Mitochondrial separation protein inhibitor inhibits cell apoptosis in rat lungs during intermittent hypoxia

  • Authors:
    • Dan Zhao
    • Chen‑Yi Yin
    • Xian‑Wei Ye
    • Zi‑Fen Wan
    • De‑Gang Zhao
    • Xiang‑Yan Zhang
  • View Affiliations

  • Published online on: January 25, 2019     https://doi.org/10.3892/etm.2019.7201
  • Pages: 2349-2358
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Obstructive sleep apnoea (OSA) is a very common sleep and breathing disorder that occurs in worldwide. It is important to develop a more effective treatment for OSA to overcome lung cell apoptosis during intermittent hypoxia (IH). A mitochondrial separation protein inhibitor (Mdivi‑1) has been demonstrated to be a powerful tool for inhibiting apoptosis. In the present study, the protective effect and possible mechanism of apoptosis in lung cells during IH was investigated using in vivo and in vitro experiments. Following IH exposure for 4 weeks, the lung tissues of Sprague Dawley rats exhibited interstitial lesions, while Mdivi‑1 reduced these pulmonary interstitial lesions. B‑cell lymphoma (Bcl)‑2 mRNA and protein expression levels were decreased however caspase‑3, caspase‑9 and dynamin‑related protein 1 (Drp‑1) mRNA and protein expression levels were increased. Following Mdivi‑1 intervention, Bcl‑2 mRNA and protein expression levels were increased while caspase‑3, caspase‑9 and Drp‑1 mRNA and protein expression levels were decreased (P<0.05). After exposure to IH for 12 h, the apoptosis rate of WTRL1 cells in rats increased gradually with the IH time (P<0.05). Bcl‑2 mRNA and protein expression levels were decreased, whereas caspase‑3, caspase‑9, cytochrome C (Cyt‑C) and Drp‑1 mRNA levels were increased, and caspase‑3, caspase‑9 and Drp‑1 protein expression levels were increased. After Mdivi‑1 intervention, Bcl‑2 mRNA and protein expression levels were increased but caspase‑3, caspase‑9, Cyt‑C and Drp‑1 mRNA levels were decreased along with caspase‑9, Cyt‑C and Drp‑1 protein expression levels which were decreased (P<0.05). The results of the present study suggest that Mdivi‑1 may be a potential agent for treating OSA because it inhibits the mitochondrial pathway and reduces apoptosis.

Introduction

Obstructive sleep apnoea (OSA) is a very common sleep and breathing disorder that is characterized by the repetitive collapse of the upper airway during sleep (1); in addition, OSA has high morbidity in paediatric and adult populations. In OSA, upper airway obstruction is usually associated with recurrent hypoxemia interspersed with periods of reoxygenation and is typically terminated by brief arousals, resulting in intermittent hypoxia (IH) and sleep fragmentation (2). The oscillatory nature of IH, especially chronic IH, may mimic the paradigm of ischaemia-reperfusion in that tissues and cells are exposed to periods of low and high O2, which may lead to oxidative stress and induces lipid peroxidation, apoptosis, stress-responsive proteins, cell death and inflammation (35). Studies have confirmed that IH via different signalling pathways induces cell apoptosis (6,7). Mitochondrial separation protein inhibitor (Mdivi-1) is a selective dynamin-related protein 1 (Drp-1) inhibitor that has been reported to be a potential therapeutic target for brain, cardiovascular, liver and tumour angiogenesis (8,9). How Mdivi-1 inhibits lung apoptosis during IH remains unclear. The aim of the present study was to investigate the protective effect and possible mechanism of Mdivi-1 in lung cell apoptosis during IH.

Materials and methods

Animals

A total of 32 specific pathogen free 6–8-week-old male Sprague Dawley (SD) rats (body weight, 180–220 g) were purchased from the Animal Care and Use Committee of the Army Medical University (Chongqing, China). The experiments were conducted in accordance with the Guide for the Care and Use of Laboratory Animals (10). All experimental procedures were approved by the Ethics Committee of Guizhou Provincial People's Hospital (Guiyang, China). All efforts were made to minimize animal suffering and to reduce the number of animals used.

