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 (3.3 MB)
Collect
Submit Manuscript AI Chat Paper
Show Outline
Outline
Show full outline
Hide outline
Outline
Show full outline
Hide outline
Research Article | Open Access

Integrating transcriptomic and proteomics revealed the response mechanism of red swamp crayfish (Procambarus clarkii) muscle under cold stress

Yuqing Lei1,§Ying Gao1,§Xuehong Li2,3Xiaoying Luo2,3Lan Wang3Wenjin Wu3Guangquan Xiong3Shang Chu2Shugang Li1( )
Engineering Research Center of Bio-Process, Ministry of Education, School of Food and Biological Engineering, Hefei University of Technology, Hefei 230601, China
Key Laboratory of Fermentation Engineering, Ministry of Education, School of Food and Biological Engineering, Hubei University of Technology, Wuhan 430068, China
Institute for Farm Products Processing and Nuclear-Agricultural Technology, Hubei Academy of Agricultural Science, Wuhan 430064, China

§ These authors contributed equally to this work.

Show Author Information

Graphical Abstract

Abstract

Related research findings indicated that the hardness of the tail meat from red swamp crayfish (Procambarus clarkii) increased when responding to cold stress during the transportation. However, the effect of low temperature on crayfish muscle was still at the phenotype level, there were few studies on the molecular mechanism of crayfish muscle response to cold stress. The effect of cold stress on the tail meat of crayfish during simulated transportation (control and low temperature stress for 12 h (LT_12), 24 h (LT_24) and 36 h (LT_36) at 4 ℃) were investigated by integrated transcriptome and proteomics. The results showed that the hardness of crayfish meat increased after cold stress. Gene ontology (GO) analysis showed that differentially expressed genes (DEGs) and differentially expressed proteins (DEPs) of crayfish coping with cold stress were mainly involved in metabolism and glycolysis. Kyoto Encyclopedia of Genes and Genomes (KEGG) metabolic analysis found that the metabolic response to cold stress included changes in amino acids such as valine and isoleucine. Low temperature activated glycolysis and amino acid metabolism pathway as well as peroxisome pathway to maintain body balance. The significant increase in the expression of cytoskeletal protein-actin related genes such as β-actin and ACT1 might cause the increase of muscle hardness under stress.

References

[1]

X. Dong, J. Wang, V. Raghavan, Impact of microwave processing on the secondary structure, in vitro protein digestibility and allergenicity of shrimp (Litopenaeus vannamei) proteins, Food Chem. 337 (2021) 127811. https://doi.org/10.1016/j.foodchem.2020.127811.

[2]

Y. Sun, M. Zhang, B. Bhandari, et al., Ultrasound treatment of frozen crayfish with chitosan nano-composite water-retaining agent: influence on cryopreservation and storage qualities, Food Res. Int. 126 (2019) 108670. https://doi.org/10.1016/j.foodres.2019.108670.

[3]

Z. F. Gu, H. J. Wei, F. Cheng, et al., Effects of air exposure time and temperature on physiological energetics and oxidative stress of winged pearl oyster (Pteria penguin), Aquac. Rep. 17 (2020) 100384. https://doi.org/10.1016/j.aqrep.2020.100384.

[4]

D. C. Behringer, E. Duermit-Moreau, Crustaceans, one health and the changing ocean, J. Invertebr. Pathol. 186 (2021) 107500. https://doi.org/10.1016/j.jip.2020.107500.

[5]

Z. L. Wang, J. Zhou, J. Y. Li, et al., The immune defense response of Pacific white shrimp (Litopenaeus vannamei) to temperature fluctuation, Fish Shellfish Immunol. 103 (2020) 103–110. https://doi.org/10.1016/j.fsi.2020.04.053.

[6]

L. F. Fan, A. L. Wang, Y. T. Miao, et al., Comparative proteomic identification of the hepatopancreas response to cold stress in white shrimp, Litopenaeus vannamei, Aquaculture 454 (2016) 27–34. https://doi.org/10.1016/j.aquaculture.2015.10.016.

[7]

X. H. Kong, G. Z. Wang, S. J. Li, Effects of low temperature acclimation on antioxidant defenses and ATPase activities in the muscle of mud crab (Scylla paramamosain), Aquaculture 370/371 (2012) 144–149. https://doi.org/10.1016/j.aquaculture.2012.10.012.

[8]

S. Y. Jin, L. Jacquin, F. Huang, et al., Optimizing reproductive performance and embryonic development of red swamp crayfish Procambarus clarkii by manipulating water temperature, Aquaculture 510 (2019) 32–42. https://doi.org/10.1016/j.aquaculture.2019.04.066.

[9]

T. Simčič, F. Pajk, M. Jaklič, et al., The thermal tolerance of crayfish could be estimated from respiratory electron transport system activity, J. Therm. Biol. 41 (2014) 21–30. https://doi.org/10.1016/j.jtherbio.2013.06.003.

[10]

T. Rőszer, The invertebrate midintestinal gland (“hepatopancreas”) is an evolutionary forerunner in the integration of immunity and metabolism, Cell Tissue Res. 358 (2014) 685–695. https://doi.org/10.1007/s00441-014-1985-7.

