Next Article in Journal
Effect of Salinity on Stomatal Conductance, Leaf Hydraulic Conductance, HvPIP2 Aquaporin, and Abscisic Acid Abundance in Barley Leaf Cells
Next Article in Special Issue
Transcriptomic and Bioinformatic Analyses Identifying a Central Mif-Cop9-Nf-kB Signaling Network in Innate Immunity Response of Ciona robusta
Previous Article in Journal
Genome-Scale Identification, Classification, and Expression Profiling of MYB Transcription Factor Genes in Cinnamomum camphora
Previous Article in Special Issue
Peritrophin-like Genes Are Associated with Delousing Drug Response and Sensitivity in the Sea Louse Caligus rogercresseyi
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Current Knowledge on Bee Innate Immunity Based on Genomics and Transcriptomics

1
College of Engineering, Hebei Normal University, Shijiazhuang 050024, China
2
Key Laboratory for Insect-Pollinator Biology of the Ministry of Agriculture and Rural Affairs, Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing 100093, China
*
Author to whom correspondence should be addressed.
Current address: College of Animal Sciences, Shanxi Agricultural University, Taigu, Jinzhong 030801, China.
Int. J. Mol. Sci. 2022, 23(22), 14278; https://doi.org/10.3390/ijms232214278
Submission received: 29 September 2022 / Revised: 28 October 2022 / Accepted: 14 November 2022 / Published: 18 November 2022
(This article belongs to the Special Issue Transcriptomic and Genomic Insights into Invertebrates)

Abstract

:
As important pollinators, bees play a critical role in maintaining the balance of the ecosystem and improving the yield and quality of crops. However, in recent years, the bee population has significantly declined due to various pathogens and environmental stressors including viruses, bacteria, parasites, and increased pesticide application. The above threats trigger or suppress the innate immunity of bees, their only immune defense system, which is essential to maintaining individual health and that of the colony. In addition, bees can be divided into solitary and eusocial bees based on their life traits, and eusocial bees possess special social immunities, such as grooming behavior, which cooperate with innate immunity to maintain the health of the colony. The omics approach gives us an opportunity to recognize the distinctive innate immunity of bees. In this regard, we summarize innate bee immunity from a genomic and transcriptomic perspective. The genetic characteristics of innate immunity were revealed by the multiple genomes of bees with different kinds of sociality, including honeybees, bumblebees, wasps, leaf-cutter bees, and so on. Further substantial transcriptomic data of different tissues from diverse bees directly present the activation or suppression of immune genes under the infestation of pathogens or toxicity of pesticides.

1. Introduction

While the demand for crop pollination by insects has tripled over the past 50 years, the pollinator-bee population has drastically declined due to climate change, habitat loss, emerging parasites and pathogens, and increased pesticide application [1,2,3,4,5,6]. Due to these challenges and environmental stresses such as poor nutrition and pesticide residues, pollinator bees use their innate immune system, which is their only defense, to maintain their individual health and that of the colony. The emergence and development of genomic and transcriptomic technology provide an opportunity to understand the mysteries of life sciences [7,8]. Similarly, genomic and transcriptomic research on bees have helped identify and understand the genetic traits of immunity and the immune response to environmental stressors and pathogens, both primordial aspects of the colony’s health [9,10]. This review focuses on the genetic characteristics of innate immunity and immune response to pathogens and pesticides in bees from a genomic and transcriptomic perspective (Figure 1). The present review outlines the uniqueness of innate bee immunity and immune genes in response to single or multiple threats to facilitate intensive study of the bee immune system.

2. Genomic Perspective of Innate Bee Immunity

The first complete bee genome, the Apis mellifera genome, was assembled and annotated in 2006 [11], and it was updated in 2014 and 2016 [12,13]. Compared to known genomes of model organisms such as Drosophila melanogaster and Anopheles gambiae, the A. mellifera genome encodes fewer immune proteins involved in the immune response process, starting from pathogen recognition to immune effectors. In fact, nearly two-thirds of the immune genes are reduced, but a small number of genes encode the components of the insect’s classical immune pathways, such as Toll, IMD, and JAK/STAT pathways [11,12,14]. Based on the genomic and transcriptomic analyses, Apis cerana, a species similar to A. mellifera, also possesses a small amount of innate immune genes and similar classical immune pathways compared to those of flies and mosquitoes, and most of its immune genes are similar to those of A. mellifera [15,16]. Recently, de novo genome assembly of Chinese plateau A. cerana has shown that the gene number of this genome is different from that of known A. cerana genomes [17]. As a representative of primary eusocial bees, bumblebee genomes from 17 species show that the major immune repertoire and immune gene number are both similar to those of A. mellifera, which is significantly lower compared with that of Dipteran models [18,19]. Moreover, another important Asian honeybee (Apis dorsata) genome also exhibits an immune repertoire similar to that of known bee genomes [20]. A reduced number of immune proteins might be seen as a result of the social immunity of social bees; eusocial and primary eusocial bees can cooperate to reduce disease transmission risk through their behavior, known as social immunity, which can be prophylactic or activated on demand [21]. However, expressed sequence tag (EST) databases of healthy and pathogen-challenged alfalfa leaf-cutting bee larvae have identified 104 putative immunity-related genes, including innate immune response genes that are highly conserved with honey bee genes, such as those involved in pathogen recognition, phagocytosis, prophenoloxidase cascade, melanization, coagulation, and several signaling pathways [22]. Similar smaller immune repertoires have been discovered in other available solitary or eusocial bee genomes, including those of A. florea, Bombus terrestris, B. impatiens, Eufriesea Mexicana, Melipona quadrifasciata, Habropoda laboriosa, Megachile rotundata, Lasioglossum albipes, and Dufourea novaeangliae [19,23]. Additionally, genomes of three parasitoid Nasonia species (N. vitripennis, N. giraulti, and N. longicornis) show an immune repertoire similar to that of A. mellifera but with a slightly higher gene count than that of the latter, although several immune genes are not yet identified [24]. Furthermore, a fig wasp (Ceratosolen solmsi) genome exhibits an immune repertoire and gene counts similar to those of A. mellifera [25]. Therefore, although different bee species possess slightly different immune gene counts, their innate immune system is characterized by integral immune pathways, and the reduced immune gene number is interestingly not related to the bees’ sociality [26]. Thus, genomic analysis is a powerful tool for exploring the innate immune components of both solitary and eusocial bees. Until now, several genomes of different bees have been determined, but the immune repertoire of these bees has to be further analyzed [27,28,29,30,31,32,33,34,35,36,37,38,39].

