Open Access

Adiponectin ameliorates the apoptotic effects of paraquat on alveolar type Ⅱ cells via improvements in mitochondrial function

  • Authors:
    • Yarong He
    • Liqun Zou
    • Yaxiong Zhou
    • Hai Hu
    • Rong Yao
    • Yaowen Jiang
    • Wayne Bond Lau
    • Tun Yuan
    • Wen Huang
    • Zhi Zeng
    • Yu Cao
  • View Affiliations

  • Published online on: May 23, 2016     https://doi.org/10.3892/mmr.2016.5328
  • Pages: 746-752
  • Copyright: © He et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

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


Abstract

Previous studies have demonstrated that excessive reactive oxygen/nitrogen species (ROS/RNS)‑induced apoptosis is an important feature of the injury to the lung epithelium in paraquat (PQ) poisoning. However the precise mechanisms of PQ‑induced dysfunction of the mitochondria, where ROS/RNS are predominantly produced, remain to be fully elucidated. Whether globular adiponectin (gAd), a potent molecule protective to mitochondria, regulates the mitochondrial function of alveolar type II cells to reduce PQ‑induced ROS/RNS production remains to be investigated. The current study aimed to investigate the precise mechanisms of PQ poisoning in the mitochondria of alveolar type II cells, and to elucidate the role of gAd in protecting against PQ‑induced lung epithelium injury. Therefore, lung epithelial injury was induced by PQ co‑culture of alveolar type II A549 cells for 24 h. gAd was administrated to and removed from the injured cells in after 24 h. PQ was observed to reduce cell viability and increase apoptosis by ~1.5 fold in A549 cells. The oxidative/nitrative stress, resulting from ROS/RNS and disordered mitochondrial function were evidenced by increased O2‑., NO production and reduced mitochondrial membrane potential (ΔΨ), adenosine 5'‑triphosphate (ATP) content in PQ‑poisoned A549 cells. gAd treatment significantly reversed the PQ‑induced cell injury and mitochondrial dysfunction in A549 cells. The protective effects of gAd were partly abrogated by an adenosine 5'‑monophosphate‑activated protein kinase (AMPK) inhibitor, compound C. The results suggest that reduced ΔΨ and ATP content may result in PQ‑induced mitochondrial dysfunction of the lung epithelium, which constitutes a novel mechanism for gAd exerting pulmonary protection against PQ poisoning via AMPK activation.

Introduction

Previous studies have demonstrated that apoptosis is an important process in injured to the lung epithelium in paraquat (PQ) poisoning (1,2). The mechanisms of apoptosis are highly complex and involve oxidative/nitrative stress impairment to the function of mitochondria with the reduction of the mitochondrial membrane potential (ΔΨ), resulting in an increase of superoxide formation and reduced production of adenosine 5′-triphosphate (ATP) (3,4). Previous studies suggest that adenosine 5′-monophosphate-activated protein kinase (AMPK) is one of the most important molecules in the stimulation of ATP production and protection against oxidative/nitrative stress in mitochondria (5,6). However, whether dysfunction of the mitochondria and reduction of ATP via inactivation of AMPK serve crucial roles in the PQ-induced apoptosis of alveolar epithelial type II cells remains to be fully elucidated.

Adiponectin (APN), an adipocytokine predominantly secreted by adipocytes and found at high levels in the plasma, has been reported to exhibit anti-insulin resistance and anti-inflammatory and anti-apoptotic properties (79). Circulating APN is present as trimeric, hexameric or oligomeric complexes of monomers, and cleavage to produce the C-terminal globular domain, gAd, has been proposed as an important regulatory step in the action of APN. This is due to the fact that this C-terminal fragment has been reported to mediate potent physiological effects (10,11). Previous studies have suggested that the potential role of gAd against apoptosis is mediated via the activation of AMPK (1214). APN-knockout mice exhibit inhibition of AMPK, a reduction in the levels of ATP, an increase of mitochondrial swelling and ROS/RNS production, resulting in augmented apoptosis, while administration of gAd reversed these effects. The protective effects of gAd have been reported to be reversed by administration of the AMPK inhibitor (compound C) (15). It remains unclear whether APN may protect alveolar type II cells from PQ poisoning via activation of AMPK and increasing the production of ATP in mitochondria.