In vivo experimental design

All animals were maintained in a controlled environment (22–25°C, 40–60% relative humidity, with a 12-h light-dark cycle with the lights on at 08:00 am) with food and water available. Animals were allowed to acclimatize for 1 week to the experimental chamber. The rats were allocated randomly to four groups: Control group (n=8), IH group (n=8), IH+25 mg/kg Mdivi-1 group (n=8) and IH+50 mg/kg Mdivi-1 group (n=8). The rats in the IH group, IH+25 mg/kg Mdivi-1 group and IH+50 mg/kg Mdivi-1 group were exposed to IH conditions (21% O2, 5% CO2 and balanced N2) for 90 sec and hypoxia (8% O2, 5% CO2 and balanced N2 for 30 sec) from 08:00 am to 04:00 pm daily for 4 consecutive weeks (1113). The chambers had a rear N2 inlet and a front air extractor, which enabled recovery to normoxia. A computerized system controlled the valve inlets and the alternating cycles of the extractors. The CO2 in the chamber was maintained at a low level by continuous air extraction (14). Rats in the control group were maintained under normoxic conditions during the experiment. The treatment groups were injected intraperitoneally with Mdivi-1 (MedChemExpress, Monmouth Junction, NJ, USA) prior to oxygen treatment every day (15). A general clinical pain score for scoring cyanosis condition for decisions about endpoint was use as presented in Table I (16).

Table I.

Assessing pain and distress in animal.

Table I.

Assessing pain and distress in animal.

1) No indication of pain and distressNormal; well groomed; alert; active; good condition; asleep or calm; normal appetite; BCS=3, 4 or 5
2) Mild or anticipated pain and distressNot well groomed; awkward gait; slightly hunched; looks at wound or pulls away when area touched; mildly agitated; BCS=2
3) Moderate pain and distressRough hair coat; dirty incision; squinted eyes; moves slowly; walks hunched and/or slowly; depressed or moderately agitated; slight dehydration; pruritic; restless; uncomfortable; not eating or drinking; BCS=2.
4) Severe pain and distressVery rough hair coat; eyes sunken (severe dehydration); slow to move or non-responsive when coaxed; hunched; large abdominal mass; dyspnea; self mutilating; violent reaction to stimuli or when approached; BCS=1

[i] BCS, body condition score.

Sample collection and tissue preparation

Trained personnel euthanized the experimental animals via CO2 inhalation following IH exposure. Rats were left in the chamber until clinical death could be ensured. The flow rate of CO2 displaced 20% of the volume chamber per minute. If no breath was seen for 60 sec the rodent was removed from the chamber. To ensure death, the rat's chest was felt to determine that there is no longer a heartbeat. Death was verified following euthanasia and prior to disposal. The lungs were dissected out and the right lower lobes were fixed in 4% paraformaldehyde at room temperature for 48 h. The remaining lung tissues were frozen in liquid nitrogen and then stored at −80°C until further analysis.

Histological examination

The right lower lobes were fixed in 4% paraformaldehyde at room temperature for 24 h, embedded in paraffin blocks and slices into 6-µm-thick sections. The sections were then stained with haematoxylin for 5 min and eosin for 90 sec at room temperature, and observed by light microscopy. Damage to the lung tissue was scored by the pathologist on a scale of 1 (no injury) to 4 (worst), as described previously (17).

In vitro experimental design

WTRL1 cells (a rat pulmonary alveolar epithelial cell line; http://www.cellresource.cn/fdetail.aspx?id=1349) were purchased from the National Infrastructure of Cell Line Resource, (Beijing, China). The cells were grown in RPMI-1640 medium (HyClone, Logan, UT, USA) with 10% FBS (Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA), penicillin (100 units/ml) and streptomycin (100 µg/ml) and were cultured at 37°C in 5% CO2 in a humidified incubator. WTRL1 cells in the logarithmic growth phase were divided randomly into five groups (3×106 cells/well) in 6-well plates: Control, IH, IH+10 µM Mdivi-1, IH+50 µM Mdivi-1 and IH+100 µM Mdivi-1. The control group was exposed to air, while the IH group, IH+10 µM Mdivi-1 group, IH+50 µM Mdivi-1 group and IH+100 µM Mdivi-1 group were placed in a low oxygen culture box containing 5% O2 for 1 h. The box was then reoxygenated with 21% O2 for 30 min and this process was repeated for 12 h. The treatment groups were incubated with Mdivi-1 for 30 min prior to IH culture.

Flow cytometry

WTRL1 cells were grown logarithmically and placed on 60-mm Petri dishes in a low oxygen culture box containing 5% O2 for 1 h. The box was reoxygenated with 21% O2 for 30 min and this process was repeated for 6, 12, 24 and 48 h. Next, the cells were resuspended in Annexin V-PI from a dual-staining assay kit (Nanjing KeyGen Biotech. Co. Ltd., Nanjing, China) for 10 min at room temperature. The fluorescence signals from Annexin V and PI were measured by flow cytometry with a FACSCalibur flow cytometer (BD Bioscience), and the data were analysed using Cell Quest Pro software (version 5.0; BD Bioscience). The flow cytometry results were measured in 1 h.

