Hostname: page-component-848d4c4894-pftt2 Total loading time: 0 Render date: 2024-06-08T01:51:42.625Z Has data issue: false hasContentIssue false

Honey bee larval culture in vitro: gut emptying determines the transition from larva to prepupa and recombinant AccApidaecin improves antibacterial activity

Published online by Cambridge University Press:  28 February 2023

Wen-Feng Chen
Affiliation:
College of Animal Science and Technology, Shandong Agricultural University, Taian, Shandong Province, China
Hong-Fang Wang
Affiliation:
College of Animal Science and Technology, Shandong Agricultural University, Taian, Shandong Province, China
Ying Wang
Affiliation:
College of Animal Science and Technology, Shandong Agricultural University, Taian, Shandong Province, China
Zhen-Guo Liu
Affiliation:
College of Animal Science and Technology, Shandong Agricultural University, Taian, Shandong Province, China
Bao-Hua Xu*
Affiliation:
College of Animal Science and Technology, Shandong Agricultural University, Taian, Shandong Province, China
*
Author for correspondence: Bao-Hua Xu, Email: bhxu@sdau.edu.cn

Abstract

In vitro rearing of honey bee larvae is ideal for bioassay studies; no honey bee stable cell lines are available. Inconsistency of internal development staging of reared larvae and a susceptibility to contamination are common problems encountered. Standardized protocols on rearing larvae in vitro to make the larvae growth and development more similar to that of natural colonies are necessary to ensure the accuracy of experimental results and promote honey bee research as a model organism. Here, we concluded that when larval fasting weight was >160 mg, the time point of gut emptying can be defined as the critical point separating the larval and prepupal stages. In this way, we can conduct precise studies on the prepupal stage, such as organ remodeling during metamorphosis. Simultaneously, we further verified that recombinant AccApidaecin in genetic engineered bacteria added to the larval diet upregulated antibacterial peptide gene expression, and did not stimulate the stress response in larvae, nor did it affect the pupation rate or eclosion rate. This demonstrated that feeding recombinant AccApidaecin can enhance the individual antibacterial ability at the molecular level.

Type
Research Paper
Copyright
Copyright © The Author(s), 2023. Published by Cambridge University Press