3. Transcriptomic Perspective of Innate Bee Immune Response

Transcriptomic analysis in bees indicates gene expression changes under certain conditions. For instance, the expression profile of the immune genes is mainly influenced by invasion by pathogens, such as viruses, bacteria, and parasites, as well as exposure to pesticides and other hazardous substances, as well as poor nutrition [40]. While nutrient status is key to an individual’s immune response, the relationship between nutrition and innate immunity is driven by energy consumption [41]. Pathogens adversely affect the health of wild and managed bees [42], and their infestation can trigger the innate immune response, thus blocking the infection and eliminating the pathogens [43]. In contrast, pesticides inhibit the innate immune response and promote pathogen spread and virulence, contributing to bee colony loss [3]. Hazardous substances such as nano- and micro-polystyrene plastics can disturb gut microbiota and inhibit intestinal immune response [44]. Regardless of the suppression or triggering of immune gene expression, transcriptomic analysis can directly reveal gene expression profile changes in different tissues of managed and wild bees infected by various pathogens and exposed to pesticides.

3.1. Immune Responses to Viruses

Bees can be infected by more than 20 viruses worldwide, most commonly by deformed wing virus (DWV), black queen cell virus (BQCV), Israeli acute paralysis virus (IAPV), and Sacbrood virus (SBV) [45,46,47]. Following an IAPV infection in A. mellifera, two immune genes involved in RNAi pathways Ago2 and Dicer, as well as other immune genes, were identified to be implicated in Toll and JAK/STAT pathways, and these findings overlap with those on immune gene response following other viral infections based on transcriptome analysis [48]. This analysis also demonstrated the dynamic changes in immune gene expression in the hours following an IAPV infection [49], and it has shown that BQCV infection triggers significant upregulation of immune genes such as those encoding antimicrobial peptides (abaecin, apidaecin, and hymenoptaecin), peptidoglycan recognition protein S2 (PGRP-S2), Ago2, and Dicer, (the latter two both implicated in RNAi pathways in A. mellifera brains [50]). A transcriptomic analysis of larvae and pupae has revealed changes in immune genes involved in antimicrobial peptides (AMPs) and melanization pathways following DWV and SBV infection in A. mellifera; both are positive-strand RNA viruses and members of the iflavirus group [51]. In SBV-carrying A. mellifera larvae, approximately 20 differentially expressed immune-related genes have been identified [52]. In A. cerana larvae naturally infected with CSBV, small interfering RNA-targeting serine proteases that are involved in the immune response are upregulated [53]. Moreover, transcriptomic analysis has revealed that the sirtuin signaling pathway may be a novel mechanism of immune response to CSBV infection in honeybees [54] and that the immune genes for AMPs, Ago2, and Dicer are involved in the innate immune response to DWV infection in A. mellifera brains [55]. The transcriptome profile of A. mellifera eggs shows the trans-generational effects of SBV and DWV on several gene expression levels, indicating the different virulence of DWV and SBV during vertical transmission [56]. Bee viruses can be transmitted by Varroa destructor mites, which drives changes in virus distribution, prevalence, and virulence [57]. Transcriptomic analysis shows that varroa-induced viral replication is closely related to the expression of immune genes PGRP-S2, NimC2, and Eater-like as well as serine protease levels in A. mellifera adults [58]. Furthermore, transcriptomic analysis revealed that the Varroa mite alone and the DWV coupled with the mite could induce upregulation of different immune genes involved in the Toll and JNK pathways, respectively [59]. In addition, multiple transcriptome data have shown that hymenoptaecin, defensin-2, PGRP-S1, and B-gluc1 are common host immune genes that respond to the major pathogens and parasites such as RNA viruses, V. destructor, N. apis, and N. ceranae in A. mellifera [60]. Meanwhile, despite the fact that some common genes are identified above, important differences in the transcription responses of honey bees to various pathogens were revealed [60].

3.2. Immune Response to Parasites

Along with acting as a virus vector, the parasitic Varroa destructor also reduces nutrient levels and suppresses individual immune function, and is an underestimated parasite threatening the health of bee colonies [41,61]. Transcriptomic analysis has shown that immune gene expression levels change as a response to the mite V. destructor (e.g., PGRP-S3, GNBP1, Toll receptors, and serine protease) [62]. Updated transcriptomic analysis of newly emerged A. mellifera has identified three immune genes encoding PGRP-2, hymenoptaecin, and glucan recognition protein, which could be good candidates as markers for immune response to Varroa infestation [63]. Moreover, Varroa parasitism could also cause downregulation of autophagic-specific gene 18 and poly (U) binding factor 68 Kd (pUf68), and Rab7 upregulation in A. mellifera [64]. A set of genes related to social immunity has been identified in A. mellifera by analyzing the comparative transcriptome of varroa-hygienic bees [65]. Nutrigenomics shows that pollen and sugar supplements positively affect the production of some AMPs but cannot reverse the harmful effects of varroa parasitism [66].
Additionally, based on the transcriptomic data, the expression of immune genes encoding serine protease, lysozyme 1, and hymenoptaecin is found to be suppressed by Nosema ceranae infection in A. mellifera [67]. Serine proteases, peptidoglycan recognition proteins, and antimicrobial peptides are downregulated following N. ceranae infection in A. mellifera [68]. Besides the differently expressed immune genes, the whole transcriptome has also identified the N. ceranae infection-related long non-coding RNAs (lncRNAs) that may participate in the A. mellifera immune response [69]. Comparative transcriptome analysis has identified the genes involved in cellular immune pathways, such as ubiquitin-mediated proteolysis, endocytosis, lysosomes, phagosomes, autophagy, and melanogenesis, and in humoral immune pathways, such as MAPK, JAK/STAT, and Toll/IMD signaling pathways, in N. ceranae-infected A. cerana [70]. Moreover, transcriptome analysis has identified CircRNAs targeting mRNAs that were annotated to cellular immunity pathways, including endocytosis, lysosomes, and phagosomes in the gut of N. ceranae-infested A. cerana [71]. Except for honeybee, many genes, including those encoding receptors (GNBPs), signaling pathway components, and AMPs, have been identified in Bombus terrestris infested by Crithidia bombi, and these genes are closely related to canonical immune pathways [72]. Transcriptomic analysis of Sphaerularia bombi-infected B. terrestris queens during and after diapause showed that increased expression of immune genes (e.g., genes encoding scavenger receptors, Toll-like receptors, domeless, C-type lectin, and draper) is mainly induced by S. bombi after diapause [73]. Interestingly, the transcriptome has been used to evaluate the role of pathogens and pesticides in reducing the B. terricola population by detecting immune and detoxification genes [74].