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

Total RNA was extracted from lung tissues or cells with TRIzol (Thermo Fisher Scientific, Inc.). The D260/D280 ratio was measured and samples with a ratio between 1.8 and 2.0 were used for reverse transcription with a High Capacity cDNA Archive kit (Bio-Rad Laboratories, Inc., Hercules, CA, USA). The reverse transcription protocol were as follows: 25°C for 10 min, 37°C for 120 min and 85°C for 5 min. qPCR was applied to analyze the cDNAs by using the 2X Maxima SYBR-Green/ROX qPCR Master Mix kit (Thermo Fisher Scientific, Inc.) following the manufacturer's protocol. The amplification conditions were as follows: 95°C for 10 min for denaturation, followed by 40 cycles of 95°C for 15 sec, 60°C for 30 sec and 72°C for 60 sec. qPCR for Bcl-2, caspase-3, caspase-9, Cyt-C and Drp-1 was performed using TaqMan gene expression assays and the 2−ΔΔCq method with GAPDH as the endogenous control (18). The primers were synthesized by Sheng Gong (Shanghai) Co., Ltd. (Shanghai, China) and the sequences are presented in Table II.

Table II.

Primer sequences for reverse transcription-quantitative polymerase chain reaction.

Table II.

Primer sequences for reverse transcription-quantitative polymerase chain reaction.

Primer namePrimer sequence (5′-3′)
GAPDHF: AGCCACATCGCTCAGACAC
R: GCCCAATACGACCAATCC
B-cell lymphoma 2F: TCCTTCCAGCCTGAGAGCAACC
R: TCACGACGGTAGCGACGAGAG
Caspase-3F: GTAGCAGTGTGCCGCTGTGTC
R: ACTCTTGTCCATCGCCTCTCCTC
Caspase-9F: ATGGAGGAGGCTGACCGGCAACTCCTG
R: TCATGAAGTTTTAAAGAACAGCTTCTTC
Cytochrome CF: AGGAGGCAAGCATAAGAC
R: ATTAGGTCTGCCCTTTCT
Dynamin-related protein 1F: TTCGCCGCCTGGAGGACC
R: CACTTCGTTGCCACAATGCTGAA

[i] F, forward; R, reverse.

Western blot analysis

Total protein cell lysates were isolated from rat lungs or from cells as described previously (19). Protein quantification was assessed using a bicinchoninic acid protein quantification assay and absorbance read at 562 nm was used to determine sample volumes for further analysis. A total of 50 µg total protein from each tissue homogenate was separated by 30% SDS-PAGE and then transferred to polyvinylidene difluoride membranes, which were blocked with 5% skim milk at room temperature for 1 h. Membranes were washed twice with TBST and incubated with primary antibodies at 4°C overnight against Bcl-2 polyclonal antibody (cat. no. YT0470), caspase-3 polyclonal antibody (cat. no. YT0656), caspase-9 polyclonal antibody (cat. no. YT0662), Drp1 Polyclonal antibody (cat. no. YT1414; all 1:1,000; ImmunoWay Biotechnology Company, Plano, TX, USA), Cyt-C antibody (cat. no. 10993-1-AP; 1:1,000; ProteinTech, Group, Inc., Chicago, IL, USA), mouse monoclonal β-actin antibody (1:4,000; cat. no. YM3138; ImmunoWay Biotechnology Company). Membranes were washed twice and incubated with horseradish peroxidase-conjugated goat anti-mouse IgG (1:6,000; cat. no. PMK-014-091; Bioprimacy, Wuhan, China) and goat anti-rabbit IgG (1:8,000; cat. no. PMK-014-090; Bioprimacy, Wuhan, China) for 2 h at room temperature. The blots were visualized with eECL western blot kit (Beyotime Institute of Biotechnology, Jiangsu, China). The intensities of the bands were measured using ImageLab 5.1 Software (Bio-Rad Laboratories, Inc.). Rat β-actin was used as the loading control in western blotting.

Statistical analysis

All experiments were repeated three times. All data are presented as mean ± standard deviation. Comparisons of two or three experimental conditions were evaluated using an unpaired Student's t-test or one-way analysis of variance and the Bonferroni t-test followed by the Bonferroni post-test using GraphPad Prism 5.0 software (GraphPad Software Corp, San Diego, CA, USA). P<0.05 was considered to indicate a statistically significant difference.