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Al-Ghamdi, A, Ali Khan, K, Javed Ansari, M, Almasaudi, SB and Al-Kahtani, S (2018) Effect of gut bacterial isolates from Apis mellifera jemenitica on Paenibacillus larvae infected bee larvae. Saudi Journal of Biological Sciences 25, 383387.CrossRefGoogle ScholarPubMed
Al-Waili, N, Salom, K, Al-Ghamdi, A and Ansari, MJ (2012) Antibiotic, pesticide, and microbial contaminants of honey: human health hazards. The Scientific World Journal 2012, 930849.CrossRefGoogle ScholarPubMed
Amparyup, P, Charoensapsri, W and Tassanakajon, A (2013) Prophenoloxidase system and its role in shrimp immune responses against major pathogens. Fish & Shellfish Immunology 34, 9901001.CrossRefGoogle ScholarPubMed
Andres, AJ and Thummel, CS (1994) Methods for quantitative analysis of transcription in larvae and prepupae. Methods in Cell Biology 44, 565573.CrossRefGoogle ScholarPubMed
Balieira, KVB, Mazzo, M, Bizerra, PFV, Guimarães, ARDJS, Nicodemo, D and Mingatto, FE (2018) Imidacloprid-induced oxidative stress in honey bees and the antioxidant action of caffeine. Apidologie 49, 562572.10.1007/s13592-018-0583-1CrossRefGoogle Scholar
BenVau, LR and Nieh, JC (2017) Larval honey bees infected with Nosema ceranae have increased vitellogenin titers as young adults. Scientific Reports 7, 14144.CrossRefGoogle ScholarPubMed
Bertholf, LM (1925) The moults of the honey bee. Journal of Economic Entomology 18, 380384.CrossRefGoogle Scholar
Casteels-Josson, K, Zhang, W, Capaci, T, Casteels, P and Tempst, P (1994) Acute transcriptional response of the honeybee peptide-antibiotics gene repertoire and required post-translational conversion of the precursor structures. Journal of Biological Chemistry 269, 2856928575.10.1016/S0021-9258(19)61943-5CrossRefGoogle ScholarPubMed
Casteels, P, Ampe, C, Jacobs, F, Vaeck, M and Tempst, P (1989) Apidaecins: antibacterial peptides from honeybees. The EMBO Journal 8, 23872391.CrossRefGoogle ScholarPubMed
Casteels, P, Ampe, C, Riviere, L, Van Damme, J, Elicone, C, Fleming, M, Jacobs, F and Tempst, P (1990) Isolation and characterization of abaecin, a major antibacterial response peptide in the honeybee (Apis mellifera). European Journal of Biochemistry 187, 381386.CrossRefGoogle Scholar
Casteels, P, Ampe, C, Jacobs, F and Tempst, P (1993) Functional and chemical characterization of Hymenoptaecin, an antimicrobial peptide that is infection-inducible in the honey bee (Apis mellifera). Journal of Biological Chemistry 268, 70447054.CrossRefGoogle Scholar
Chan, QW, Melathopoulos, AP, Pernal, SF and Foster, LJ (2009) The innate immune and systemic response in honey bees to a bacterial pathogen, Paenibacillus larvae. BMC Genomics 10, 387.10.1186/1471-2164-10-387CrossRefGoogle ScholarPubMed
Chen, WF, Wang, HF, Liu, ZG, Zhang, WX and Xu, BH (2019) Recombinant expression and antimicrobial activity of Apidaecin in Apis cerana cerana. Scientia Agricultura Sinica 52, 767776 (in Chinese).Google Scholar
Chen, WF, Wang, Y, Zhang, WX, Liu, ZG, Xu, BH and Wang, HF (2021) Methionine as a methyl donor regulates caste differentiation in the European honey bee (Apis mellifera). Insect Science 28, 746756.10.1111/1744-7917.12788CrossRefGoogle ScholarPubMed
Christe, P, Oppliger, A, Bancalà, F, Castella, G and Chapuisat, M (2003) Evidence for collective medication in ants. Ecology Letters 6, 1922.10.1046/j.1461-0248.2003.00395.xCrossRefGoogle Scholar
da Cruz-Landim, C and Cavalcante, VM (2003) Ultrastructural and cytochemical aspects of metamorphosis in the midgut of Apis mellifera L. (Hymenoptera: Apidae: Apinae). Zoological Science 20, 10991107.CrossRefGoogle ScholarPubMed
DeGrandi-Hoffman, G and Chen, Y (2015) Nutrition, immunity and viral infections in honey bees. Current Opinion in Insect Science 10, 170176.CrossRefGoogle ScholarPubMed
Dobrovsky, TM (1951) Postembryonic changes in the digestive tract of the worker honey bee (Apis mellifera L.). Cornell University, pp. 3–47.Google Scholar
El-Seedi, HR, Ahmed, HR, El-Wahed, AAA, Saeed, A, Algethami, AF, Attia, NF, Guo, Z, Musharraf, SG, Khatib, A, Alsharif, SM, Naggar, YA, Khalifa, SAM and Wang, K (2022) Bee stressors from an immunological perspective and strategies to improve bee health. Veterinary Sciences 9, 199.CrossRefGoogle ScholarPubMed
El Chamy, L, Leclerc, V, Caldelari, I and Reichhart, JM (2008) Sensing of ‘danger signals’ and pathogen-associated molecular patterns defines binary signaling pathways ‘upstream’ of toll. Nature Immunology 9, 11651170.CrossRefGoogle ScholarPubMed
Fine, JD, Cox-Foster, DL and Mullin, CA (2017) An inert pesticide adjuvant synergizes viral pathogenicity and mortality in honey bee larvae. Scientific Reports 7, 40499.10.1038/srep40499CrossRefGoogle ScholarPubMed
Goncalves, WG, Fernandes, KM, Santana, WC, Martins, GF, Zanuncio, JC and Serrao, JE (2017) Post-embryonic changes in the hindgut of honeybee Apis mellifera workers: morphology, cuticle deposition, apoptosis, and cell proliferation. Developmental Biology 431, 194204.CrossRefGoogle ScholarPubMed
Guidugli, KR, Nascimento, AM, Amdam, GV, Barchuk, AR, Omholt, S, Simoes, ZL and Hartfelder, K (2005) Vitellogenin regulates hormonal dynamics in the worker caste of a eusocial insect. FEBS Letters 579, 49614965.CrossRefGoogle ScholarPubMed