[11]

X. H. Li, S. G. Li, G. P. Shi, et al., Quantitative proteomics insights into gel properties changes of myofibrillar protein from Procambarus clarkii under cold stress, Food Chem. 372 (2022) 130935. https://doi.org/10.1016/j.foodchem.2021.130935.

[12]

X. Y. Ren, Z. X. Yu, Y. Xu, et al., Integrated transcriptomic and metabolomic responses in the hepatopancreas of kuruma shrimp (Marsupenaeus japonicus) under cold stress, Ecotoxicol. Environ. Saf. 206 (2020) 111360. https://doi.org/10.1016/j.ecoenv.2020.111360.

[13]

L. Luo, J. H. Huang, D. L. Liu, et al., Transcriptome reveals the important role of metabolic imbalances, immune disorders and apoptosis in the treatment of Procambarus clarkii at super high temperature, Comp. Biochem. Physiol. Part D: Genom. Proteom. 37 (2021) 100781. https://doi.org/10.1016/j.cbd.2020.100781.

[14]

L. J. Chen, J. Liu, G. Kaneko, et al., Quantitative phosphoproteomic analysis of soft and firm grass carp muscle, Food Chem. 303 (2020) 125367. https://doi.org/10.1016/j.foodchem.2019.125367.

[15]

X. Lu, S. Luan, P. Dai, et al., iTRAQ-based comparative proteome analysis for molecular mechanism of defense against acute ammonia toxicity in Pacific white shrimp Litopenaeus vannamei, Fish Shellfish Immunol. 74 (2018) 52–61. https://doi.org/10.1016/j.fsi.2017.12.030.

[16]

Y. M. Tao, L. Ma, D. D. Li, et al., Proteomics analysis to investigate the effect of oxidized protein on meat color and water holding capacity in Tan mutton under low temperature storage, LWT-Food Sci. Technol. 146 (2021) 111429. https://doi.org/10.1016/j.lwt.2021.111429.

[17]

W. Wu, X. P. You, W. Q. Sun, et al., Insight into acute heat stress on meat qualities of rainbow trout (Oncorhynchus mykiss) during short-time transportation, Aquaculture 543 (2021) 737013. https://doi.org/10.1016/j.aquaculture.2021.737013.

[18]

H. L. Fan, D. M. Fan, J. L. Huang, et al., Cooking evaluation of crayfish (Procambarus clarkia) subjected to microwave and conduction heating: a visualized strategy to understand the heat-induced quality changes of food, Innov. Food Sci. Emerg. 62 (2020) 102368. https://doi.org/10.1016/j.ifset.2020.102368.

[19]

J. Qin, F. Gu, D. Liu, et al., Proteomic analysis of elite soybean Jidou17 and its parents using iTRAQ-based quantitative approaches, Proteome Sci. 11 (2013) 12. https://doi.org/10.1186/1477-5956-11-12.

[20]

Q. X. Sun, B. H. Kong, S. C. Liu, et al., Ultrasound-assisted thawing accelerates the thawing of common carp (Cyprinus carpio) and improves its muscle quality, LWT-Food Sci. Technol. 141 (2021) 111080. https://doi.org/10.1016/j.lwt.2021.111080.

[21]
W. Huang, C. H. Ren, H. M. Li, et al., Transcriptomic analyses on muscle tissues of Litopenaeus vannamei provide the first profile insight into the response to low temperature stress, PLoS ONE 12 (2017) e0178604. https://doi.org/10.1371/journal.pone.0178604.
[22]

I. M. Sokolova, M. Frederich, R. Bagwe, et al., Energy homeostasis as an integrative tool for assessing limits of environmental stress tolerance in aquatic invertebrates, Mar. Environ. Res. 79 (2012) 1–15. https://doi.org/10.1016/j.marenvres.2012.04.003.

[23]

J. Bao, X. H. Wang, C. C. Feng, et al., Trehalose metabolism in the Chinese mitten crab Eriocheir sinensis: molecular cloning of trehalase and its expression during temperature stress, Aquac. Rep. 20 (2021) 100770. https://doi.org/10.1016/j.aqrep.2021.100770.

[24]

S. L. Hsieh, S. M. Chen, Y. H. Yang, et al., Involvement of norepinephrine in the hyperglycemic responses of the freshwater giant prawn, Macrobrachium rosenbergii, under cold shock, Comp. Biochem. Physiol. Part A: Mol. Integr. Physiol. 143 (2006) 254–263. https://doi.org/10.1016/j.cbpa.2005.12.009.

[25]

B. Shi, X. Y. Tao, M. B. Betancor, et al., Dietary chromium modulates glucose homeostasis and induces oxidative stress in Pacific white shrimp (Litopenaeus vannamei), Aquat. Toxicol. 240 (2021) 105967. https://doi.org/10.1016/j.aquatox.2021.105967.

[26]

D. Adinarayana, S. Kishore, Alterations in the carbohydrate metabolism during deltamethrin-induced toxicity in Ciprinus carpio, Int. J. Pharm. Life Sci. 5 (2014) 3311–3317. https://doi.org/10.1016/ijplsjournal.2014.109727.