Results

Lung histopathology

Histopathological examinations were performed for the lungs of rats subjected to normoxia, IH, IH+25 mg/kg Mdivi-1 and IH+50 mg/kg Mdivi-1. IH caused severe interstitial pneumonia and a large number of inflammatory cells, while following Mdivi-1 intervention, interstitial pneumonia and inflammatory cells were reduced. The effect in the IH+50 mg/kg Mdivi-1 was more obvious compared with in the IH+25 mg/kg Mdivi-1 group (Fig. 1). Consistent with these histopathological observations, the lung injury score in the IH group was significantly increased compared with the control group (P<0.005) and the score in the IH+50 mg/kg Mdivi-1 group was significantly decreased compared with in the IH group (P<0.005).

Detection of Bcl-2, caspase-3, caspase-9, Cyt-C and Drp-1 expression levels via RT-qPCR

After SD rats were exposed to IH for 4 weeks and after WTRL1 cells were exposed to IH for 12 h in culture, the expression of apoptosis markers was increased at the gene level in the rat lung tissue cells and WRTL1 cells. In addition, Mdivi-1 treatment effectively reduced the apoptosis rate of the hypoxia-exposed cells. After SD rats were exposed to IH for 4 weeks, Bcl-2 mRNA levels were significantly decreased (P<0.05; Fig. 2A), but caspase-3 and caspase-9 mRNA levels were significantly increased compared with the control group rats (P<0.05; Fig. 2B and C). After Mdivi-1 intervention, Bcl-2 mRNA levels were significantly increased and caspase-3, and Drp-1 mRNA levels were significantly decreased in the IH+50 mg/kg Mdivi-1 group compared with the IH group (P<0.05; Fig. 2A, B and D). However, no significant difference in Cyt C levels was identified between the groups (Fig. 2E). After WTRL1 cells were exposed to IH for 12 h, Bcl-2 mRNA levels were significantly decreased, but caspase-3, caspase-9, Cyt-C and Drp-1 mRNA levels were significantly increased compared with the control group (P<0.05; Fig. 3A-E). After Mdivi-1 intervention, Bcl-2 mRNA levels were significantly increased and caspase-3, caspase-9, Cyt-C and Drp-1 mRNA levels were significantly decreased compared with in the IH group (P<0.05).

Western blotting results for Bcl-2, caspase-3, caspase-9, Drp-1 and Cyt-C

After SD rats were exposed to IH for 4 weeks and after WTRL1 cells were exposed to IH for 12 h in culture, the expression of apoptosis markers was increased at the protein level in rat lung tissue cells and WRTL1 cells. In addition, Mdivi-1 treatment reduced the apoptosis rate of the hypoxia-exposed cells. Western blotting results for Bcl-2, caspase-3, caspase-9, Cyt-C and Drp-1 protein expression in rat lungs are presented in Fig. 4. After SD rats were exposed to IH for 4 weeks, the protein expression of Bcl-2 in the IH group was significantly decreased, but the protein expression of caspase-3, caspase-9 and Drp-1 was significantly increased compared with the control group (P<0.05; Fig. 4A-E). After Mdivi-1 intervention, the protein expression of Bcl-2 was significantly increased and the protein expression of caspase-3, caspase-9, and Drp-1 was significantly decreased in the IH+50 mg/kg Mdivi-1 group compared with the IH group (P<0.05). After WTRL1 cells were exposed to IH for 12 h, Bcl-2 protein expression was significantly decreased in the IH group and caspase-3, caspase-9 and Drp-1 protein expression was significantly increased (P<0.05; Fig. 5A-E). After Mdivi-1 intervention, Bcl-2 protein expression was significantly increased and caspase-9, Cyt-C, and Drp-1 protein expression was significantly decreased in the IH+100 µM Mdivi-1 group (P<0.05; Fig. 5 A, B and D-F).

Detection of apoptosis levels

The apoptosis rate of WTRL1 cells was positively correlated with the time of exposure to IH. After WTRL1 cell exposure to IH for 6, 12, 24 and 48 h, the rate of apoptosis in WTRL1 cells increased gradually with the IH exposure time, and the apoptosis rate was the highest at 48 h (P<0.05; Fig. 6).