3.3. Immune Response to Bacteria

Genome microarrays demonstrate that immunostimulants such as bacterial infection and wounds could induce hundreds of significantly differentially expressed genes, including the previously identified canonical immune genes and other major unidentified new genes [75]. Transcriptomic analysis showed that the expression levels of hymenoptaecin, apidaecin, and defensin-1 are significantly upregulated in A. mellifera larvae infested with the bacterial pathogen Paenibacillus [76]. Transcriptome profiling has revealed an upregulation of immune-related genes, such as those encoding Toll-like receptors, integrin, and antimicrobial peptides, in Ascosphaera apis-infected A. mellifera larvae [77]. Moreover, 13 differently expressed immune genes involved in humoral and cellular immunity were identified in the A. mellifera gut following A. apis infestation [78]. Transcriptomic analysis of the A. cerana larval gut showed upregulation of immune genes such as humoral and cellular immune genes following A. apis infestation [79]. In addition to the pathogenic bacteria, the beneficial gut microbe Frischella perrara can strongly activate the host immune response and upregulate important immune genes, including those encoding pattern-recognition receptors, antimicrobial peptides, transporter genes, and melanization cascade in A. mellifera [80]. Interestingly, transcriptome analysis has shown that the gut microbe Lactobacillus apis triggers the expression of PGRP-S3, Spätzle, and antibacterial proteins, which can inhibit infection by Hafnia alvei; further genomic analysis suggested that the S-layer proteins of L. apis are potentially involved in honeybee Toll signaling and in the activation of antibacterial protein production in honeybees [81]. The gut microbiota can be altered by polystyrene microplastic exposure and might influence the expression of gut immune genes; for instance, it can cause an upregulation of apidaecin and abaecin and dose-dependent downregulation of domeless, hopscotch, and symplekin in A. mellifera [82]. Another A. mellifera gut transcriptome has shown that microplastic polystyrene ingestion triggers upregulation of PGRP-S3, defensin-2, and dose-dependent differently expressed genes encoding Toll-like receptors, PGRP-S2, defensin-1, hymenoptaecin, and apidemins [83]. Additionally, as a parasitoid wasp differing from honeybees [84], transcriptomic analysis suggests that Nasonia vitripennis may possess novel immune components against bacterial infection [85]. As an important model system, the transcriptome of N. vitripennis will contribute to our comprehensive understanding of innate bee immunity.

3.4. Immune Suppression Due to Pesticides

Based on a transcriptomic analysis, five differently expressed immune genes encoding hymenoptaecin, abaecin, apidaecin, apisimin, and lysozyme are found in A. mellifera larvae exposed to sublethal levels of imidacloprid [86]. Another transcriptome analysis has identified immune-related genes (abaecin, eater, hymenoptaecin, defensin1, defensin2, vitellogenin, and apidaecin) involved in the immune response against neonicotinoids such as imidacloprid and clothianidin in honeybees; it has also shown that abaecin and hymenoptaecin expression levels are significantly higher in neonicotinoid-exposed A. cerana than in neonicotinoid-exposed A. mellifera [87]. Moreover, a transcriptomic analysis also demonstrated that imidacloprid could alter the innate immune gene expression of brain tissue in the bumblebee B. terrestris [88]. Meanwhile, following exposure to sublethal doses of imidacloprid and deltamethrin, detoxification genes are upregulated, and immune genes encoding apidaecin and hymenoptaecin are significantly downregulated in the brain tissue of A. mellifera [89]. Another transcriptomic analysis has indicated that environmentally relevant concentrations of the neonicotinoid clothianidin can induce the downregulation of scavenger receptor class B member 1 and upregulation of hymenoptaecin and apidaecin, while imidacloprid can cause hymenoptaecin upregulation in the brain tissue of A. mellifera [90]. Chronic oral exposure to the neonicotinoid clothianidin may alter the expression of immune defense-related genes by upregulating exosome complex component RRP46 and downregulating C-Maf-inducing protein-like in worker bees but not in male B. impatiens [91]. Moreover, immune gene expression following V. destructor mite infestation differs from that following exposure to the neonicotinoid insecticide clothianidin in a single A. mellifera colony [92].
Transcriptome analysis has also helped identify the impacts of various pesticides on bee immunity. For instance, immune genes encoding defensin1, vitellogenin, and scavenger receptor class B member 1 are shown to be downregulated in thiamethoxam-treated A. mellifera brain tissues [90,93], while innate immunity-related proteins like apolipophorin-III-like proteins are significantly upregulated in the brains of A. mellifera exposed to environmental concentrations of the neonicotinoid thiacloprid [94]. Additionally, dinotefuran treatment significantly affects the expression of immune-related genes such as those encoding glutathione S-transferase S4, prolactin-releasing peptide receptor, defensin 2 (Def2), and vesicle-associated membrane protein 2 in the brains of A. mellifera [95]. The immune genes and expression levels of that in response to four insecticides, namely chlorpyrifos, malathion, cypermethrin, and chlorantraniliprole, are different but the differently expressed immune genes are all involved in the IMD pathway and production of AMPs in A. mellifera [96]. Transcriptome analysis of midguts also identified differentially expressed genes involved in immunity in nitenpyram-treated A. mellifera [97]. Dimethoate, an insecticide, can cause apisimin and Toll downregulation, and flupyradifurone or chlorantraniliprole may induce defensin1 and processing enzyme downregulation in the larvae of A. mellifera [98]. Differently expressed immune genes are identified in A. mellifera under benomyl stress [99]. Transcriptomic analysis has revealed that several immune genes such as those encoding abaecin, Def1, SP28, Toll-1, Toll-6, Toll-8, Toll-10, and MyD88 are upregulated in benomyl-treated A. mellifera [100]. Additionally, pesticides (acaricides) used to treat varroa mites in bee colonies can also induce Dscam downregulation, an immune gene important to cellular immunity, and basket downregulation, an orthologue of JNK signaling, in A. mellifera [101]. Interestingly, transcriptomic analysis of different tissues suggests that AMPs (e.g., apisimin and defensin) are simultaneously expressed with nectar processing enzymes in the hypopharyngeal and mandibular glands of foragers but not in the nurses of A. mellifera as a response to potential environmental threats during nectar and pollen collection [102].

4. Conclusions

Advances in genomic and transcriptomic analyses permit recognizing the fundamental genetic characteristics of bees and help understanding of gene expression changes as part of the response to various pathogens and/or internal or external environmental stressors. Multiple bee genomes have revealed that a small number of immune genes are involved in classical insect immune pathways in the bee’s innate immunity system. Based on genomes, the transcriptomic analysis has revealed some of the immune genes acting as a response to various pathogens such as viruses, bacteria, and parasites; these genes are suppressed by hazardous pesticides. However, more in-depth studies are needed to identify more immune genes critical to the immune response against threatening factors and maintain the bee colony’s health. Indeed, recognizing these immune genes provides a basis for the subsequent elaboration of the function and structure of these genes by other molecular biological methods. Moreover, further research is needed for a more comprehensive understanding of innate bee immunity. Future genomic analysis of different species and transcriptomic analysis of different tissues following various internal and/or external environment stressors could help identify related immune genes.