Therefore, the aims of the current study were: i) To determine whether mitochondrial function is impaired and ATP is reduced in PQ-poisoned alveolar type II cells via inactivation of AMPK; ii) to determine the effect of gAd against PQ-induced apoptosis and in alveolar type II cells; and if so, iii) whether gAd mediates protective effects via activation of AMPK and increasing production of ATP.

Materials and methods

Materials

All reagents were purchased from Sigma-Aldrich (St. Louis, MO, USA), unless otherwise specified. RPMI 1640 medium was obtained from Gibco; Thermo Fisher Scientific, Inc. (Waltham, MA, USA). Penicillin/streptomycin was obtained from Wisent, Inc. (Saint-Bruno, QC, Canada). PQ was purchased from Tokyo Chemical Industry Co., Ltd. (Tokyo, Japan). The globular domain of APN (gAd) was purchased from Beijing Adipobiotech, Inc. (Beijing, China). Compound C, an AMPK Inhibitor, was obtained from Merck Millipore (Darmstadt, Germany). The Annexin V-Fluorescein Isothiocyanate (FITC) Apoptosis Detection kit I was from Nanjing KeyGen Biotech Co., Ltd. (Nanjing, China). JC-1 and dihydroethidium (DHE) were purchased from Molecular Probes; Thermo Fisher Scientific, Inc. The Nitrate/Nitrite Colorimetric Assay kit was from Nanjing Jiancheng Biochemical Reagent Co. (Nanjing, China). The ATP Bioluminescent Somatic Cell assay kit was from Sigma-Aldrich.

Cell cultures and treatments

The human lung adenocarcinoma cell line A549 (Shanghai Institute of Biochemistry and Cell Biology, Shanghai, China) was grown in a monolayer culture in RPMI 1640 medium supplemented with 10% fetal bovine serum, 1% glutamine, 1% (v/v) streptomycin/penicillin. The culture medium was refreshed two to three times per week and was subcultured upon reaching 80% confluence. The cells were then seeded onto flat-bottom six-well or 96-well plates with growth medium at 37°C in a humidified atmosphere at 5% CO2, followed by administration of PQ at 300 µM for 24 h for the PQ, PA and PCA groups. Subsequently, gAd (2.5 µg/ml) was added and the cells were cultured for another 24 h, in order to investigate the role of gAd in PQ-induced cytotoxicity. PCA group cells were also treated with compound C (1 µM) for 30 min prior to the addition of PQ. The optimal doses and time points for gAd, PQ and compound C were determined through preliminary experiments (data not shown).

Measurement of cell viability

The 3-(4, 5-methylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay was conducted as previously described (16). Briefly, following PQ challenge, culture media was refreshed with media containing MTT reagent (5 mg/ml) and cells were incubated under standard conditions for an additional 4 h. The culture media was then carefully aspirated and 100 µl dimethylsulfoxide was added per well to solubilize the formazan crystals. Following agitation, absorbance was measured spectrophotometrically at a wavelength of 490 nm using a Benchmark Plus Microplate Spectrophotometer (Bio-Rad Laboratories, Inc., Hercules, CA, USA). Viabilities of the challenged cells were expressed relative to control cells.

Measurement of necrosis and apoptosis

Apoptotic and necrotic cell death were measured using flow cytometry with the Annexin V-FITC Apoptosis Detection kit I (17). At 24 h after administration of gAd, cells were trypsinized (Thermo Fisher Scientific, Inc.) without ethylenediaminetetraacetic acid and labelled with the fluorochromes in the absence of light for 10 min at room temperature. Fluorescence was measured by flow cytometric analysis using a FACSCalibur flow cytometer (BD Biosciences, San Jose, CA, USA) to monitor green fluorescence (525 nm band-pass filter) for Annexin V-fluorescein conjugate and red fluorescence (575 nm band-pass filter) for PI respectively. The Annexin V-FITC-/PI-cell population was regarded as normal, while Annexin V-FITC+/PI-cells were taken as an indicator of early apoptosis, Annexin V-FITC+/PI+ as late apoptosis and Annexin V-FITC-/PI+ as necrosis. The data were analyzed using CellQuest software (BD Biosciences). A minimum of 30,000 gated events were acquired per sample.