Discussion

OSA is characterized by intermittent airway obstruction and systemic hypoxia during sleep. OSA affects 5–25% of the general population. However, previous studies have illustrated a rising prevalence of moderate-to-severe sleep disordered breathing in men (ranging from 10–49%, depending on age and cohort) and women (ranging from 10–23%) in western countries (20,21). OSA frequently goes unnoticed by people who are at highest risk for developing the disease (2225). OSA is recognized as a low-grade systemic inflammatory disease; it induces inflammatory responses and can be an indication of metabolic deregulation, in addition to posing a great risk for apoptotic cell death (26). Apoptosis can be triggered by two distinct signalling pathways, namely, the intrinsic and extrinsic pathways (27,28). The intrinsic apoptotic pathway is elicited by a wide range of intracellular stress conditions, including cytokine deprivation, DNA damage, oxidative stress, cytosolic Ca2+ overload and endoplasmic reticulum stress. In the extrinsic pathway, apoptosis is triggered by death receptors, including FAS-associated death domain protein, therefore activating caspases-8 and 10 (the initiator caspases), which in turn activate executioner caspases-3, 6 and 7. In the intrinsic pathway, the mitochondrial permeability transition pore serves a pivotal role in regulating the release of pro-apoptotic proteins, including Cyt-C. The Cyt-C released from mitochondria initiates apoptosome assembly, activating factor 1 and caspase-9, an initiator caspase that cleaves and activates caspases-3 and 7 (29).

IH causes mitochondrial and endoplasmic reticulum system damage and produces inflammatory factors that cause lung injury. Sustained and IH-induced cell apoptosis mediates the mitochondrial apoptosis signalling pathway (30). A number of hypoxia and inflammatory factors activate caspases via the mitochondrial apoptosis signalling pathway. Mitochondrial morphology is now recognized as an important determinant of the energetic state of mitochondria. IH/hypercapnia leads to oxidative stress due to the mitochondrial response. Mitochondria constantly undergo fusion and fission, which are essential for organelle fidelity. Lung cell apoptosis occurs through a highly regulated proteolytic process that eliminates unwanted, damaged, or altered cells. However, abnormal mitochondrial fission is involved in regulating apoptosis (31).

Mdivi-1 has emerged as a critical regulator of cellular function and differentiation under hypoxic conditions. However, the role of mitochondrial dynamics in hypoxia-induced angiogenesis remains to be elucidated. The aim of the present study was to investigate the protective effect and possible mechanism of Mdivi-1 in lung cell apoptosis induced by IH.

Several studies have revealed that Drp-1 participates in mitochondrial fission and the subsequent apoptosis induced by various stimuli (32,33). Drp-1 assembles from the cytosol onto mitochondria at focal sites of mitochondrial fission; emerging evidence indicates that it is concomitantly involved in apoptosis with mitochondrial outer membrane permeabilization and that Drp-1 inhibition prevents partially intrinsic apoptosis (34,35). The activity of the prototypical Bcl-2 protein is usually considered anti-apoptotic (36). The Bcl-2 family is classified into three subgroups: Pro-apoptotic proteins (Bax, BAD, Bak and Bok); anti-apoptotic proteins (Bcl-2, Bcl-xL and Bcl-w) and BH3-only members [Bad, Bid, Bim, Noxa and PUMA (a p53-upregulated modulator of apoptosis)]. The caspase family of cysteine-aspartic proteases serves an important role in the initiation and completion of cell apoptosis and executes cellular apoptosis. For instance, caspase-3 is the primary activator of apoptotic DNA fragmentation. Certain reports have demonstrated that the pro-apoptotic signalling of Cyt-C triggers the caspase cascade amplification reaction, which leads to the cleavage of a set of proteins (37,38).

Mitochondria are autonomous and morphologically dynamic organelles that structurally reflect a precise balance of ongoing fission and fusion within a cell. In the two models, in vitro and in vivo, it was observed that Mdivi-1 decreases mitochondrial fission and inhibits cell apoptosis in the lungs. These results agree with evidence from previous studies (39,40). In this study, it was demonstrated that inhibiting mitochondrial fission by Mdivi-1 prevents lung apoptosis during IH and it was demonstrated that IH induced mitochondrial fission and apoptosis via a mitochondrial mechanism involving the transcriptional activation of caspase-3, caspase-9 and Drp-1. The results of the present study provide a well-characterized mechanism explaining how IH stimulates mitochondrial fission and consequent apoptosis, which may offer a novel therapeutic strategy for treating OSA via the mitochondrial apoptosis pathway.

Acknowledgements

Not applicable.

Funding

The present study was funded by the Youth Fund Project of Guizhou Provincial People's Hospital (GZSYQN [2016] grant no. 16); the Technology Project of Guizhou Province (contract [2016] No. 2907); the Major Disease Prevention and Control Research of Guizhou Province (contract [2015] grant no. 78); the Science and Technology Project of a tobacco corporation (Guizhou Province division) of China (contract [201608]).

Availability of data and materials

The datasets used and/or analysed in the present study are available from the corresponding author upon reasonable request.