Author Contributions

Conceptualization, X.Z. and Y.L.; writing—original draft preparation, X.Z. and Y.L.; writing—review and editing, X.Z. and Y.L.; supervision, Y.L.; funding acquisition, Y.L. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the National Natural Science Foundation of China, grant number 32102606.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Goulson, D.; Nicholls, E.; Botías, C.; Rotheray, E.L. Bee declines driven by combined stress from parasites, pesticides, and lack of flowers. Science 2015, 347, 1255957. [Google Scholar] [CrossRef] [PubMed]
  2. Hristov, P.; Shumkova, R.; Palova, N.; Neov, B. Factors Associated with Honey Bee Colony Losses: A Mini-Review. Vet. Sci. 2020, 7, 166. [Google Scholar] [CrossRef] [PubMed]
  3. Sánchez-Bayo, F.; Goulson, D.; Pennacchio, F.; Nazzi, F.; Goka, K.; Desneux, N. Are bee diseases linked to pesticides?—A brief review. Environ. Int. 2016, 89–90, 7–11. [Google Scholar] [CrossRef]
  4. Cameron, S.A.; Sadd, B.M. Global Trends in Bumble Bee Health. Annu. Rev. Entomol. 2020, 65, 209–232. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Kluser, S.; Peduzzi, P. Global Pollinator Decline: A Literature Review; UNEP/GRIDEurope: Geneva, Switzerland, 2007; Volume 8, 10p. [Google Scholar]
  6. LeBuhn, G.; Vargas Luna, J. Pollinator decline: What do we know about the drivers of solitary bee declines? Curr. Opin. Insect Sci. 2021, 46, 106–111. [Google Scholar] [CrossRef] [PubMed]
  7. Pareek, C.S.; Smoczynski, R.; Tretyn, A. Sequencing technologies and genome sequencing. J. Appl. Genet. 2011, 52, 413–435. [Google Scholar] [CrossRef] [Green Version]
  8. Martin, J.A.; Wang, Z. Next-generation transcriptome assembly. Nat. Rev. Genet. 2011, 12, 671–682. [Google Scholar] [CrossRef]
  9. Branstetter, M.G.; Childers, A.K.; Cox-Foster, D.; Hopper, K.R.; Kapheim, K.M.; Toth, A.L.; Worley, K.C. Genomes of the Hymenoptera. Curr. Opin. Insect Sci. 2018, 25, 65–75. [Google Scholar] [CrossRef]
  10. Grozinger, C.M.; Robinson, G.E. The power and promise of applying genomics to honey bee health. Curr. Opin. Insect Sci. 2015, 10, 124–132. [Google Scholar] [CrossRef] [Green Version]
  11. Consortium, H.G.S. Insights into social insects from the genome of the honeybee Apis mellifera. Nature 2006, 443, 931–949. [Google Scholar] [CrossRef]
  12. Elsik, C.G.; Worley, K.C.; Bennett, A.K.; Beye, M.; Camara, F.; Childers, C.P.; de Graaf, D.C.; Debyser, G.; Deng, J.; Devreese, B. Finding the missing honey bee genes: Lessons learned from a genome upgrade. BMC Genom. 2014, 15, 86. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. McAfee, A.; Harpur, B.A.; Michaud, S.; Beavis, R.C.; Kent, C.F.; Zayed, A.; Foster, L.J. Toward an Upgraded Honey Bee (Apis mellifera L.) Genome Annotation Using Proteogenomics. J. Proteome Res. 2016, 15, 411–421. [Google Scholar] [CrossRef] [PubMed]
  14. Evans, J.; Aronstein, K.; Chen, Y.P.; Hetru, C.; Imler, J.L.; Jiang, H.; Kanost, M.; Thompson, G.; Zou, Z.; Hultmark, D. Immune pathways and defence mechanisms in honey bees Apis mellifera. Insect Mol. Biol. 2006, 15, 645–656. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Park, D.; Jung, J.W.; Choi, B.-S.; Jayakodi, M.; Lee, J.; Lim, J.; Yu, Y.; Choi, Y.-S.; Lee, M.-L.; Park, Y. Uncovering the novel characteristics of Asian honey bee, Apis cerana, by whole genome sequencing. BMC Genom. 2015, 16, 1. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Diao, Q.; Sun, L.; Zheng, H.; Zeng, Z.; Wang, S.; Xu, S.; Zheng, H.; Chen, Y.; Shi, Y.; Wang, Y.; et al. Genomic and transcriptomic analysis of the Asian honeybee Apis cerana provides novel insights into honeybee biology. Sci. Rep. 2018, 8, 822. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Lan, L.; Shi, P.; Song, H.; Tang, X.; Zhou, J.; Yang, J.; Yang, M.; Xu, J. De Novo Genome Assembly of Chinese Plateau Honeybee Unravels Intraspecies Genetic Diversity in the Eastern Honeybee, Apis cerana. Insects 2021, 12, 891. [Google Scholar] [CrossRef]
  18. Sun, C.; Huang, J.; Wang, Y.; Zhao, X.; Su, L.; Thomas, G.W.; Zhao, M.; Zhang, X.; Jungreis, I.; Kellis, M. Genus-wide characterization of bumblebee genomes provides insights into their evolution and variation in ecological and behavioral traits. Mol. Biol. Evol. 2021, 38, 486–501. [Google Scholar] [CrossRef]
  19. Sadd, B.M.; Barribeau, S.M.; Bloch, G.; De Graaf, D.C.; Dearden, P.; Elsik, C.G.; Gadau, J.; Grimmelikhuijzen, C.J.; Hasselmann, M.; Lozier, J.D. The genomes of two key bumblebee species with primitive eusocial organization. Genome Biol. 2015, 16, 76. [Google Scholar] [CrossRef] [Green Version]
  20. Oppenheim, S.; Cao, X.; Rueppel, O.; Krongdang, S.; Phokasem, P.; DeSalle, R.; Goodwin, S.; Xing, J.; Chantawannakul, P.; Rosenfeld, J.A. Whole genome sequencing and assembly of the Asian honey bee Apis dorsata. Genome Biol. Evol. 2020, 12, 3677–3683. [Google Scholar] [CrossRef] [Green Version]
  21. Cremer, S.; Armitage, S.A.O.; Schmid-Hempel, P. Social Immunity. Curr. Biol. 2007, 17, R693–R702. [Google Scholar] [CrossRef]
  22. Xu, J.; James, R. Genes related to immunity, as expressed in the alfalfa leafcutting bee, Megachile rotundata, during pathogen challenge. Insect Mol. Biol. 2009, 18, 785–794. [Google Scholar] [CrossRef] [PubMed]
  23. Kapheim, K.M.; Pan, H.; Li, C.; Salzberg, S.L.; Puiu, D.; Magoc, T.; Robertson, H.M.; Hudson, M.E.; Venkat, A.; Fischman, B.J. Genomic signatures of evolutionary transitions from solitary to group living. Science 2015, 348, 1139–1143. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Werren, J.H.; Richards, S.; Desjardins, C.A.; Niehuis, O.; Gadau, J.; Colbourne, J.K.; Group, N.G.W.; Beukeboom, L.W.; Desplan, C.; Elsik, C.G. Functional and evolutionary insights from the genomes of three parasitoid Nasonia species. Science 2010, 327, 343–348. [Google Scholar] [CrossRef]