Measurement of superoxide production

Production of superoxides was evaluated intracellularly using the superoxide-sensitive dye DHE (18). DHE is oxidized by superoxides to a novel product, which binds to DNA enhancing intracellular fluorescence. Culture medium was aspirated and cells were incubated with DHE (5 µM final concentration) for 10 min at 37°C in the dark. Following two rinses in phosphate-buffered saline, cells were photographed using an Axiostart 50 (Zeiss, Oberkochen, Germany) microscope equipped with a Canon PowerShot G5 epifluorescence attachment (Canon, Inc., Tokyo, Japan). Between five and six photographs were captured from each well. The fluorescence intensity values for each photo were determined using Adobe Photoshop software (version 4; Adobe Systems, Inc., San Jose, CA, USA).

Measurement of nitrites

Nitrites were measured in the culture supernatants by a colorimetric assay using a procedure based on the Greiss reaction with sodium nitrite as the standard (19). Culture medium was collected and stored at −80°C until analysis with the Nitrate/Nitrite Colorimetric Assay kit, according to the manufacturer's instructions.

Measurement of ΔΨ

The membrane potential assay was conducted as previously described (20). The relative ΔΨ was assessed using a laser scanning confocal microscope (SP8; Leica Microsystems GmbH, Wetzlar, Germany) analysis of cells stained with JC-1. Cells were incubated with 10 µg/ml JC-1 for 30 min at 37°C. Subsequent to applying the dye, the cells were scanned with the confocal microscope using a 10x objective lens. Fluorescence was excited by the 488-nm line of an argon laser and the 543-nm line of a helium/neon laser. The red emission of the dye is due to a potential-dependent aggregation in the mitochondria, reflecting high ΔΨ. Green fluorescence indicates the monomeric form of JC-1, appearing in the cytosol following mitochondrial membrane depolarization, indicating low ΔΨ. The ratio of red/green indicated the mitochondrial membrane depolarization states of different cells.

Measurement of ATP

The amount of ATP was measured in the cultured cells using a luminometer (Bio Orbit 1251 Luminometer; Bio-Orbit, Turku, Finland) as previously described (21). Subsequent to exposure to different experimental conditions, cells were collected to react with different working solutions of from the ATP Bioluminescent Somatic Cell assay kit, according to the manufacturer's instructions. All reactions for a given sample were monitored simultaneously and calibrated with the addition of an ATP standard from the kit.

Statistics

Data are presented as the mean ± standard error (n=5) and were analyzed for statistical significance by one- or two-way analysis of variance with were least significant difference or Student-Newman-Keuls post hoc analyses. Statistical analysis and the significance of the data were determined using SPSS software, version 12.0 (SPSS, Inc., Chicago, IL, USA). P<0.05 was considered to indicate a statistically significant difference.

Results

gAd reduced PQ-mediated cytotoxicity

As presented in Fig. 1, PQ significantly reduced the cell viability of A549 cells, while gAd alleviated the PQ-induced cytotoxicity without any effects on the cells alone. However, pretreatment with compound C significantly reversed the protective effects of gAd on the viability reduced by PQ.

gAd reduced PQ-mediated apoptosis

The results indicated that there was a low level of cell death in the control group (Fig. 2A). However, PQ was observed to induce apoptosis in ~37.5% cells (Fig. 2B). However, gAd significantly reversed the PQ-induced cytotoxicity, reducing the percentage of apoptotic cells to 22.3% (P<0.01). However, the pretreatment with compound C significantly reversed gAd's protective effect (Fig. 2).