Authors' contributions

DZ and XY contributed to the conception and design of the study. CY and ZW collected the data and revised the manuscript for important intellectual content. DZ and XZ analysed and interpreted the data and drafted the manuscript. All authors read and approved the final manuscript.

Ethics approval and consent to participate

All procedures performed in studies involving animals were in accordance with the ethical standards of the institution or practice at which the studies were conducted. The study was approved by the Ethics Committee of Guizhou Provincial People's Hospital.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Young T, Evans L, Finn L and Palta M: Estimation of the clinically diagnosed proportion of sleep apnea syndrome in middle-aged men and women. Sleep. 20:705–706. 1997. View Article : Google Scholar : PubMed/NCBI

2 

Punjabi NM: The epidemiology of adult obstructive sleep apnea. Proc Am Thorac Soc. 5:136–143. 2008. View Article : Google Scholar : PubMed/NCBI

3 

Sakao S, Taraseviciene-Stewart L, Lee JD, Wood K, Cool CD and Voelkel NF: Initial apoptosis is followed by increased proliferation of apoptosis-resistant endothelial cells. FASEB J. 19:1178–1180. 2005. View Article : Google Scholar : PubMed/NCBI

4 

Haslip M, Dostanic I, Huang Y, Zhang Y, Russell KS, Jurczak MJ, Mannam P, Giordano F, Erzurum SC and Lee PJ: Endothelial Ucp2 regulates mitophagy and pulmonary hypertension during intermittent-hypoxia. Arterioscler Thromb Vasc Biol. 35:1166–1178. 2015. View Article : Google Scholar : PubMed/NCBI

5 

Zhou S, Yin X, Zheng Y, Miao X, Feng W, Cai J and Cai L: Metallothionein prevents intermittent hypoxia-induced cardiac endoplasmic reticulum stress and cell death likely via activation of Akt signaling pathway in mice. Toxicol Lett. 227:113–123. 2014. View Article : Google Scholar : PubMed/NCBI

6 

Liu KX, Chen GP, Lin PL, Huang JC, Lin X, Qi JC and Lin QC: Detection and analysis of apoptosis- and autophagy-related miRNAs of mouse vascular endothelial cells in chronic intermittent hypoxia model. Life Sci. 193:194–199. 2018. View Article : Google Scholar : PubMed/NCBI

7 

Ren J, Liu W, Deng Y, Li GC, Pan YY, Xie S, Jin M and Liu HG: Losartan attenuates aortic endothelial apoptosis induced by chronic intermittent hypoxia partly via the phospholipase C pathway. Sleep Breath. 21:679–689. 2017. View Article : Google Scholar : PubMed/NCBI

8 

Wang J, Hansen K, Edwards R, Van Houten B and Qian W: Mitochondrial division inhibitor 1 (mdivi-1) enhances death receptor-mediated apoptosis in human ovarian cancer cells. Biochem Biophys Res Commun. 456:7–12. 2015. View Article : Google Scholar : PubMed/NCBI

9 

Kim DY, Jung SY, Kim YJ, Kang S, Park JH, Ji ST, Jang WB, Lamichane S, Lamichane BD, Chae YC, et al: Hypoxia-dependent mitochondrial fission regulates endothelial progenitor cell migration, invasion, and tube formation. Korean J Physiol Pharmacol. 22:203–213. 2018. View Article : Google Scholar : PubMed/NCBI

10 

Gonder JC and Laber K: A renewed look at laboratory rodent housing and management. ILAR J. 48:29–36. 2007. View Article : Google Scholar : PubMed/NCBI

11 

Shortt CM, Fredsted A, Chow HB, Williams R, Skelly JR, Edge D, Bradford A and O'Halloran KD: Reactive oxygen species mediated diaphragm fatigue in a rat model of chronic intermittent hypoxia. Experiment physiology. 99:688–700. 2014. View Article : Google Scholar

12 

Briancon-Marjollet A, Monneret D, Henri M, Hazane-Puch F, Pepin JL, Faure P and Godin-Ribuot D: Endothelin regulates intermittent hypoxia-induced lipolytic remodelling of adipose tissue and phosphorylation of hormone-sensitive lipase. J Physiol. 594:1727–1740. 2016. View Article : Google Scholar : PubMed/NCBI

13 

Poorhassan M, Navae F, Mahakizadeh S, Bazrafkan M, Nikmehr B, Abolhassani F, Ijaz S, Yamini N, Dashti N, Mehrannia K, et al: Flaxseed can reduce hypoxia-induced damages in rat testes. Int J Fertil Steril. 12:235–241. 2018.PubMed/NCBI