  25. Xiao, J.-H.; Yue, Z.; Jia, L.-Y.; Yang, X.-H.; Niu, L.-H.; Wang, Z.; Zhang, P.; Sun, B.-F.; He, S.-M.; Li, Z.; et al. Obligate mutualism within a host drives the extreme specialization of a fig wasp genome. Genome Biol. 2013, 14, R141. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Barribeau, S.M.; Sadd, B.M.; du Plessis, L.; Brown, M.J.F.; Buechel, S.D.; Cappelle, K.; Carolan, J.C.; Christiaens, O.; Colgan, T.J.; Erler, S.; et al. A depauperate immune repertoire precedes evolution of sociality in bees. Genome Biol. 2015, 16, 83. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Zhou, Q.-S.; Luo, A.; Zhang, F.; Niu, Z.-Q.; Wu, Q.-T.; Xiong, M.; Orr, M.C.; Zhu, C.-D. The First Draft Genome of the Plasterer Bee Colletes gigas (Hymenoptera: Colletidae: Colletes). Genome Biol. Evol. 2020, 12, 860–866. [Google Scholar] [CrossRef]
  28. Wey, B.; Heavner, M.E.; Wittmeyer, K.T.; Briese, T.; Hopper, K.R.; Govind, S. Immune suppressive extracellular vesicle proteins of Leptopilina heterotoma are encoded in the wasp genome. G3 Genes Genomes Genet. 2020, 10, 1–12. [Google Scholar] [CrossRef] [Green Version]
  29. Heraghty, S.D.; Sutton, J.M.; Pimsler, M.L.; Fierst, J.L.; Strange, J.P.; Lozier, J.D. De Novo Genome Assemblies for Three North American Bumble Bee Species: Bombus bifarius, Bombus vancouverensis, and Bombus vosnesenskii. G3 Genes Genomes Genet. 2020, 10, 2585–2592. [Google Scholar] [CrossRef]
  30. Tvedte, E.S.; Walden, K.K.; McElroy, K.E.; Werren, J.H.; Forbes, A.A.; Hood, G.R.; Logsdon, J.M., Jr.; Feder, J.L.; Robertson, H.M. Genome of the parasitoid wasp Diachasma alloeum, an emerging model for ecological speciation and transitions to asexual reproduction. Genome Biol. Evol. 2019, 11, 2767–2773. [Google Scholar] [CrossRef]
  31. Kapheim, K.M.; Pan, H.; Li, C.; Blatti, C., III; Harpur, B.A.; Ioannidis, P.; Jones, B.M.; Kent, C.F.; Ruzzante, L.; Sloofman, L. Draft genome assembly and population genetics of an agricultural pollinator, the solitary alkali bee (Halictidae: Nomia melanderi). G3 Genes Genomes Genet. 2019, 9, 625–634. [Google Scholar] [CrossRef]
  32. Yin, C.; Li, M.; Hu, J.; Lang, K.; Chen, Q.; Liu, J.; Guo, D.; He, K.; Dong, Y.; Luo, J. The genomic features of parasitism, polyembryony and immune evasion in the endoparasitic wasp Macrocentrus cingulum. BMC Genom. 2018, 19, 420. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Haddad, N.J.; Adjlane, N.; Saini, D.; Menon, A.; Krishnamurthy, V.; Jonklaas, D.; Tomkins, J.P.; Loucif-Ayad, W.; Horth, L. Whole-genome sequencing of north African honey bee Apis mellifera intermissa to assess its beneficial traits. Entomol. Res. 2018, 48, 174–186. [Google Scholar] [CrossRef]
  34. Geib, S.M.; Liang, G.H.; Murphy, T.D.; Sim, S.B. Whole genome sequencing of the braconid parasitoid wasp Fopius arisanus, an important biocontrol agent of pest tepritid fruit flies. G3 Genes Genomes Genet. 2017, 7, 2407–2411. [Google Scholar] [CrossRef] [Green Version]
  35. Brand, P.; Saleh, N.; Pan, H.; Li, C.; Kapheim, K.M.; Ramírez, S.R. The nuclear and mitochondrial genomes of the facultatively eusocial orchid bee Euglossa dilemma. G3 Genes Genomes Genet. 2017, 7, 2891–2898. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Standage, D.S.; Berens, A.J.; Glastad, K.M.; Severin, A.J.; Brendel, V.P.; Toth, A.L. Genome, transcriptome and methylome sequencing of a primitively eusocial wasp reveal a greatly reduced DNA methylation system in a social insect. Mol. Ecol. 2016, 25, 1769–1784. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Rehan, S.M.; Glastad, K.M.; Lawson, S.P.; Hunt, B.G. The genome and methylome of a subsocial small carpenter bee, Ceratina calcarata. Genome Biol. Evol. 2016, 8, 1401–1410. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Haddad, N.J.; Loucif-Ayad, W.; Adjlane, N.; Saini, D.; Manchiganti, R.; Krishnamurthy, V.; AlShagoor, B.; Batainh, A.M.; Mugasimangalam, R. Draft genome sequence of the Algerian bee Apis mellifera intermissa. Genom. Data 2015, 4, 24–25. [Google Scholar] [CrossRef] [Green Version]
  39. Kocher, S.D.; Li, C.; Yang, W.; Tan, H.; Yi, S.V.; Yang, X.; Hoekstra, H.E.; Zhang, G.; Pierce, N.E.; Yu, D.W. The draft genome of a socially polymorphic halictid bee, Lasioglossum albipes. Genome Biol. 2013, 14, R142. [Google Scholar] [CrossRef] [Green Version]
  40. Hrdlickova, R.; Toloue, M.; Tian, B. RNA-Seq methods for transcriptome analysis. Wiley Interdiscip. Rev. RNA 2017, 8, e1364. [Google Scholar] [CrossRef] [Green Version]
  41. DeGrandi-Hoffman, G.; Chen, Y. Nutrition, immunity and viral infections in honey bees. Curr. Opin. Insect Sci. 2015, 10, 170–176. [Google Scholar] [CrossRef]
  42. Grozinger, C.M.; Flenniken, M.L. Bee Viruses: Ecology, Pathogenicity, and Impacts. Annu. Rev. Entomol. 2019, 64, 205–226. [Google Scholar] [CrossRef] [PubMed]
  43. McMenamin, A.J.; Daughenbaugh, K.F.; Parekh, F.; Pizzorno, M.C.; Flenniken, M.L. Honey Bee and Bumble Bee Antiviral Defense. Viruses 2018, 10, 395. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Wang, K.; Zhu, L.; Rao, L.; Zhao, L.; Wang, Y.; Wu, X.; Zheng, H.; Liao, X. Nano- and micro-polystyrene plastics disturb gut microbiota and intestinal immune system in honeybee. Sci. Total Environ. 2022, 842, 156819. [Google Scholar] [CrossRef] [PubMed]