gAd reduced PQ-mediated superoxide production

As presented in Fig. 3, control cultures generally exhibited low fluorescence intensity while PQ-treated cultures exhibited bright red fluorescence. This indicated that PQ significanlty increased O2−. production in cells compared with that of the controls (P<0.001). gAd inhibited the O2−. production induced by PQ significanlty. However, pre-treatment with compound C partly reversed gAd's effects on O2−. production.

gAd reduced PQ-mediated nitrite production

Addition of PQ to the cultures significantly increased nitrite production compared with the controls (P<0.01). Administration of gAd or compound C significantly reversed the increase, however pre-treatment with compound C did not reverse the effects of gAd on nitrite production (Fig. 4).

gAd reduced PQ-mediated ΔΨ depolarization

Incubation with PQ depolarized the ΔΨ significantly, resulting in a low ratio of red/green. Treatment with gAd repolarized the ΔΨ of the cells, resulting in a greater ratio of red/green, compared with PQ group (P<0.01). However, compound C reversed the repolarization of gAd, resulting in a lower ratio of red/green as compared with the gAd group (P<0.01; Fig. 5).

gAd partly reversed the PQ-mediated reduction in ATP

Addition of PQ significantly reduced the intracellular ATP concentration, as compared with the initial control levels: 21% after 24 h, irrespective of the presence of other medium supplements or stimulation (P<0.05). Conversely, treatment with gAd increased the ATP level to ~50% of that of the control group (P<0.05). However, compound C reversed the gAd-mediated increase (P<0.05; Fig. 6).

Discussion

The most prevalent cause of morbidity and mortality in patients with PQ poisoning is organ injury, in particular, injury to the lungs (22). It has been demonstrated that excessive ROS/RNS stress resulting from impaired mitochondria, is an important feature of the injured lung epithelium in PQ poisoning, though the underlying mechanism responsible for the mitochondrial dysfunction remains unclear (23). Therefore, studies have focussed upon clarifying the underlying mechanisms for PQ-associated mitochondria injury of alveolar type II cells (24,25) and developing novel strategies for the treatment of PQ-poisoned patients (26). In the present study, an established PQ poisoning model with A549 cells cultured with PQ for 24 h was used, and it was confirmed that apoptosis and cell death were exacerbated in the presence of PQ. Furthermore, it was indicated that PQ markedly disturbed mitochondrial function, depolarizing ΔΨ and reducing ATP production, resulting in progressive increases of O2−. and NO. This may be explained by the fact that PQ2+ induced the release of Ca2+ from cytoplasm (27), which may induce the depolarization of mitochondria and reduce ΔΨ (28). This results in reductions of ATP synthesis to less than 50% of the normal level, thus activating pro-apoptotic and pro-necrotic proteins, including Bax and caspase 3/7/8/9 (29,30). As a result, the injured mitochondria produce excessive O2−. and NO, which attached to the mitochondrial membrane in turn, and resulted in increases in ROS/RNS production and ultimately inducing apoptosis (31).

Injury resulting from PQ poisoning was however, reversed by treatment with gAd in the current study. Although the associated mechanisms remain to be fully elucidated, a previous study demonstrated the potential cellular protective effects of gAd against apoptosis via inhibiting mitochondrial dysfunction and ROS/RNS attachment (32). Therefore, the current study suggested that gAd may protect the alveolar type II cells aginst PQ-induced cell injury via inhibiting mitochondrial dysfunction and reducing oxidative/nitrative injury.

In order to further elucidate the underlying mechanisms for the cellular-protective effects of gAd in PQ poisoning against mitochondrial dysfunction and oxidative/nitrative injury, the signaling of AMPK, a protective kinase in oxidative/nitrative injury and a downstream mediator of gAd's mitochondrial protective actions, was investigated (33). It was identified that the protective effects of gAd could be reversed by an AMPK inhibitor, compound C, which has been previously identified to block the cardiac protective benefits of gAd treatment against cardiac myocardial hypoxia/reoxygenation injury (15). This result clarified the critical role of AMPK in the pneumonocyte-protective effects of gAd, indicating it may be used in the therapy for PQ poisoning. This observation is consistent with a previous study by Yan et al (15), which demonstrated that gAd was able to activate AMPK-peroxisome proliferator-activated receptor γ coactivator 1-α, resulting in increased ATP production, improving ΔΨ, thus leading to return to normal function. These results indicate that AMPK may be one of the key molecules in gAd's inhibition of mitochondrial dysfunction induced by PQ. However, interspecies differences (34,35), animal model establishment (36,37) and the injury procedure (38) may affect whether AMPK signaling serves a beneficial or detrimental role in the mitochondria. Experiments using animal models should be conducted in order to further clarify the mechanisms.