14 

Krause BJ, Casanello P, Dias AC, Arias P, Velarde V, Arenas GA, Preite MD and Iturriaga R: Chronic intermittent hypoxia-Induced vascular dysfunction in rats is reverted by N-Acetylcysteine supplementation and arginase inhibition. Front Physiol. 9:9012018. View Article : Google Scholar : PubMed/NCBI

15 

Park SW, Kim KY, Lindsey JD, Dai Y, Heo H, Nguyen DH, Ellisman MH, Weinreb RN and Ju WK: A selective inhibitor of drp1, mdivi-1, increases retinal ganglion cell survival in acute ischemic mouse retina. Invest Ophthalmol Vis Sci. 52:2837–2843. 2011. View Article : Google Scholar : PubMed/NCBI

16 

Burkholder T, Foltz C, Karlsson E, Linton CG and Smith JM: Health evaluation of experimental laboratory mice. Curr Protoc Mouse Biol. 2:145–165. 2012. View Article : Google Scholar : PubMed/NCBI

17 

Kostopanagiotou G, Avgerinos E, Costopanagiotou C, Arkadopoulos N, Andreadou I, Diamantopoulou K, Lekka M, Smyrniotis V and Nakos G: Acute lung injury in a rat model of intestinal ischemia-reperfusion: The potential time depended role of phospholipases A(2). J Surg Res. 147:108–116. 2008. View Article : Google Scholar : PubMed/NCBI

18 

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

19 

Nicole NL, Satriotomo I, Harrigan DJ and Mitchell GS: Acute intermittent hypoxia induced phrenic long-term facilitation despite increased SOD1 expression in a rat model of ALS. Exp Neurol. 273:138–150. 2015. View Article : Google Scholar : PubMed/NCBI

20 

Peppard PE, Young T, Barnet JH, Palta M, Hagen EW and Hla KM: Increased prevalence of sleep-disordered breathing in adults. Am J Epidemiol. 177:1006–10014. 2013. View Article : Google Scholar : PubMed/NCBI

21 

Heinzer R, Vat S, Marques-Vidal P, Marti-Soler H, Andries D, Tobback N, Mooser V, Preisig M, Malhotra A, Waeber G, et al: Prevalence of sleep-disordered breathing in the general population: The HypnoLaus study. Lancet Respir Med. 3:310–318. 2015. View Article : Google Scholar : PubMed/NCBI

22 

Drager LF, Togeiro SM, Polotsky VY and Lorenzi-Filho G: Obstructive sleep apnea: A cardiometabolic risk in obesity and the metabolic syndrome. J Am Coll Cardiol. 62:569–576. 2013. View Article : Google Scholar : PubMed/NCBI

23 

Drager LF, Polotsky VY and Lorenzi-Filho G: Obstructive sleep apnea: An emerging risk factor for atherosclerosis. Chest J. 140:534–542. 2011. View Article : Google Scholar

24 

Sahlin C, Sandberg O, Gustafson Y, Bucht G, Carlberg B, Stenlund H and Franklin KA: Obstructive sleep apnea is a risk factor for death in patients with stroke: A 10-year follow-up. Arch Intern Med. 168:297–301. 2008. View Article : Google Scholar : PubMed/NCBI

25 

Xie C, Zhu R, Tian Y and Wang K: Association of obstructive sleep apnoea with the risk of vascular outcomes and all-cause mortality: A meta-analysis. BMJ Open. 7:e0139832017. View Article : Google Scholar : PubMed/NCBI

26 

Zychowski KE, Sanchez B, Pedrosa RP, Lorenzi-Filho G, Drager LF, Polotsky VY and Campen MJ: Serum from obstructive sleep apnea patients induces inflammatory responses in coronary artery endothelial cells. Atherosclerosis. 254:59–66. 2016. View Article : Google Scholar : PubMed/NCBI

27 

Galluzzi L, Vitale I, Abrams JM, Alnemri ES, Baehrecke EH, Blagosklonny MV, Dawson TM, Dawson VL, El-Deiry WS, Fulda S, et al: Molecular definitions of cell death subroutines: Recommendations of the nomenclature committee on cell death 2012. Cell Death Differ. 19:107–120. 2012. View Article : Google Scholar : PubMed/NCBI

28 

Elmore S: Apoptosis: A review of programmed cell death. Toxicol Pathol. 35:495–516. 2007. View Article : Google Scholar : PubMed/NCBI

29 

Mariño G, Niso-Santano M, Baehrecke EH and Kroemer G: Self-consumption: The interplay of autophagy and apoptosis. Nat Rev Mol Cell Biol. 15:81–94. 2014. View Article : Google Scholar : PubMed/NCBI

30 

Douglas RM, Ryu J, Kanaan A, Del Carmen Rivero M, Dugan LL, Haddad GG and Ali SS: Neuronal death during combined intermittent hypoxia/hypercapnia is due to mitochondrial dysfunction. Am J Physiol Cell Physiol. 298:C1594–C1602. 2012. View Article : Google Scholar

31 

Li J, Donath S, Li Y, Qin D, Prabhakar BS and Li P: miR-30 regulates mitochondrial fission through targeting p53 and the dynamin-related protein-1 pathway. PLoS Genet. 6:e10007952010. View Article : Google Scholar : PubMed/NCBI

32 

Chen Y, Lin JR and Gao PJ: Mitochondrial division inhibitor Mdivi-1 ameliorates angiotensin II-induced endothelial dysfunction. Sheng Li Xue Bao. 68:669–676. 2016.PubMed/NCBI

33 

Oettinghaus B, D'Alonzo D, Barbieri E, Restelli LM, Savoia C, Licci M, Tolnay M, Frank S and Scorrano L: DRP1-dependent apoptotic mitochondrial fission occurs independently of BAX, BAK and APAF1 to amplify cell death by BID and oxidative stress. Biochim Biophys Acta 1857. 1267–1276. 2016.

34 

Tanaka A and Youle RJ: A chemical inhibitor of DRP1 uncouples mitochondrial fission and apoptosis. Mol Cell. 29:409–410. 2008. View Article : Google Scholar : PubMed/NCBI

35 

Frank S, Gaume B, Bergmann-Leitner ES, Leitner WW, Robert EG, Catez F, Smith CL and Youle RJ: The role of dynamin-related protein 1, a mediator of mitochondrial fission, in apoptosis. Dev Cell. 1:515–525. 2001. View Article : Google Scholar : PubMed/NCBI

36 

Czabotar PE, Lessene G, Strasser A and Adams JM: Control of apoptosis by the BCL-2 protein family: Implications for physiology and therapy. Nat Rev Mol Cell Biol. 15:49–63. 2014. View Article : Google Scholar : PubMed/NCBI

37 

Di Pietro R and Zauli G: Emerging non-apoptotic functions of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)/Apo2L. J Cell Physiol. 201:331–340. 2004. View Article : Google Scholar : PubMed/NCBI

38 

Sikora E, Bielak-Zmijewska A, Magalska A, Piwocka K, Mosieniak G, Kalinowska M, Widlak P, Cymerman IA and Bujnicki JM: Curcumin induces caspase-3-dependent apoptotic pathway but inhibits DNA fragmentation factor 40/caspase-activated DNase endonuclease in human Jurkat cells. Mol Cancer Ther. 5:927–934. 2006. View Article : Google Scholar : PubMed/NCBI

39 

Han XJ, Yang ZJ, Jiang LP, Wei YF, Liao MF, Qian Y, Li Y, Huang X, Wang JB, Xin HB and Wan YY: Mitochondrial dynamics regulates hypoxia-induced migration and antineoplastic activity of cisplatin in breast cancer cells. Int J Oncol. 46:691–700. 2015. View Article : Google Scholar : PubMed/NCBI

40 

Sanderson TH, Raghunayakula S and Kumar R: Neuronal hypoxia disrupts mitochondrial fusion. Neuroscience. 301:71–78. 2015. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March-2019
Volume 17 Issue 3

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

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhao D, Yin CY, Ye XW, Wan ZF, Zhao DG and Zhang XY: Mitochondrial separation protein inhibitor inhibits cell apoptosis in rat lungs during intermittent hypoxia . Exp Ther Med 17: 2349-2358, 2019
APA
Zhao, D., Yin, C., Ye, X., Wan, Z., Zhao, D., & Zhang, X. (2019). Mitochondrial separation protein inhibitor inhibits cell apoptosis in rat lungs during intermittent hypoxia . Experimental and Therapeutic Medicine, 17, 2349-2358. https://doi.org/10.3892/etm.2019.7201
MLA
Zhao, D., Yin, C., Ye, X., Wan, Z., Zhao, D., Zhang, X."Mitochondrial separation protein inhibitor inhibits cell apoptosis in rat lungs during intermittent hypoxia ". Experimental and Therapeutic Medicine 17.3 (2019): 2349-2358.
Chicago
Zhao, D., Yin, C., Ye, X., Wan, Z., Zhao, D., Zhang, X."Mitochondrial separation protein inhibitor inhibits cell apoptosis in rat lungs during intermittent hypoxia ". Experimental and Therapeutic Medicine 17, no. 3 (2019): 2349-2358. https://doi.org/10.3892/etm.2019.7201