Kaftanoglu, O, Linksvayer, TA and Page, RE (2015) Rearing honey bees (Apis mellifera L.) in vitro: effects of feeding intervals on survival and development. Journal of Apicultural Research 49, 311317.CrossRefGoogle Scholar
Kupke, J, Spaethe, J, Mueller, MJ, Rossler, W and Albert, S (2012) Molecular and biochemical characterization of the major royal jelly protein in bumblebees suggest a non-nutritive function. Insect Biochemistry and Molecular Biology 42, 647654.CrossRefGoogle ScholarPubMed
Li, G, Zhao, H, Liu, Z, Wang, H, Xu, B and Guo, X (2018) The wisdom of honeybee defenses against environmental stresses. Frontiers in Microbiology 9, 722.10.3389/fmicb.2018.00722CrossRefGoogle ScholarPubMed
Ligoxygakis, P, Pelte, N, Hoffmann, JA and Reichhart, JM (2002) Activation of Drosophila toll during fungal infection by a blood serine protease. Science (New York, N.Y.) 297, 114116.CrossRefGoogle ScholarPubMed
Lu, A, Zhang, Q, Zhang, J, Yang, B, Wu, K, Xie, W, Luan, YX and Ling, E (2014) Insect prophenoloxidase: the view beyond immunity. Frontiers in Physiology 5, 252.CrossRefGoogle ScholarPubMed
Ma, L, Wang, Y, Zhang, W, Wang, H, Liu, Z and Xu, B (2016) Alterations in protein and amino acid metabolism in honeybees (Apis mellifera) fed different l-leucine diets during the larval stage. Journal of Asia-Pacific Entomology 19, 769774.CrossRefGoogle Scholar
Martins, GF, Neves, CA, Campos, LA and Serrao, JE (2006) The regenerative cells during the metamorphosis in the midgut of bees. Micron 37, 161168.CrossRefGoogle ScholarPubMed
McAfee, A, Chapman, A, Iovinella, I, Gallagher-Kurtzke, Y, Collins, TF, Higo, H, Madilao, LL, Pelosi, P and Foster, LJ (2018) A death pheromone, oleic acid, triggers hygienic behavior in honey bees (Apis mellifera L.). Scientific Reports 8, 5719.CrossRefGoogle ScholarPubMed
McDermott, BJ, Millar, BC and Piper, HM (1993) Cardiovascular effects of neuropeptide Y: receptor interactions and cellular mechanisms. Cardiovascular Research 27, 893905.CrossRefGoogle ScholarPubMed
Millar, BC, Weis, T, Piper, HM, Weber, M, Borchard, U, Mcdermott, BJ and Balasubramaniam, A (1991) Positive and negative contractile effects of neuropeptide Y on ventricular cardiomyocytes. The American Journal of Physiology 261, H1727H1H33.Google ScholarPubMed
Myser, WC (1954) The larval and pupal development of the honey bee Apis mellifera Linnaeus. Annals of the Entomological Society of America 47, 683711.10.1093/aesa/47.4.683CrossRefGoogle Scholar
Reim, T and Scheiner, R (2014) Division of labour in honey bees: age- and task-related changes in the expression of octopamine receptor genes. Insect Molecular Biology 23, 833841.CrossRefGoogle ScholarPubMed
Rembold, HD, Kremer, JP and Ulrich, GM (1980) Characterization of postembryonic developmental stages of the female castes of the honey bee, Apis mellifera L. Apidologie 11, 2938.10.1051/apido:19800104CrossRefGoogle Scholar
Rolff, J, Johnston, PR and Reynolds, S (2019) Complete metamorphosis of insects. Philosophical Transactions of the Royal Society of London. Series B, Biological Sciences 374, 20190063.10.1098/rstb.2019.0063CrossRefGoogle ScholarPubMed
Rothenbuhler, WC and Thompso, WC (1964) Resistance to American foulbrood in honey bees. I. Differential survival of larvae of different genetic lines. Journal of Economic Entomology 49, 470475.CrossRefGoogle Scholar
Schmittgen, TD and Livak, KJ (2008) Analyzing real-time PCR data by the comparative C(T) method. Nature Protocols 3, 11011108.CrossRefGoogle ScholarPubMed
Sreelakshmi, P and Paul, A (2014) In vitro rearing of spotted pod borer, Maruca vitrata (Fabricius) (Lepidoptera: Pyralidae) on different diets. Research Journal of Agricultural Sciences 6, 102104.Google Scholar
Tsuruda, JM, Chakrabarti, P and Sagili, RR (2021) Honey bee nutrition. The veterinary clinics of North America. Food Animal Practice 37, 505519.10.1016/j.cvfa.2021.06.006CrossRefGoogle Scholar
vanEngelsdorp, D, Traynor, KS, Andree, M, Lichtenberg, EM, Chen, Y, Saegerman, C and Cox-Foster, DL (2017) Colony collapse disorder (CCD) and bee age impact honey bee pathophysiology. PLoS ONE 12, e0179535.CrossRefGoogle ScholarPubMed
Veloso, JA and Lourenço, AP (2013) Pollen diet for in vitro rearing of Africanized honey bee larvae, Apis mellifera (Hymenoptera: Apidae) = Dieta polínica para criação in vitro de larvas de abelhas melíferas Africanizadas, Apis mellifera (Hymenoptera: Apidae). Bioscience Journal 30, 288296.Google Scholar
Wang, Y, Ma, LT and Xu, BH (2015) Diversity in life history of queen and worker honey bees, Apis mellifera L. Journal of Asia-Pacific Entomology 18, 145149.CrossRefGoogle Scholar
Wu, YY, Zhou, T, Wang, Q, Dai, PL, Xu, SF, Jia, HR and Wang, X (2015) Programmed cell death in the honey bee (Apis mellifera) (Hymenoptera: Apidae) worker brain induced by imidacloprid. Journal of Economic Entomology 108, 14861494.CrossRefGoogle ScholarPubMed
Xu, P, Shi, M and Chen, XX (2009) Antimicrobial peptide evolution in the Asiatic honey bee Apis cerana. PLoS ONE 4, e4239.CrossRefGoogle ScholarPubMed
Zhang, G and Xu, B (2015) Effects of dietary calcium levels on development, haemolymph and antioxidant status of honey bee (Apis mellifera) larva reared in vitro. Journal of Apicultural Research 54, 4854.CrossRefGoogle Scholar

Chen et al. supplementary material

Movie 1

Download Chen et al. supplementary material(Video)
Video 23.3 MB