There were limitations in the present study. The inactivation of AMPK was not introduced when PQ was added to the cells, due to the high rate of mortality, thus the ability to directly demonstrate the role of AMPK in PQ-poisoned mitochondria following oxidative/nitrative injury was compromised. In addition, gAd treatment in PQ poisoning may modulate numerous other key molecules in addition to AMPK-ATP (3941). Thus, investigation of these molecules in oxidative/nitrative injury requires additional investigation.

In conclusion, it was demonstrated that the ΔΨ exhibited progressive reductions with increased oxidative/nitrative injury in PQ-induced apoptosis. In addition, it was identified that in PQ poisoning, ΔΨ is increased in response to gAd treatment, which significantly attenuated mitochondrial dysfunction and oxidative/nitrative injury in PQ-poisoned alveolar type II cells. The present study provided indirect evidence for the possible role of the AMPK-ATP signaling pathway in gAd against oxidative/nitrative injury originating from the mitochondria following PQ poisoning. To the best of our knowledge, this is the first study to demonstrate the ability of gAd in attenuating oxidative/nitrative injury in PQ poisoning of lung cells beyond its metabolic actions. These observations add to the current understanding of the pathophysiology of PQ poisoning and the mechanisms of the widely used gAd therapy. This may aid in the development of future therapeutic strategies capable of assisting gAd in protecting against PQ-induced lung injury.

Acknowledgments

The current study was supported by the National Natural Science Foundation of China (grant nos. 30900493 and 81471836) and the Science and Technology Department Foundation of Sichuan Province (grant no. 2013JY0011). The authors would additionally like to acknowledge Professor Xin-liang Ma of Thomas Jefferson University (Philadelphia, PA, USA) for his help and advice aiding in the completion of the present study.

Abbreviations:

ROS/RNS

reactive oxygen/nitrogen species

PQ

paraquat

gAd

globular adiponectin

ATP

adenosine 5′-triphosphate

AMPK

adenosine 5′-monophosphate-activated protein kinase

References

1 

Vieira DN and de Azevedo-Bernarda R: Paraquat poisoning. Pulmonary lesions. J Toxicol Clin Exp. 9:177–186. 1989.In French. PubMed/NCBI

2 

Cai Q, Lu Z, Hong G, Jiang X, Wu Z, Zheng J, Song Q and Chang Z: Recombinant adenovirus Ad-RUNrf2 reduces paraquat-induced A549 injury. Hum Exp Toxicol. 31:1102–1112. 2012. View Article : Google Scholar : PubMed/NCBI

3 

Golbidi S, Botta A, Gottfred S, Nusrat A, Laher I and Ghosh S: Glutathione administration reduces mitochondrial damage and shifts cell death from necrosis to apoptosis in ageing diabetic mice hearts during exercise. Br J Pharmacol. 171:5345–5360. 2014. View Article : Google Scholar : PubMed/NCBI

4 

Haagsman HP, Schuurmans EA, Batenburg JJ and Van Golde LM: Phospholipid synthesis in isolated alveolar type II cells exposed in vitro to paraquat and hyperoxia. Biochem J. 245:119–126. 1987. View Article : Google Scholar : PubMed/NCBI

5 

Kurokawa H, Sugiyama S, Nozaki T, Sugamura K, Toyama K, Matsubara J, Fujisue K, Ohba K, Maeda H, Konishi M, et al: Telmisartan enhances mitochondrial activity and alters cellular functions in human coronary artery endothelial cells via AMP-activated protein kinase pathway. Atherosclerosis. 239:375–385. 2015. View Article : Google Scholar : PubMed/NCBI