  45. McMenamin, A.J.; Flenniken, M.L. Recently identified bee viruses and their impact on bee pollinators. Curr. Opin. Insect Sci. 2018, 26, 120–129. [Google Scholar] [CrossRef] [PubMed]
  46. de Miranda, J.R.; Gauthier, L.; Ribière, M.; Chen, Y.P. Honey bee viruses and their effect on bee and colony health. In Honey Bee Colony Health; CRC Press: Boca Raton, FL, USA, 2011; pp. 71–102. [Google Scholar] [CrossRef]
  47. Yañez, O.; Piot, N.; Dalmon, A.; de Miranda, J.R.; Chantawannakul, P.; Panziera, D.; Amiri, E.; Smagghe, G.; Schroeder, D.; Chejanovsky, N. Bee Viruses: Routes of Infection in Hymenoptera. Front. Microbiol. 2020, 11, 943. [Google Scholar] [CrossRef]
  48. Galbraith, D.A.; Yang, X.; Niño, E.L.; Yi, S.; Grozinger, C. Parallel Epigenomic and Transcriptomic Responses to Viral Infection in Honey Bees (Apis mellifera). PLoS Pathog. 2015, 11, e1004713. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Li-Byarlay, H.; Boncristiani, H.; Howell, G.; Herman, J.; Clark, L.; Strand, M.K.; Tarpy, D.; Rueppell, O. Transcriptomic and Epigenomic Dynamics of Honey Bees in Response to Lethal Viral Infection. Front. Genet. 2020, 11, 566320. [Google Scholar] [CrossRef]
  50. Doublet, V.; Paxton, R.J.; McDonnell, C.M.; Dubois, E.; Nidelet, S.; Moritz, R.F.A.; Alaux, C.; Le Conte, Y. Brain transcriptomes of honey bees (Apis mellifera) experimentally infected by two pathogens: Black queen cell virus and Nosema ceranae. Genom. Data 2016, 10, 79–82. [Google Scholar] [CrossRef]
  51. Ryabov, E.V.; Fannon, J.M.; Moore, J.D.; Wood, G.R.; Evans, D.J. The Iflaviruses Sacbrood virus and Deformed wing virus evoke different transcriptional responses in the honeybee which may facilitate their horizontal or vertical transmission. PeerJ 2016, 4, e1591. [Google Scholar] [CrossRef]
  52. Kim, W.J.; Lee, S.-H.; Kim, J.H.; Fang, Y.; Ha, K.B.; Park, D.H.; Choi, J.Y.; Je, Y.H. Differential gene expressions of innate immune related genes of the Asian honeybee, Apis cerana, latently infected with sacbrood virus. J. Asia-Pac. Entomol. 2017, 20, 17–21. [Google Scholar] [CrossRef]
  53. Deng, Y.; Zhao, H.; Shen, S.; Yang, S.; Yang, D.; Deng, S.; Hou, C. Identification of Immune Response to Sacbrood Virus Infection in Apis cerana Under Natural Condition. Front. Genet. 2020, 11, 587509. [Google Scholar] [CrossRef] [PubMed]
  54. Guo, Y.; Zhang, Z.; Zhuang, M.; Wang, L.; Li, K.; Yao, J.; Yang, H.; Huang, J.; Hao, Y.; Ying, F. Transcriptome profiling reveals a novel mechanism of antiviral immunity upon sacbrood virus infection in honey bee larvae (Apis cerana). Front. Microbiol. 2021, 12, 615893. [Google Scholar] [CrossRef] [PubMed]
  55. Pizzorno, M.C.; Field, K.; Kobokovich, A.L.; Martin, P.L.; Gupta, R.A.; Mammone, R.; Rovnyak, D.; Capaldi, E.A. Transcriptomic responses of the honey bee brain to infection with deformed wing virus. Viruses 2021, 13, 287. [Google Scholar] [CrossRef] [PubMed]
  56. Amiri, E.; Herman, J.J.; Strand, M.K.; Tarpy, D.R.; Rueppell, O. Egg transcriptome profile responds to maternal virus infection in honey bees, Apis mellifera. Infect. Genet. Evol. 2020, 85, 104558. [Google Scholar] [CrossRef]
  57. Traynor, K.S.; Mondet, F.; de Miranda, J.R.; Techer, M.; Kowallik, V.; Oddie, M.A.Y.; Chantawannakul, P.; McAfee, A. Varroa destructor: A Complex Parasite, Crippling Honey Bees Worldwide. Trends Parasitol. 2020, 36, 592–606. [Google Scholar] [CrossRef]
  58. Nazzi, F.; Brown, S.P.; Annoscia, D.; Del Piccolo, F.; Di Prisco, G.; Varricchio, P.; Della Vedova, G.; Cattonaro, F.; Caprio, E.; Pennacchio, F. Synergistic Parasite-Pathogen Interactions Mediated by Host Immunity Can Drive the Collapse of Honeybee Colonies. PLoS Pathog. 2012, 8, e1002735. [Google Scholar] [CrossRef] [Green Version]
  59. Annoscia, D.; Brown, S.P.; Di Prisco, G.; De Paoli, E.; Del Fabbro, S.; Frizzera, D.; Zanni, V.; Galbraith, D.A.; Caprio, E.; Grozinger, C.M.; et al. Haemolymph removal by Varroa mite destabilizes the dynamical interaction between immune effectors and virus in bees, as predicted by Volterra’s model. Proc. R. Soc. B Biol. Sci. 2019, 286, 20190331. [Google Scholar] [CrossRef] [Green Version]
  60. Doublet, V.; Poeschl, Y.; Gogol-Döring, A.; Alaux, C.; Annoscia, D.; Aurori, C.; Barribeau, S.M.; Bedoya-Reina, O.C.; Brown, M.J.F.; Bull, J.C.; et al. Unity in defence: Honeybee workers exhibit conserved molecular responses to diverse pathogens. BMC Genom. 2017, 18, 207. [Google Scholar] [CrossRef] [Green Version]
  61. Noël, A.; Le Conte, Y.; Mondet, F. Varroa destructor: How does it harm Apis mellifera honey bees and what can be done about it? Emerg. Top. Life Sci. 2020, 4, 45–57. [Google Scholar] [CrossRef]
  62. Ryabov, E.V.; Wood, G.R.; Fannon, J.M.; Moore, J.D.; Bull, J.C.; Chandler, D.; Mead, A.; Burroughs, N.; Evans, D.J. A Virulent Strain of Deformed Wing Virus (DWV) of Honeybees (Apis mellifera) Prevails after Varroa destructor-Mediated, or In Vitro, Transmission. PLoS Pathog. 2014, 10, e1004230. [Google Scholar] [CrossRef] [Green Version]
  63. Zanni, V.; Galbraith, D.A.; Annoscia, D.; Grozinger, C.M.; Nazzi, F. Transcriptional signatures of parasitization and markers of colony decline in Varroa-infested honey bees (Apis mellifera). Insect Biochem. Mol. Biol. 2017, 87, 1–13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Navajas, M.; Migeon, A.; Alaux, C.; Martin-Magniette, M.L.; Robinson, G.E.; Evans, J.D.; Cros-Arteil, S.; Crauser, D.; Le Conte, Y. Differential gene expression of the honey bee Apis mellifera associated with Varroa destructor infection. BMC Genom. 2008, 9, 301. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Le Conte, Y.; Alaux, C.; Martin, J.F.; Harbo, J.R.; Harris, J.W.; Dantec, C.; Séverac, D.; Cros-Arteil, S.; Navajas, M. Social immunity in honeybees (Apis mellifera): Transcriptome analysis of varroa-hygienic behaviour. Insect Mol. Biol. 2011, 20, 399–408. [Google Scholar] [CrossRef] [PubMed]
