Y., Wang, W., & Wang, X. (2017). Excessive nNOS/NO/AMPK signaling activation mediated by the blockage of the CBS/H2S system contributes to oxygen‑glucose deprivation‑induced endoplasmic reticulum stress in PC12 cells. International Journal of Molecular Medicine, 40, 549-557. https://doi.org/10.3892/ijmm.2017.3035
MLA
Zhang, R., Lin, Y., Wang, W., Wang, X."Excessive nNOS/NO/AMPK signaling activation mediated by the blockage of the CBS/H2S system contributes to oxygen‑glucose deprivation‑induced endoplasmic reticulum stress in PC12 cells". International Journal of Molecular Medicine 40.2 (2017): 549-557.
Chicago
Zhang, R., Lin, Y., Wang, W., Wang, X."Excessive nNOS/NO/AMPK signaling activation mediated by the blockage of the CBS/H2S system contributes to oxygen‑glucose deprivation‑induced endoplasmic reticulum stress in PC12 cells". International Journal of Molecular Medicine 40, no. 2 (2017): 549-557. https://doi.org/10.3892/ijmm.2017.3035