6 

Barreto-Torres G, Hernandez JS, Jang S, Rodríguez-Muñoz AR, Torres-Ramos CA, Basnakian AG and Javadov S: The beneficial effects of AMP kinase activation against oxidative stress are associated with prevention of PPARα-cyclophilin D interaction in cardiomyocytes. Am J Physiol Heart Circ Physiol. 308:H749–H758. 2015. View Article : Google Scholar : PubMed/NCBI

7 

Berg AH, Combs TP and Scherer PE: ACRP30/adiponectin: An adipokine regulating glucose and lipid metabolism. Trends Endocrinol Metab. 13:84–89. 2002. View Article : Google Scholar : PubMed/NCBI

8 

Okamoto Y, Kihara S, Funahashi T, Matsuzawa Y and Libby P: Adiponectin: A key adipocytokine in metabolic syndrome. Clin Sci (Lond). 110:267–278. 2006. View Article : Google Scholar

9 

Robinson K, Prins J and Venkatesh B: Clinical review: Adiponectin biology and its role in inflammation and critical illness. Crit Care. 15:2212011. View Article : Google Scholar : PubMed/NCBI

10 

Kadowaki T, Yamauchi T and Kubota N: The physiological and pathophysiological role of adiponectin and adiponectin receptors in the peripheral tissues and CNS. FEBS Lett. 582:74–80. 2008. View Article : Google Scholar

11 

Fang X and Sweeney G: Mechanisms regulating energy metabolism by adiponectin in obesity and diabetes. Biochem Soc Trans. 34:798–801. 2006. View Article : Google Scholar : PubMed/NCBI

12 

Tao L, Gao E, Jiao X, Yuan Y, Li S, Christopher TA, Lopez BL, Koch W, Chan L, Goldstein BJ and Ma XL: Adiponectin cardioprotection after myocardial ischemia/reperfusion involves the reduction of oxidative/nitrative stress. Circulation. 115:1408–1416. 2007. View Article : Google Scholar : PubMed/NCBI

13 

Kim JE, Song SE, Kim YW, Kim JY, Park SC, Park YK, Baek SH, Lee IK and Park SY: Adiponectin inhibits palmitate-induced apoptosis through suppression of reactive oxygen species in endothelial cells: Involvement of cAMP/protein kinase A and AMP-activated protein kinase. J Endocrinol. 207:35–44. 2010. View Article : Google Scholar : PubMed/NCBI

14 

Wei CD, Li Y, Zheng HY, Sun KS, Tong YQ, Dai W, et al: Globular adiponectin protects H9c2 cells from palmitate-induced apoptosis via Akt and ERK1/2 signaling pathways. Lipids Health Dis. 11:1352012. View Article : Google Scholar : PubMed/NCBI

15 

Yan W, Zhang H, Liu P, Wang H, Liu J, Gao C, Liu Y, Lian K, Yang L, Sun L, et al: Impaired mitochondrial biogenesis due to dysfunctional adiponectin-AMPK-PGC-1α signaling contributing to increased vulnerability in diabetic heart. Basic Res Cardiol. 108:3292013. View Article : Google Scholar

16 

Kim JE, Ahn MW, Baek SH, Lee IK, Ki YW, Kim JY, Dan JM and Park SY: AMPK activator, AICAR, inhibits palmitate-induced apoptosis in osteoblast. Bone. 43:394–404. 2008. View Article : Google Scholar : PubMed/NCBI

17 

Krysko DV, Vanden Berghe T, D'Herde K and Vandenabeele P: Apoptosis and necrosis: Detection, discrimination and phagocytosis. Methods. 44:205–221. 2008. View Article : Google Scholar : PubMed/NCBI

18 

Radad K, Rausch WD and Gille G: Rotenone induces cell death in primary dopaminergic culture by increasing ROS production and inhibiting mitochondrial respiration. Neurochem Int. 49:379–386. 2006. View Article : Google Scholar : PubMed/NCBI