  66. Alaux, C.; Dantec, C.; Parrinello, H.; Le Conte, Y. Nutrigenomics in honey bees: Digital gene expression analysis of pollen’s nutritive effects on healthy and varroa-parasitized bees. BMC Genom. 2011, 12, 496. [Google Scholar] [CrossRef] [Green Version]
  67. Aufauvre, J.; Misme-Aucouturier, B.; Viguès, B.; Texier, C.; Delbac, F.; Blot, N. Transcriptome Analyses of the Honeybee Response to Nosema ceranae and Insecticides. PLoS ONE 2014, 9, e91686. [Google Scholar] [CrossRef] [PubMed]
  68. Badaoui, B.; Fougeroux, A.; Petit, F.; Anselmo, A.; Gorni, C.; Cucurachi, M.; Cersini, A.; Granato, A.; Cardeti, G.; Formato, G.; et al. RNA-sequence analysis of gene expression from honeybees (Apis mellifera) infected with Nosema ceranae. PLoS ONE 2017, 12, e0173438. [Google Scholar] [CrossRef] [Green Version]
  69. Chen, D.; Chen, H.; Du, Y.; Zhou, D.; Geng, S.; Wang, H.; Wan, J.; Xiong, C.; Zheng, Y.; Guo, R. Genome-Wide Identification of Long Non-Coding RNAs and Their Regulatory Networks Involved in Apis mellifera ligustica Response to Nosema ceranae Infection. Insects 2019, 10, 245. [Google Scholar] [CrossRef] [Green Version]
  70. Xing, W.; Zhou, D.; Long, Q.; Sun, M.; Guo, R.; Wang, L. Immune response of eastern honeybee worker to Nosema ceranae infection revealed by transcriptomic investigation. Insects 2021, 12, 728. [Google Scholar] [CrossRef]
  71. Zhu, Z.; Wang, J.; Fan, X.; Long, Q.; Chen, H.; Ye, Y.; Zhang, K.; Ren, Z.; Zhang, Y.; Niu, Q.; et al. CircRNA-regulated immune response of Asian honey bee workers to microsporidian infection. bioRxiv 2022. [Google Scholar] [CrossRef]
  72. Riddell, C.E.; Lobaton Garces, J.D.; Adams, S.; Barribeau, S.M.; Twell, D.; Mallon, E.B. Differential gene expression and alternative splicing in insect immune specificity. BMC Genom. 2014, 15, 1031. [Google Scholar] [CrossRef] [Green Version]
  73. Colgan, T.J.; Carolan, J.C.; Sumner, S.; Blaxter, M.L.; Brown, M.J.F. Infection by the castrating parasitic nematode Sphaerularia bombi changes gene expression in Bombus terrestris bumblebee queens. Insect Mol. Biol. 2020, 29, 170–182. [Google Scholar] [CrossRef] [PubMed]
  74. Tsvetkov, N.; MacPhail, V.J.; Colla, S.R.; Zayed, A. Conservation genomics reveals pesticide and pathogen exposure in the declining bumble bee Bombus terricola. Mol. Ecol. 2021, 30, 4220–4230. [Google Scholar] [CrossRef] [PubMed]
  75. Richard, F.-J.; Holt, H.L.; Grozinger, C.M. Effects of immunostimulation on social behavior, chemical communication and genome-wide gene expression in honey bee workers (Apis mellifera). BMC Genom. 2012, 13, 558. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Cornman, R.S.; Lopez, D.; Evans, J.D. Transcriptional response of honey bee larvae infected with the bacterial pathogen Paenibacillus larvae. PLoS ONE 2013, 8, e65424. [Google Scholar] [CrossRef] [Green Version]
  77. Xu, B.; Wang, Y.; Zhu, S.; Zhou, H.; Gou, C.; Dong, W.; Wang, Y.; Gao, Y.; Ma, H. Transcriptional profiling reveals the molecular bases of immune regulation in Apis mellifera in response to Ascosphaera apis infection. Entomol. Res. 2019, 49, 26–34. [Google Scholar] [CrossRef] [Green Version]
  78. Chen, D.; Guo, R.; Xu, X.; Xiong, C.; Liang, Q.; Zheng, Y.; Luo, Q.; Zhang, Z.; Huang, Z.; Kumar, D.; et al. Uncovering the immune responses of Apis mellifera ligustica larval gut to Ascosphaera apis infection utilizing transcriptome sequencing. Gene 2017, 621, 40–50. [Google Scholar] [CrossRef]
  79. Guo, R.; Chen, D.; Diao, Q.; Xiong, C.; Zheng, Y.; Hou, C. Transcriptomic investigation of immune responses of the Apis cerana cerana larval gut infected by Ascosphaera apis. J. Invertebr. Pathol. 2019, 166, 107210. [Google Scholar] [CrossRef]
  80. Emery, O.; Schmidt, K.; Engel, P. Immune system stimulation by the gut symbiont Frischella perrara in the honey bee (Apis mellifera). Mol. Ecol. 2017, 26, 2576–2590. [Google Scholar] [CrossRef]
  81. Lang, H.; Duan, H.; Wang, J.; Zhang, W.; Guo, J.; Zhang, X.; Hu, X.; Zheng, H. Specific Strains of Honeybee Gut Lactobacillus Stimulate Host Immune System to Protect against Pathogenic Hafnia alvei. Microbiol. Spectr. 2022, 10, e01896-21. [Google Scholar] [CrossRef]
  82. Wang, K.; Li, J.; Zhao, L.; Mu, X.; Wang, C.; Wang, M.; Xue, X.; Qi, S.; Wu, L. Gut microbiota protects honey bees (Apis mellifera L.) against polystyrene microplastics exposure risks. J. Hazard. Mater. 2021, 402, 123828. [Google Scholar] [CrossRef]
  83. Deng, Y.; Jiang, X.; Zhao, H.; Yang, S.; Gao, J.; Wu, Y.; Diao, Q.; Hou, C. Microplastic Polystyrene Ingestion Promotes the Susceptibility of Honeybee to Viral Infection. Environ. Sci. Technol. 2021, 55, 11680–11692. [Google Scholar] [CrossRef] [PubMed]
  84. Whiting, A.R. The Biology of the Parasitic Wasp Mormoniella vitripennis [=Nasonia brevicornis] (Walker). Q. Rev. Biol. 1967, 42, 333–406. [Google Scholar] [CrossRef]
  85. Sackton, T.B.; Werren, J.H.; Clark, A.G. Characterizing the Infection-Induced Transcriptome of Nasonia vitripennis Reveals a Preponderance of Taxonomically-Restricted Immune Genes. PLoS ONE 2013, 8, e83984. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Wu, M.-C.; Chang, Y.-W.; Lu, K.-H.; Yang, E.-C. Gene expression changes in honey bees induced by sublethal imidacloprid exposure during the larval stage. Insect Biochem. Mol. Biol. 2017, 88, 12–20. [Google Scholar] [CrossRef] [PubMed]