[27]

X. Y. Ren, Y. Xu, Y. B. Zhang, et al., Comparative accumulation and transcriptomic analysis of juvenile Marsupenaeus japonicus under cadmium or copper exposure, Chemosphere 249 (2020) 126157. https://doi.org/10.1016/j.chemosphere.2020.126157.

[28]

K. G. Holden, E. J. Gangloff, E. Gomez-Mancillas, et al., Surviving winter: physiological regulation of energy balance in a temperate ectotherm entering and exiting brumation, Gen. Comp. Endocr. 307 (2021) 113758. https://doi.org/10.1016/j.ygcen.2021.113758.

[29]

I. Jerez-Cepa, M. Fernandez-Castro, T. J. Del Santo O’Neill, et al., Transport and recovery of gilthead seabream (Sparus aurata L.) sedated with clove oil and MS-222: effects on stress axis regulation and intermediary metabolism, Front. Physiol. 10 (2019) 612. https://doi.org/10.3389/fphys.2019.00612.

[30]

D. L. Wu, Y. H. Huang, Q. Chen, et al., Effects and transcriptional responses in the hepatopancreas of red claw crayfish Cherax quadricarinatus under cold stress, J. Therm. Biol. 85 (2019) 102404. https://doi.org/10.1016/j.jtherbio.2019.102404.

[31]

S. H. Zhang, X. F. Zeng, M. Ren, et al., Novel metabolic and physiological functions of branched chain amino acids: a review, J. Anim. Sci. Biotechnol. 8 (2017) 10. https://doi.org/10.1186/s40104-016-0139-z.

[32]

H. A. MacMillan, J. M. Knee, A. B. Dennis, et al., Cold acclimation wholly reorganizes the Drosophila melanogaster transcriptome and metabolome, Sci. Rep. 6 (2016) 28999. https://doi.org/10.1038/srep28999.

[33]

P. Segges, S. Corrêa, B. D. Rocher, et al., Targeting hodgkin and reed-sternberg cells with an inhibitor of heat-shock protein 90: molecular pathways of response and potential mechanisms of resistance, Int. J. Mol. Sci. 19 (2018) 836. https://doi.org/10.3390/ijms19030836.

[34]

Y. P. Sun, R. J. Deng, K. X. Zhang, et al., Single-cell study of the extracellular matrix effect on cell growth by in situ imaging of gene expression, Chem. Sci. 8 (2017) 8019–8024. https://doi.org/10.1039/c7sc03880a.

[35]

H. L. R. Gomez, J. P. Peralta, L. A. Tejano, et al., In silico and in vitro assessment of Portuguese oyster (Crassostrea angulata) proteins as precursor of bioactive peptides, Int. J. Mol. Sci. 20 (2019) 5191. https://doi.org/10.3390/ijms20205191.

[36]

J. Xiao, Q. Y. Liu, J. H. Du, et al., Integrated analysis of physiological, transcriptomic and metabolomic responses and tolerance mechanism of nitrite exposure in Litopenaeus vannamei, Sci. Total Environ. 711 (2020) 134416. https://doi.org/10.1016/j.scitotenv.2019.134416.

[37]

M. Kim, R. M. Robich, J. P. Rinehart, et al., Upregulation of two actin genes and redistribution of actin during diapause and cold stress in the northern house mosquito, Culex pipiens, J. Insect Physiol. 52 (2006) 1226–1233. https://doi.org/10.1016/j.jinsphys.2006.09.007.

[38]

P. E. Purdue, P. B. Lazarow, Peroxisome biogenesis, Annu. Rev. Cell Dev. Bi. 17 (2001) 701–752. https://doi.org/10.1146/annurev.cellbio.17.1.701.

[39]

T. A. Rodrigues, I. S. Alencastre, T. Francisco, et al., A PEX7-centered perspective on the peroxisomal targeting signal type 2-mediated protein import pathway, Mol. Cell. Biol. 34 (2014) 2917–2928. https://doi.org/10.1128/mcb.01727-13.

[40]

T. Finkel, N. J. Holbrook, Oxidants, oxidative stress and the biology of ageing, Nature 408 (2000) 239–247. https://doi.org/10.1038/35041687.

[41]

S. Krick, S. L. Shi, W. J. Ju, et al., Mpv17l protects against mitochondrial oxidative stress and apoptosis by activation of Omi/HtrA2 protease, PNAS 105 (2008) 14106–14111. https://doi.org/10.1073/pnas.0801146105.

Food Science of Animal Products
Article number: 9240007
Cite this article:
Lei Y, Gao Y, Li X, et al. Integrating transcriptomic and proteomics revealed the response mechanism of red swamp crayfish (Procambarus clarkii) muscle under cold stress. Food Science of Animal Products, 2023, 1(1): 9240007. https://doi.org/10.26599/FSAP.2023.9240007

1571

Views

228

Downloads

0

Crossref

Altmetrics

Received: 11 February 2023
Revised: 23 February 2023
Accepted: 06 March 2023
Published: 10 April 2023
© Beijing Academy of Food Sciences 2023.

Food Science of Animal Products published 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