19 

Green LC, Wagner DA, Glogowski J, Skipper PL, Wishnok JS and Tannenbaum SR: Analysis of nitrate, nitrite and 15 N.nitrate in biological fluids. Anal Biochem. 126:131–138. 1982. View Article : Google Scholar : PubMed/NCBI

20 

Akao M, Ohler A, O'Rourke B and Marbán E: Mitochondrial ATP-sensitive potassium channels inhibit apoptosis induced by oxidative stress in cardiac cells. Circ Res. 88:1267–1275. 2001. View Article : Google Scholar : PubMed/NCBI

21 

Yeh ST, Guo HR, Su YS, Lin HJ, Hou CC, Chen HM, Chang MC and Wang YJ: Protective effects of N-acetylcysteine treatment post acute paraquat intoxication in rats and in human lung epithelial cells. Toxicology. 223:181–190. 2006. View Article : Google Scholar : PubMed/NCBI

22 

Onyon LJ and Volans GN: The epidemiology and prevention of paraquat poisoning. Hum Toxicol. 6:19–29. 1987. View Article : Google Scholar : PubMed/NCBI

23 

Gaudreault P, Karl PI and Friedman PA: Paraquat and putrescine uptake by lung slices of fetal and newborn rats. Drug Metab Dispos. 12:550–552. 1984.PubMed/NCBI

24 

Wang GY, Hirai K and Shimada H: Mitochondrial breakage induced by the herbicide paraquat in cultured human lung cell. J Electron Microsc (Tokyo). 3:181–184. 1992.

25 

Chen YW, Yang YT, Hung DZ, Su CC and Chen KL: Paraquat induces lung alveolar epithelial cell apoptosis via Nrf-2-regulated mitochondrialdysfunction and ER stress. Arch Toxicol. 86:1547–58. 2012. View Article : Google Scholar : PubMed/NCBI

26 

Shokrzadeh M, Shaki F, Mohammadi E, Rezagholizadeh N and Ebrahimi F: Edaravone decreases paraquat toxicity in a549 cells and lung isolated mitochondria. Iran J Pharm Res. 13:675–681. 2014.PubMed/NCBI

27 

Chang X, Lu W, Dou T, Wang X, Lou D, Sun X, et al: Paraquat inhibits cell viability via enhanced oxidative stress and apoptosis in human neural progenitor cells. Chem Biol Interact. 206:248–255. 2013. View Article : Google Scholar : PubMed/NCBI

28 

Huang CL, Lee YC, Yang YC, Kuo TY and Huang NK: Minocycline prevents paraquat-induced cell death through attenuating endoplasmic reticulum stress and mitochondrial dysfunction. Toxicol Lett. 209:203–210. 2012. View Article : Google Scholar : PubMed/NCBI

29 

Helewski KJ, Kowalczyk-Ziomek GI and Konecki J: Apoptosis and necrosis-two different ways leading to the same target. Wiad Lek. 59:679–684. 2006.In Polish.

30 

Kasahara A and Scorrano L: Mitochondria: From cell death executioners to regulators of cell differentiation. Trends Cell Biol. 24:761–770. 2014. View Article : Google Scholar : PubMed/NCBI

31 

Wang Z, Wang J, Xie R, Liu R and Lu Y: Mitochondria-derived reactive oxygen species play an important role in Doxorubicin-induced platelet apoptosis. Int J Mol Sci. 16:11087–11100. 2015. View Article : Google Scholar : PubMed/NCBI

32 

Park M, Youn B, Zheng XL, Wu D, Xu A and Sweeney G: Globular adiponectin, acting via AdipoR1/APPL1, protects H9c2 cells from hypoxia/reoxygenation-induced apoptosis. PLoS One. 6:e191432011. View Article : Google Scholar : PubMed/NCBI

33 

Lin Z, Wu F, Lin S, Pan X, Jin L, Lu T, Shi L, Wang Y, Xu A and Li X: Adiponectin protects against acetaminophen-induced mitochondrial dysfunction and acute liver injury by promoting autophagy in mice. J Hepatol. 61:825–831. 2014. View Article : Google Scholar : PubMed/NCBI

34 

Zheng A, Li H, Xu J, Cao K, Li H, Pu W, Yang Z, Peng Y, Long J, Liu J and Feng Z: Hydroxytyrosol improves mitochondrial function and reduces oxidative stress in the brain of db/db mice: Role of AMP-activated protein kinase activation. Br J Nutr. 113:1667–1676. 2015. View Article : Google Scholar : PubMed/NCBI

35 

Chen H, Wang JP, Santen RJ and Yue W: Adenosine monophosphate activated protein kinase (AMPK), a mediator of estradiol-induced apoptosis in long-term estrogen deprived breast cancer cells. Apoptosis. 20:821–830. 2015. View Article : Google Scholar : PubMed/NCBI

36 

Chen LJ, Na R, Gu M and Ran QT: Reduction in mitochondria H2O2 protects against insulin resistance through a mechanism of Akt and AMPK activation. Free Radic Biol Med. 47:S852009.

37 

Pan JS, Huang L, Belousova T, Lu L, Yang Y, Reddel R, Chang A, Ju H, DiMattia G, Tong Q, et al: Stanniocalcin-1 inhibits renal ischemia/reperfusion injury via an AMP-activated protein kinase-dependent pathway. J Am Soc Nephrol. 26:364–378. 2015. View Article : Google Scholar :

38 

Wu SB, Wu YT, Wu TP and Wei YH: Role of AMPK-mediated adaptive responses in human cells with mitochondrial dysfunction to oxidative stress. Biochim Biophys Acta. 1840.1331–1344. 2014.

39 

Qiao LP, Kinney B, Yoo HS, Lee B, Schaack J and Shao JH: Adiponectin increases skeletal muscle mitochondrial biogenesis by suppressing mitogen-activated protein kinase phosphatase-1. Diabetes. 61:1463–1470. 2012. View Article : Google Scholar : PubMed/NCBI

40 

Zhou M, Xu A, Tam PK, Lam KS, Chan L, Hoo RL, Liu J, Chow KH and Wang Y: Mitochondrial dysfunction contributes to the increased vulnerabilities of adiponectin knockout mice to liver injury. Hepatology. 48:1087–1096. 2008. View Article : Google Scholar : PubMed/NCBI

41 

Yamauchi T, Iwabu M, Okada-Iwabu M and Kadowaki T: Adiponectin receptors: A review of their structure, function and how they work. Best Pract Res Clin Endocrinol Metab. 28:15–23. 2014. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2016
Volume 14 Issue 1

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

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
He Y, Zou L, Zhou Y, Hu H, Yao R, Jiang Y, Lau WB, Yuan T, Huang W, Zeng Z, Zeng Z, et al: Adiponectin ameliorates the apoptotic effects of paraquat on alveolar type Ⅱ cells via improvements in mitochondrial function. Mol Med Rep 14: 746-752, 2016
APA
He, Y., Zou, L., Zhou, Y., Hu, H., Yao, R., Jiang, Y. ... Cao, Y. (2016). Adiponectin ameliorates the apoptotic effects of paraquat on alveolar type Ⅱ cells via improvements in mitochondrial function. Molecular Medicine Reports, 14, 746-752. https://doi.org/10.3892/mmr.2016.5328
MLA
He, Y., Zou, L., Zhou, Y., Hu, H., Yao, R., Jiang, Y., Lau, W. B., Yuan, T., Huang, W., Zeng, Z., Cao, Y."Adiponectin ameliorates the apoptotic effects of paraquat on alveolar type Ⅱ cells via improvements in mitochondrial function". Molecular Medicine Reports 14.1 (2016): 746-752.
Chicago
He, Y., Zou, L., Zhou, Y., Hu, H., Yao, R., Jiang, Y., Lau, W. B., Yuan, T., Huang, W., Zeng, Z., Cao, Y."Adiponectin ameliorates the apoptotic effects of paraquat on alveolar type Ⅱ cells via improvements in mitochondrial function". Molecular Medicine Reports 14, no. 1 (2016): 746-752. https://doi.org/10.3892/mmr.2016.5328