  87. Li, Z.; Li, M.; He, J.; Zhao, X.; Chaimanee, V.; Huang, W.F.; Nie, H.; Zhao, Y.; Su, S. Differential physiological effects of neonicotinoid insecticides on honey bees: A comparison between Apis mellifera and Apis cerana. Pestic. Biochem. Physiol. 2017, 140, 1–8. [Google Scholar] [CrossRef] [PubMed]
  88. Bebane, P.S.A.; Hunt, B.J.; Pegoraro, M.; Jones, A.R.C.; Marshall, H.; Rosato, E.; Mallon, E.B. The effects of the neonicotinoid imidacloprid on gene expression and DNA methylation in the buff-tailed bumblebee Bombus terrestris. Proc. R. Soc. B Biol. Sci. 2019, 286, 20190718. [Google Scholar] [CrossRef] [PubMed]
  89. Zhang, Z.Y.; Li, Z.; Huang, Q.; Zeng, Z.J. The effects of sublethal doses of imidacloprid and deltamethrin on honeybee foraging time and the brain transcriptome. J. Appl. Entomol. 2022, 146, 1169–1177. [Google Scholar] [CrossRef]
  90. Christen, V.; Schirrmann, M.; Frey, J.E.; Fent, K. Global Transcriptomic Effects of Environmentally Relevant Concentrations of the Neonicotinoids Clothianidin, Imidacloprid, and Thiamethoxam in the Brain of Honey Bees (Apis mellifera). Environ. Sci. Technol. 2018, 52, 7534–7544. [Google Scholar] [CrossRef]
  91. Mobley, M.W.; Gegear, R.J. One size does not fit all: Caste and sex differences in the response of bumblebees (Bombus impatiens) to chronic oral neonicotinoid exposure. PLoS ONE 2018, 13, e0200041. [Google Scholar] [CrossRef]
  92. Morfin, N.; Goodwin, P.H.; Guzman-Novoa, E. Interaction of Varroa destructor and Sublethal Clothianidin Doses during the Larval Stage on Subsequent Adult Honey Bee (Apis mellifera L.) Health, Cellular Immunity, Deformed Wing Virus Levels and Differential Gene Expression. Microorganisms 2020, 8, 858. [Google Scholar] [CrossRef]
  93. Shi, T.F.; Wang, Y.F.; Liu, F.; Qi, L.; Yu, L.S. Sublethal Effects of the Neonicotinoid Insecticide Thiamethoxam on the Transcriptome of the Honey Bees (Hymenoptera: Apidae). J. Econ. Entomol. 2017, 110, 2283–2289. [Google Scholar] [CrossRef] [PubMed]
  94. Fent, K.; Schmid, M.; Hettich, T.; Schmid, S. The neonicotinoid thiacloprid causes transcriptional alteration of genes associated with mitochondria at environmental concentrations in honey bees. Environ. Pollut. 2020, 266, 115297. [Google Scholar] [CrossRef] [PubMed]
  95. Huang, M.; Dong, J.; Guo, H.; Xiao, M.; Wang, D. Identification of long noncoding RNAs reveals the effects of dinotefuran on the brain in Apis mellifera (Hymenopptera: Apidae). BMC Genom. 2021, 22, 502. [Google Scholar] [CrossRef] [PubMed]
  96. Christen, V.; Fent, K. Exposure of honey bees (Apis mellifera) to different classes of insecticides exhibit distinct molecular effect patterns at concentrations that mimic environmental contamination. Environ. Pollut. 2017, 226, 48–59. [Google Scholar] [CrossRef] [PubMed]
  97. Zhu, L.; Qi, S.; Xue, X.; Niu, X.; Wu, L. Nitenpyram disturbs gut microbiota and influences metabolic homeostasis and immunity in honey bee (Apis mellifera L.). Environ. Pollut. 2020, 258, 113671. [Google Scholar] [CrossRef] [PubMed]
  98. Kablau, A.; Eckert, J.H.; Pistorius, J.; Sharbati, S.; Einspanier, R. Effects of selected insecticidal substances on mRNA transcriptome in larvae of Apis mellifera. Pestic. Biochem. Physiol. 2020, 170, 104703. [Google Scholar] [CrossRef]
  99. Korb, J.; Meusemann, K.; Aumer, D.; Bernadou, A.; Elsner, D.; Feldmeyer, B.; Foitzik, S.; Heinze, J.; Libbrecht, R.; Lin, S.; et al. Comparative transcriptomic analysis of the mechanisms underpinning ageing and fecundity in social insects. Philos. Trans. R. Soc. B Biol. Sci. 2021, 376, 20190728. [Google Scholar] [CrossRef]
  100. Dai, J.; Shu, R.; Liu, J.; Xia, J.; Jiang, X.; Zhao, P. Transcriptome analysis of Apis mellifera under benomyl stress to discriminate the gene expression in response to development and immune systems. J. Environ. Sci. Health Part B 2021, 56, 594–605. [Google Scholar] [CrossRef]
  101. Boncristiani, H.; Underwood, R.; Schwarz, R.; Evans, J.D.; Pettis, J.; vanEngelsdorp, D. Direct effect of acaricides on pathogen loads and gene expression levels in honey bees Apis mellifera. J. Insect Physiol. 2012, 58, 613–620. [Google Scholar] [CrossRef]
  102. Vannette, R.L.; Mohamed, A.; Johnson, B.R. Forager bees (Apis mellifera) highly express immune and detoxification genes in tissues associated with nectar processing. Sci. Rep. 2015, 5, 16224. [Google Scholar] [CrossRef]
Figure 1. Overview of the innate immunity of bees from the genome and transcriptome perspectives.
Figure 1. Overview of the innate immunity of bees from the genome and transcriptome perspectives.
Ijms 23 14278 g001
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Zhao, X.; Liu, Y. Current Knowledge on Bee Innate Immunity Based on Genomics and Transcriptomics. Int. J. Mol. Sci. 2022, 23, 14278. https://doi.org/10.3390/ijms232214278

AMA Style

Zhao X, Liu Y. Current Knowledge on Bee Innate Immunity Based on Genomics and Transcriptomics. International Journal of Molecular Sciences. 2022; 23(22):14278. https://doi.org/10.3390/ijms232214278

Chicago/Turabian Style

Zhao, Xiaomeng, and Yanjie Liu. 2022. "Current Knowledge on Bee Innate Immunity Based on Genomics and Transcriptomics" International Journal of Molecular Sciences 23, no. 22: 14278. https://doi.org/10.3390/ijms232214278

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop