AI Chat Paper
Note: Please note that the following content is generated by AMiner AI. SciOpen does not take any responsibility related to this content.
{{lang === 'zh_CN' ? '文章概述' : 'Summary'}}
{{lang === 'en_US' ? '中' : 'Eng'}}
Chat more with AI
PDF (8.3 MB)
Collect
Submit Manuscript AI Chat Paper
Show Outline
Outline
Show full outline
Hide outline
Outline
Show full outline
Hide outline
Open Access

Endoplasmic reticulum stress improved chicken tenderness, promoted apoptosis and autophagy during postmortem ageing

Yuwei ChaiaLin Chena( )Shuya XiangaLinxuan WuaXuebo LiuaJun LuobXianchao Fenga,( )
College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China
College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China

Peer review under responsibility of Tsinghua University Press.

Show Author Information

Highlights

• Endoplasmic reticulum (ER) stress reduced shear force and increased MFI.

• ER stress promoted degradation of myofibrillar proteins and structural destruction of myofibrils.

• ER stress induced apoptosis through regulation of the Bcl-2 family.

• ER stress promoted autophagy in postmortem ageing

Graphical Abstract

Abstract

In this study, endoplasmic reticulum (ER) stress inducer tunicamycin (TM) and inhibitor 4-phenylbutyric acid (4-PBA) were used to treat postmortem chicken breast muscle to investigate changes in tenderness and effects on apoptosis and autophagy during 5 days ageing. TM-induced ER stress reduced shear force, enhanced myofibril fragmentation index (MFI), disrupted myofibril structure, increased desmin degradation, and activated μ-calpain and caspase-12. In addition, TM-induced ER stress increased the expression of Bax, Bim, and cytochrome c, and decreased the expression of Bcl-xL. Furthermore, TM-induced ER stress improved the conversion of LC3I to LC3II, raised the expression of Beclin-1, and decreased the expression of p62, PI3K, and mTOR. The opposite results were observed after 4-PBA treatment. These results suggested that ER stress could improve chicken tenderness, promote apoptosis and autophagy during chicken postmortem ageing.

References

[1]

G.Y. Xiong, X.Y. Fu, D.M. Pan, et al., Influence of ultrasound-assisted sodium bicarbonate marination on the curing efficiency of chicken breast meat, Ultrason. Sonochem. 60 (2020) 104808. https://doi.org/10.1016/j.ultsonch.2019.104808.

[2]

T.J. Yan, C.L. Hou, Z.Y. Wang, et al., Effects of chilling rate on progression of rigor mortis in postmortem lamb meat, Food Chem. 373 (2022) 131463. https://doi.org/10.1016/j.foodchem.2021.131463.

[3]

Z. Soji, Effect of the muscle nanostructure changes during post-mortem aging on tenderness of different beef breeds, Anim. Biosci. 34 (2021) 1849-1858. https://doi.org/10.5713/ajas.20.0488.

[4]

C.M. Kemp, P.L. Sensky, R.G. Bardsley, et al., Tenderness: an enzymatic view, Meat Sci. 84 (2010) 248-256. https://doi.org/10.1016/j.meatsci.2009.06.008.

[5]

Z.J. Ding, F. Huang, C.J. Zhang, et al., Effect of heat shock protein 27 on the invitro degradation of myofibrils by caspase-3 and μ-calpain, Int. J. Food Sci. Technol. 53 (2018) 121-128. https://doi.org/10.1111/ijfs.13565.

[6]

X. Li, L. Hu, X.R. Zhu, et al., The effect of caspase-3 in mitochondrial apoptosis activation on degradation of structure proteins of Esox lucius during postmortem storage, Food Chem. 367 (2022) 130767. https://doi.org/10.1016/j.foodchem.2021.130767.

[7]

Q. Hou, R. Liu, X.N. Tian, et al., Involvement of protein S-nitrosylation in regulating beef apoptosis during postmortem aging, Food Chem. 326 (2020) 126975. https://doi.org/10.1016/j.foodchem.2020.126975.

[8]

T.T. Wang, X.C. Feng, L.Z. Li, et al., Effects of quercetin on tenderness, apoptotic and autophagy signalling in chickens during post-mortem ageing, Food Chem. 383 (2022) 132409. https://doi.org/10.1016/j.foodchem.2022.132409.

[9]

M. Izadi, T. A. Ali, E. Pourkarimi, Over fifty years of life, death, and cannibalism: a historical recollection of apoptosis and autophagy, Int. J. Mol. Sci. 22 (2021) 12466. https://doi.org/10.3390/ijms222212466.

[10]

J.Y. Zhang, G.Y. Ma, Z.B. Guo, et al., Study on the apoptosis mediated by apoptosis-inducing-factor and influencing factors of bovine muscle during postmortem aging, Food Chem. 266 (2018) 359-367. https://doi.org/10.1016/j.foodchem.2018.06.032.

[11]

F. Huang, M. Huang, H. Zhang, et al., Changes in apoptotic factors and caspase activation pathways during the postmortem aging of beef muscle, Food Chem. 190 (2016) 110-114. https://doi.org/10.1016/j.foodchem.2015.05.056.

[12]

J.Y. Zhang, Q.L. Yu, L. Han, et al., Effects of lysosomal iron involvement in the mechanism of mitochondrial apoptosis on postmortem muscle protein degradation, Food Chem. 328 (2020) 127174. https://doi.org/10.1016/j.foodchem.2020.127174.

[13]

A.E. Frakes, A. Dillin, The UPRER: sensor and coordinator of organismal homeostasis, Mol. Cell 66 (2017) 761-771. https://doi.org/10.1016/j.molcel.2017.05.031.

[14]

J. Doherty, E.H. Baehrecke, Life, death and autophagy, Nat. Cell Biol. 20 (2018) 1110-1117. https://doi.org/10.1038/s41556-018-0201-5.

[15]

M. Garcia-Macia, V. Sierra, A. Palanca, et al., Autophagy during beef aging, Autophagy 10 (2014) 137-143. https://doi.org/10.4161/auto.26659.

[16]

C. Chen, J.Y. Zhang, Z.B. Guo, et al., Effect of oxidative stress on AIF-mediated apoptosis and bovine muscle tenderness during postmortem aging, J. Food Sci. 85 (2020) 77-85. https://doi.org/10.1111/1750-3841.14969.

[17]

L. Chen, X.C. Feng, Y.Y. Zhang, et al., Effects of ultrasonic processing on caspase-3, calpain expression and myofibrillar structure of chicken during post-mortem ageing, Food Chem. 177 (2015) 280-287. https://doi.org/10.1016/j.foodchem.2014.11.064.

[18]

L. Chen, Y.W. Chai, J. Luo, et al., Apoptotic changes and myofibrils degradation in post-mortem chicken muscles by ultrasonic processing, LWT-Food Sci. Technol. 142 (2021) 110985. https://doi.org/10.1016/j.lwt.2021.110985.

[19]

J.A. Martinez, Z. Zhang, S.I. Svetlov, et al., Calpain and caspase processing of caspase-12 contribute to the ER stress-induced cell death pathway in differentiated PC12 cells, Apoptosis 15 (2010) 1480-1493. https://doi.org/10.1007/s10495-010-0526-4.

[20]

A.M. Gorman, S.J.M. Healy, R. Jaeger, et al., Stress management at the ER: regulators of ER stress-induced apoptosis, Pharmacol. Ther. 134 (2012) 306-316. https://doi.org/10.1016/j.pharmthera.2012.02.003.

[21]

Y. Yang, L. Liu, I. Naik, et al., Transcription factor C/EBP homologous protein in health and diseases, Front. Immunol. 8 (2017) 1612. https://doi.org/10.3389/fimmu.2017.01612.

[22]

L. Wang, J.J. Li, S. Teng, et al., Changes in collagen properties and cathepsin activity of beef M. semitendinosus by the application of ultrasound during post-mortem aging, Meat Sci. 185 (2022) 108718. https://doi.org/10.1016/j.meatsci.2021.108718.

[23]

F. Yang, S.N. Jia, J.X. Liu, et al., The relationship between degradation of myofibrillar structural proteins and texture of superchilled grass carp (Ctenopharyngodon idella) fillet, Food Chem. 301 (2019) 125278. https://doi.org/10.1016/j.foodchem.2019.125278.

[24]

S.G. Cadena, L. Massieu, Caspases and their role in inflammation and ischemic neuronal death. Focus on caspase-12, Apoptosis 21 (2016) 763-777. https://doi.org/10.1007/s10495-016-1247-0.

[25]

C.M. Kemp, T. Parr, R.G. Bardsley, et al., Comparison of the relative expression of caspase isoforms in different porcine skeletal muscles, Meat Sci. 73 (2006) 426-431. https://doi.org/10.1016/j.meatsci.2005.12.009.

[26]

L. Zhao, T. Xing, J.C. Huang, et al., Involvement of μ/m-calpain in the proteolysis and meat quality changes during postmortem storage of chicken breast muscle, Anim. Sci. J. 89 (2018) 423-431. https://doi.org/10.1111/asj.12921.

[27]

X.L. Yan, R. Liu, C.Y. Zhang, et al., The postmortem: calpain activity, protein degradation and tenderness of sheep meat from Duolang and Hu breeds, Int. J. Food Sci. Technol. 53 (2018) 904-912. https://doi.org/10.1111/ijfs.13661.

[28]

F. Raynaud, E. Fernandez, G. Coulis, et al., Calpain 1-titin interactions concentrate calpain 1 in the Z-band edges and in the N2-line region within the skeletal myofibril, FEBS J. 272 (2005) 2578-2590. https://doi.org/10.1111/j.1742-4658.2005.04683.x.

[29]

F. Huang, M. Huang, H. Zhang, et al., Cleavage of the calpain inhibitor, calpastatin, during postmortem ageing of beef skeletal muscle, Food Chem. 148 (2014) 1-6. https://doi.org/10.1016/j.foodchem.2013.10.016.

[30]

L. Chen, X.C. Feng, F. Lu, et al., Effects of camptothecin, etoposide and Ca2+ on caspase-3 activity and myofibrillar disruption of chicken during postmortem ageing, Meat Sci. 87 (2011) 165-174. https://doi.org/10.1016/j.meatsci.2010.10.002.

[31]

L.L. Wang, G.Y. Ma, Y.B. Zhang, et al., Effect of mitochondrial cytochrome c release and its redox state on the mitochondrial-dependent apoptotic cascade reaction and tenderization of yak meat during postmortem aging, Food Res. Int. 111 (2018) 488-497. https://doi.org/10.1016/j.foodres.2018.05.049.

[32]

I. Tabas and D. Ron, Integrating the mechanisms of apoptosis induced by endoplasmic reticulum stress, Nat. Cell Biol. 13 (2011) 184-190. https://doi.org/10.1038/ncb0311-184.

[33]

J.P. Upton, L. Wang, D. Han, et al., IRE1 alpha cleaves select microRNAs during ER stress to derepress translation of proapoptotic caspase-2, Science 338 (2012) 818-822. https://doi.org/10.1126/science.1226191.

[34]

A.A. Tinnikov, H.H. Samuels, A novel cell lysis approach reveals that caspase-2 rapidly translocates from the nucleus to the cytoplasm in response to apoptotic stimuli, PLoS ONE 8 (2013) e61085. https://doi.org/10.1371/journal.pone.0061085.

[35]

C. Hetz, P. Bernasconi, J. Fisher, et al., Proapoptotic BAX and BAK modulate the unfolded protein response by a direct interaction with IRE1 alpha, Science 312 (2006) 572-576. https://doi.org/10.1126/science.1123480.

[36]

Y.C. Cheng, J.M. Chang, C.A. Chen, et al., Autophagy modulates endoplasmic reticulum stress-induced cell death in podocytes: a protective role, Exp. Biol. Med. 240 (2015) 467-476. https://doi.org/10.1177/1535370214553772.

[37]

J.T. Zhang, M. Yi, L.Y. Zha, et al., Sodium butyrate induces endoplasmic reticulum stress and autophagy in colorectal cells: implications for apoptosis, PLoS ONE 11 (2016) e0147218. https://doi.org/10.1371/journal.pone.0147218.

[38]

Y. Zhang, Y. Zhang, X.F. Jin, et al., The role of astragaloside Ⅳ against cerebral ischemia/reperfusion injury: suppression of apoptosis via promotion of P62-LC3-autophagy, Molecules 24 (2019) 1838. https://doi.org/10.3390/molecules24091838.

[39]

J. Wang, C. Garbutt, H. Ma, et al., Expression and role of autophagy-associated p62 (SQSTM1) in multidrug resistant ovarian cancer, Gynecol. Oncol. 150 (2018) 143-150. https://doi.org/10.1016/j.ygyno.2018.04.557.

[40]

Y. Potes, M. Olivan, A. Rubio-Gonzalez, et al., Pig cognitive bias affects the conversion of muscle into meat by antioxidant and autophagy mechanisms, Animal 11 (2017) 2027-2035. https://doi.org/10.1017/s1751731117000714.

[41]

M. Ogata, S.I. Hino, A. Saito, et al., Autophagy is activated for cell survival after endoplasmic reticulum stress, Mol. Cell. Biol. 26 (2006) 9220-9231. https://doi.org/10.1128/mcb.01453-06.

[42]

A. Margariti, H. Li, T. Chen, et al., XBP1 mRNA splicing triggers an autophagic response in endothelial cells through BECLIN-1 transcriptional activation, J. Biol. Chem. 288 (2013) 859-872. https://doi.org/10.1074/jbc.m112.412783.

[43]

Y.F. Wang, J.Y. Xie, H. Wang, et al., Beclin-1 suppresses gastric cancer progression by promoting apoptosis and reducing cell migration, Oncol. Lett. 14 (2017) 6857-6862. https://doi.org/10.3892/ol.2017.7046.

[44]

A. Bahrami, M. Khazaei, S. Shahidsales, et al., The Therapeutic potential of PI3K/Akt/mTOR inhibitors in breast cancer: rational and progress, J. Cell. Biochem. 119 (2018) 213-222. https://doi.org/10.1002/jcb.26136.

Food Science and Human Wellness
Pages 2141-2151
Cite this article:
Chai Y, Chen L, Xiang S, et al. Endoplasmic reticulum stress improved chicken tenderness, promoted apoptosis and autophagy during postmortem ageing. Food Science and Human Wellness, 2024, 13(4): 2141-2151. https://doi.org/10.26599/FSHW.2022.9250178

586

Views

55

Downloads

0

Crossref

0

Web of Science

0

Scopus

0

CSCD

Altmetrics

Received: 23 November 2022
Revised: 22 December 2022
Accepted: 24 January 2023
Published: 20 May 2024
© 2024 Beijing Academy of Food Sciences. Publishing services by Tsinghua University Press.

This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

Return