Acessibilidade / Reportar erro

Tailed bacteriophages (Caudoviricetes) dominate the microbiome of a diseased stingless bee

Abstract

Bacteriophages, viruses that infect bacterial hosts, are known to rule the dynamics and diversity of bacterial populations in a number of ecosystems. Bacterial communities residing in the gut of animals, known as the gut microbiome, have revolutionized our understanding of many diseases. However, the gut phageome, while of apparent importance in this context, remains an underexplored area of research. Here we identify for the first time genomic sequences from tailed viruses (Caudoviricetes) that are associated with the microbiome of stingless bees (Melipona quadrifasciata). Both DNA and RNA were extracted from virus particles isolated from healthy and diseased forager bees, the latter showing symptoms from an annual syndrome that only affects M. quadrifasciata. Viral contigs from previously sequenced metagenomes of healthy and diseased forager bees were used for the analyses. Using conserved proteins deduced from their genomes, we found that Caudoviricetes were only present in the worker bee gut microbiome from diseased stingless bees. The most abundant phages are phylogenetically related to phages that infect Gram-positive bacteria from the order Lactobacillales and Gram-negative bacteria from the genus Gilliamella and Bartonella, that are common honey bee symbionts. The potential implication of these viruses in the M. quadrifasciata syndrome is discussed.

Keywords:
Metagenome; stingless bee; bacteriophage; virus; disease

Metagenomics revealed that viruses are the most abundant biological entities on Earth (Edwards and Rohwer 2005Edwards RA and Rohwer F (2005) Viral metagenomics. Nat Rev Microbiol 3:504-510.). Most of these viruses are bacteriophages, predators of bacteria, and archaeal viruses. It is estimated that every 48 hours half of all bacteria on Earth are killed by them (Shkoporov et al., 2022Shkoporov AN, Turkington CJ and Hill C (2022) Mutualistic interplay between bacteriophages and bacteria in the human gut. Nat Rev Microbiol 20:737-749.). Bacteriophages have fundamental roles in niches as distinct as the oceans’ sediments, where they participate in key biogeochemical processes, such as nutrient cycling (Fuhrman, 1999Fuhrman JA (1999) Marine viruses and their biogeochemical and ecological effects. Nature 399:541-548.), and the gut microbiome, where they control bacterial densities (Townsend et al., 2021Townsend EM, Kelly L, Muscatt G, Box JD, Hargraves N, Lilley D and Jameson E (2021) The Human Gut Phageome: Origins and roles in the human gut microbiome. Front Cell Infect Microbiol 11:643214. ). In the human gut, phages interact directly with their prey, and indirectly with the human immune system, being major players in human health and disease (Seo and Kweon, 2019Seo S-U and Kweon M-N (2019) Virome-host interactions in intestinal health and disease. Curr Opin Virol 37:63-71.). Honey bees are models for the study of microbiomes (Zheng et al., 2018Zheng H, Steele MI, Leonard SP, Motta EVS and Moran NA (2018) Honey bees as models for gut microbiota research. Lab Anim 47:317-325.) and their gut ‘phageome’ has been recently characterized focusing on the diversity, host range and functional potential of bacteriophages (Bonilla-Rosso et al., 2020Bonilla-Rosso G, Steiner T, Wichmann F, Bexkens E and Engel P (2020) Honey bees harbor a diverse gut virome engaging in nested strain-level interactions with the microbiota. Proc Natl Acad Sci U S A 117:7355-7362.; Deboutte et al., 2020Deboutte W, Beller L, Yinda CK, Maes P, de Graaf DC and Matthijnssens J (2020) Honey-bee-associated prokaryotic viral communities reveal wide viral diversity and a profound metabolic coding potential. Proc Natl Acad Sci U S A 117:10511-10519.; Busby et al., 2022Busby TJ, Miller CR, Moran NA and Van Leuven JT (2022) Global composition of the bacteriophage community in honey bees. mSystems 7:e01195-21.). It was found that most phages in the honey bee gut are virulent (Busby et al., 2022), viruses that do not insert themselves in bacterial genomes in the form of prophages, but instead hijack their host cells and use their resources to make new phages, causing the cell to lyse and die in the process. There is clear evidence that not all bacteria are infected by all phages, and that most phages can only infect a subset of bacterial species, i.e., they show host specificity (Koskella and Meaden, 2013Koskella B and Meaden S (2013) Understanding bacteriophage specificity in natural microbial communities. Viruses 5:806-823.). Thus, phages are modulators of the gut microbiota, which is essential for bee development, pollen digestion and immunity (Deboutte et al., 2020Deboutte W, Beller L, Yinda CK, Maes P, de Graaf DC and Matthijnssens J (2020) Honey-bee-associated prokaryotic viral communities reveal wide viral diversity and a profound metabolic coding potential. Proc Natl Acad Sci U S A 117:10511-10519.).

In recent years we have been investigating the composition and dynamics of the bacterial and fungal gut communities of a stingless bee, Melipona quadrifasciata Lepeletier 1836. Some microbiome changes are associated with an annual syndrome that often leads the colony to collapse (Díaz et al., 2017Díaz S, de Souza Urbano S, Caesar L, Blochtein B, Sattler A, Zuge V and Haag KL (2017) Report on the microbiota of Melipona quadrifasciata affected by a recurrent disease. J Invertebr Pathol 143:35-39.; Haag et al., 2023Haag KL, Caesar L, da Silveira Regueira-Neto M, de Sousa DR, Montenegro Marcelino V, de Queiroz Balbino V and Torres Carvalho A (2023) Temporal changes in gut microbiota composition and pollen diet associated with colony weakness of a stingless bee. Microb Ecol 85:1514-1526.). We also used metagenomics to identify differences in the virome composition of healthy and diseased stingless bees. Our data allowed us to characterize seven novel viruses with the potential of causing the neurological symptoms that we observed in some of the diseased colonies, but none of them was consistently associated with the disease outbreaks (Caesar et al., 2019Caesar L, Cibulski SP, Canal CW, Blochtein B, Sattler A and Haag KL (2019) The virome of an endangered stingless bee suffering from annual mortality in southern Brazil. J Gen Virol 100:1153-1164.). Gene expression studies on diseased vs. healthy M. quadrifasciata, rather suggest that the underlying causes of the annual syndrome are multifactorial, and involve a weakening process of the bee colony that culminates in March when outbreaks occur synchronously in different regions of southern Brazil (Caesar et al., 2022Caesar L, Lopes AMC, Radaeski JN, Bauermann SG, Konzen ER, Pombert J-F, Sattler A, Blochtein B, Carvalho AT and Haag KL (2022) Longitudinal survey reveals delayed effects of low gene expression on stingless bee colony health. J Apic Res 61:654-663.). Between January and March forager bees lose weight and bacterial counts increase in the gut, suggesting a relaxation in the bee immunologic mechanisms that regulate bacterial growth (Haag et al., 2023Haag KL, Caesar L, da Silveira Regueira-Neto M, de Sousa DR, Montenegro Marcelino V, de Queiroz Balbino V and Torres Carvalho A (2023) Temporal changes in gut microbiota composition and pollen diet associated with colony weakness of a stingless bee. Microb Ecol 85:1514-1526.). Here we use the metagenomic data generated in our previous virome characterization (Caesar et al., 2019Caesar L, Cibulski SP, Canal CW, Blochtein B, Sattler A and Haag KL (2019) The virome of an endangered stingless bee suffering from annual mortality in southern Brazil. J Gen Virol 100:1153-1164.) to identify bacteriophages that may participate in the process of modulating the M. quadrifasciata gut microbiota.

Our analyses were run on four assemblies built from sequences generated by high throughput sequencing of virus particles separated by centrifugation (Caesar et al., 2019Caesar L, Cibulski SP, Canal CW, Blochtein B, Sattler A and Haag KL (2019) The virome of an endangered stingless bee suffering from annual mortality in southern Brazil. J Gen Virol 100:1153-1164.; BioProject PRJNA960650BioProject: Raw virome data, BioProject: Raw virome data, https://www.ncbi.nlm.nih.gov/bioproject/PRJNA960650/ (accessed 3 January 2024)
https://www.ncbi.nlm.nih.gov/bioproject/...
). Two viral contig assemblies (DNA and RNA) came from a single pooled sample of worker bees from a diseased hive of M. quadrifasciata. The other two assemblies (DNA and RNA) are from a healthy hive of M. quadrifasciata sampled simultaneously at the same meliponary. For the identification of bacteriophage contigs we used the four sets as inputs on two softwares, geNomad (Camargo et al., 2023Camargo AP, Roux S, Schulz F, Babinski M, Xu Y, Hu B, Chain PSG, Nayfach S and Kyrpides NC (2023) You can move, but you can’t hide: Identification of mobile genetic elements with geNomad. bioRXiv:2023.03.05.531206. ) and VirSorter2 (Guo et al., 2021Guo J, Bolduc B, Zayed AA, Varsani A, Dominguez-Huerta G, Delmont TO, Pratama AA, Gazitúa MC, Vik D, Sullivan MB et al. (2021) VirSorter2: a multi-classifier, expert-guided approach to detect diverse DNA and RNA viruses. Microbiome 9:37.). GeNomad was run end-to-end, using the flags --cleanup --splits 8 and the default virus score cutoff of >0.7. VirSorter2 was run with the flags --min-score 0.5 --keep-original-seq --hallmark-required-on-short. To confirm the identification of phages and check the quality of metagenome-assembled genomes (MAGs) we used CheckV (Nayfach et al., 2021Nayfach S, Camargo AP, Schulz F, Eloe-Fadrosh E, Roux S and Kyrpides NC (2021) CheckV assesses the quality and completeness of metagenome-assembled viral genomes. Nat Biotechnol 39:578-585.) with default commands. All identified phage contigs were retained for the following analyses since they had 0 host genes, and the majority of them had at least 1 viral hallmark gene.

Using both geNomad and VirSorter a total of 193 viral contigs were identified as phages, and went through the downstream analyses. Within the four assemblies, bacteriophages were only found among the unhealthy DNA and RNA contigs (n=64 and 129, respectively; Table 1). GeNomad also performs gene prediction with a modified version of Prodigal (Hyatt et al., 2010Hyatt D, Chen G-L, Locascio PF, Land ML, Larimer FW and Hauser LJ (2010) Prodigal: Prokaryotic gene recognition and translation initiation site identification. BMC Bioinformatics 11:119.) called prodigal-gv, and assigns the predicted proteins to geNomad’s markers using MMseqs2 (Steinegger and Söding, 2017Steinegger M and Söding J (2017) MMseqs2 enables sensitive protein sequence searching for the analysis of massive data sets. Nat Biotechnol 35:1026-1028.). The vast majority of the annotations of predicted genes from both datasets (DNA and RNA) were assigned to bacteriophage markers belonging to the Caudoviricetes (Tables 1 and S1 Table S1 - Summary of DNA and RNA contig taxonomy. ), a class of virulent phages known as the tailed phages, which currently contains the majority of the total phage sequences in public databases (Zhu et al., 2022Zhu Y, Shang J, Peng C and Sun Y (2022) Phage family classification under Caudoviricetes: A review of current tools using the latest ICTV classification framework. Front Microbiol 13:1032186. ).

Table 1 -
Phage distribution, taxonomy and abundance among the four metagenomes analyzed in the present study.

Next, contigs on each sample were de-replicated with dRep (Olm et al., 2017Olm MR, Brown CT, Brooks B and Banfield JF (2017) dRep: a tool for fast and accurate genomic comparisons that enables improved genome recovery from metagenomes through de-replication. ISME J 11:2864-2868.), using flags --P_ani 0.95 --cov_thresh 0.85, and coverage estimated by mapping the original reads against them with Bowtie2 (Langmead and Salzberg 2012Langmead B and Salzberg SL (2012) Fast gapped-read alignment with Bowtie 2. Nat Methods 9:357-359.), using the flag --no-discordant and --very-sensitive. The database for mapping reads included also non-phage contigs from the assembly to avoid non-specific or low quality reads mapping against the phage sequences. Coverage bam files were used as inputs along with the contigs for binning the phage genomes with vRhyme, using default commands (Kieft et al., 2022Kieft K, Adams A, Salamzade R, Kalan L and Anantharaman K (2022) vRhyme enables binning of viral genomes from metagenomes. Nucleic Acids Res 50:e83.). Binned and non-binned sequences from both samples were then de-replicated with dRep using the same parameters that follow the standard thresholds for vOTU classification (Roux et al., 2019Roux S, Adriaenssens EM, Dutilh BE, Koonin EV, Kropinski AM, Krupovic M, Kuhn JH, Lavigne R, Brister JR, Varsani A et al. (2019) Minimum information about an uncultivated virus genome (MIUViG). Nat Biotechnol 37:29-37.). Coverage of vOTUs in each sample was recovered again with Bowtie2 and command depth -a from Samtools (Li et al., 2009Li H, Handsaker B, Wysoker A, Fennell T, Ruan J, Homer N, Marth G, Abecasis G, Durbin R and 1000 Genome Project Data Processing Subgroup (2009) The Sequence Alignment/Map format and SAMtools. Bioinformatics 25:2078-2079.). Coverage and completeness estimates for the 182 vOTUs from both assemblies (DNA and RNA) are listed in Table S2 Table S2 - Metagenome coverage and contig length. . Coverage estimates ranged between 0.05 - 2704.15X (DNA) and 1.28 - 3134.75X (RNA). Completeness varied between 0.2 - 47.95% and 0.11 - 45.29%, for vOTUs from the DNA and RNA assemblies, respectively (Table 1). Thus, with the sequencing effort employed in our study, we assembled incomplete genomes, probably from the most abundant bacteriophages found in the gut of our diseased stingless bees (Figure 1).

Figure 1 -
Coverage estimates for vOTUs of variable lengths (bp) assembled from the unhealthy M. quadrifasciata DNA and RNA samples. Each dot on the chart corresponds to a phage MAG. Horizontal and vertical dashed lines mark the coverage and length thresholds, respectively, to keep vOTUs for the downstream analyses (see text for details).

We attempted to predict phage taxa and their putative hosts based on MAGs similarity to known phages, and on their match to CRISPR-spacers. First, proteins encoded by all phage vOTUs were predicted and primarily annotated with Prokka (Seemann, 2014Seemann T (2014) Prokka: Rapid prokaryotic genome annotation. Bioinforma Oxf Engl 30:2068-2069.), using flag -- kingdom Viruses. Proteins and contigs from the 5 largest vOTUs were used together with previously described bee phage sequences (Bonilla-Rosso et al., 2020Bonilla-Rosso G, Steiner T, Wichmann F, Bexkens E and Engel P (2020) Honey bees harbor a diverse gut virome engaging in nested strain-level interactions with the microbiota. Proc Natl Acad Sci U S A 117:7355-7362.; Deboutte et al., 2020Deboutte W, Beller L, Yinda CK, Maes P, de Graaf DC and Matthijnssens J (2020) Honey-bee-associated prokaryotic viral communities reveal wide viral diversity and a profound metabolic coding potential. Proc Natl Acad Sci U S A 117:10511-10519.; Busby et al., 2022Busby TJ, Miller CR, Moran NA and Van Leuven JT (2022) Global composition of the bacteriophage community in honey bees. mSystems 7:e01195-21.) as inputs for vConTACT2 (Bin Jang et al., 2019Bin Jang H, Bolduc B, Zablocki O, Kuhn JH, Roux S, Adriaenssens EM, Brister JR, Kropinski AM, Krupovic M, Lavigne R et al. (2019) Taxonomic assignment of uncultivated prokaryotic virus genomes is enabled by gene-sharing networks. Nat Biotechnol 37:632-639.). The program vConTACT2 was ran with default commands, using the ‘ProkaryoticViralRefSeq211-Merged’ database, and results were visualized with the R package IgraphIgraph for R, Igraph for R, https://zenodo.org/record/824064. (accessed 10 September 2023)
https://zenodo.org/record/824064....
. Only vOTU1 and vOTU3 could be connected to previously described phages, but with low confidence and none of the vOTUs clustered with any phage (Figure 2). Additionally, we used all phage contigs as queries in BLASTn, using flags -evalue 1e-3 -ungapped -perc_identity 95, against three CRISPR-spacer databases: CrisprOpenDB (Dion et al., 2021Dion MB, Plante P-L, Zufferey E, Shah SA, Corbeil J and Moineau S (2021) Streamlining CRISPR spacer-based bacterial host predictions to decipher the viral dark matter. Nucleic Acids Res 49:3127-3138.), spacers from honey bee microbiome-associated bacteria (Bonilla-Rosso et al., 2020Bonilla-Rosso G, Steiner T, Wichmann F, Bexkens E and Engel P (2020) Honey bees harbor a diverse gut virome engaging in nested strain-level interactions with the microbiota. Proc Natl Acad Sci U S A 117:7355-7362.), and spacers from stingless bees’ microbiome-associated bacteria. The latter database was built based on spacers that we predicted in complete genomes of bacteria sequenced from six species of stingless bees (Sarton-Lohéac et al., 2023Sarton-Lohéac G, Nunes da Silva CG, Mazel F, Baud G, de Bakker V, Das S, El Chazli Y, Ellegaard K, Garcia-Garcera M, Glover N et al. (2023) Deep divergence and genomic diversification of gut symbionts of Neotropical stingless bees. mBio 14:e0353822.) using the ‘CRISPRCasFinder’ online tool, with default settings, and evidence level 3 or 4. Unfortunately, none of our vOTUs have matches with CRISPR spacers from any of the three searched databases. We think that traditional approaches to identify viral hosts that are based on genomic data from bacteria and their phages are of limited use in the context of our study, since there are so far only a few characterized genomes from stingless bee microbiomes (e.g. Sarton-Lohéac et al., 2023Sarton-Lohéac G, Nunes da Silva CG, Mazel F, Baud G, de Bakker V, Das S, El Chazli Y, Ellegaard K, Garcia-Garcera M, Glover N et al. (2023) Deep divergence and genomic diversification of gut symbionts of Neotropical stingless bees. mBio 14:e0353822.).

Figure 2 -
Protein-sharing network displaying unclustered M. quadrifasciata phages and clustered phages of Apis mellifera. Nodes represent phage partial to complete genomes, and edges connecting them indicate a statistically significant similar protein profile between their genomes. No vOTU >10 kb clustered with known phages in the database, and only two had weak connections with known phages.

In a further attempt to infer the putative bacteria used as hosts by the dominant tailed phages inferred from our study (vOTUs 2, 5, 61, 62 and 65; see Figure 1 and Table S2) we used phylogenetic analyses. These MAGs showed the highest coverage in the RNA metagenome, which we interpreted as a proxy for their abundance in the unhealthy bee microbiome. Moreover, these MAGs contain hallmark genes that could be used for BLAST searches of related sequences. For vOTUs 2, 5 and 65 we used a gene encoding the DNA encapsidation protein (terminase) as phylogenetic marker. The protein deduced from vOTUs 5 and 65 was identical. For vOTUs 61 and 62 we used the predicted major capsid protein, which was also identical in both MAGs. Briefly, we selected reference protein sequences from GenBank (nr database) using BLASTp and aligned them to the corresponding sequences predicted from the vOTUs with MAFFT (Katoh and Standley, 2013Katoh K and Standley DM (2013) MAFFT multiple sequence alignment software version 7: improvements in performance and usability. Mol Biol Evol 30:772-780.). The alignment was used as input for phylogenetic analyses with PHYML (Guindon and Gascuel, 2003Guindon S and Gascuel O (2003) A simple, fast, and accurate algorithm to estimate large phylogenies by maximum likelihood. Syst Biol 52:696-704.) implemented in Geneious Prime 2022.1.1 (Biomatters Ltd.) using the LG evolutionary model and 500 bootstrap replicates. Whereas vOTUs 2, 61 and 62 seem to be related to tailed viruses isolated from Gram-negative bacteria, vOTUs 5 and 65 cluster with virus sequences obtained from Gram-positive bacteria belonging to the order Lactobacillales (Figure 3). Viral OTU2, which appeared both in the DNA and RNA metagenomes, clusters with phages from Bartonella, a facultative symbiont of the honey bee, and vOTUs 61 and 62 are related to a Gilliamella phage isolated from bumble bees in China. Proteobacteria such as Bartonella and Gilliamella are not as abundant in the M. quadrifasciata microbiome as yet unidentified Acetobacteraceae, in contrast to the Lactobacillales, that are highly abundant and diverse (Díaz et al., 2017Díaz S, de Souza Urbano S, Caesar L, Blochtein B, Sattler A, Zuge V and Haag KL (2017) Report on the microbiota of Melipona quadrifasciata affected by a recurrent disease. J Invertebr Pathol 143:35-39.; Cerqueira et al., 2021Cerqueira AES, Hammer TJ, Moran NA, Santana WC, Kasuya MCM and da Silva CC (2021) Extinction of anciently associated gut bacterial symbionts in a clade of stingless bees. ISME J 15:2813-2816.; Haag et al., 2023Haag KL, Caesar L, da Silveira Regueira-Neto M, de Sousa DR, Montenegro Marcelino V, de Queiroz Balbino V and Torres Carvalho A (2023) Temporal changes in gut microbiota composition and pollen diet associated with colony weakness of a stingless bee. Microb Ecol 85:1514-1526.; Sarton-Lohéac et al., 2023Sarton-Lohéac G, Nunes da Silva CG, Mazel F, Baud G, de Bakker V, Das S, El Chazli Y, Ellegaard K, Garcia-Garcera M, Glover N et al. (2023) Deep divergence and genomic diversification of gut symbionts of Neotropical stingless bees. mBio 14:e0353822.).

Figure 3 -
Maximum Likelihood phylogenies obtained using DNA encapsidation (A) and major capsid (B) proteins (see text for details). Proteins predicted from vOTUs 5 and 65, as well as vOTUs 61 and 62 were identical, and therefore correspond to a single branch on their respective phylogenetic trees.

Surprisingly, none of the sequences from our phageomes could be classified as the same phages previously characterized in honey bees (Bonilla-Rosso et al., 2020Bonilla-Rosso G, Steiner T, Wichmann F, Bexkens E and Engel P (2020) Honey bees harbor a diverse gut virome engaging in nested strain-level interactions with the microbiota. Proc Natl Acad Sci U S A 117:7355-7362.; Deboutte et al., 2020Deboutte W, Beller L, Yinda CK, Maes P, de Graaf DC and Matthijnssens J (2020) Honey-bee-associated prokaryotic viral communities reveal wide viral diversity and a profound metabolic coding potential. Proc Natl Acad Sci U S A 117:10511-10519.; Busby et al., 2022Busby TJ, Miller CR, Moran NA and Van Leuven JT (2022) Global composition of the bacteriophage community in honey bees. mSystems 7:e01195-21.). Nonetheless, Caudoviricetes were also the most abundant phages identified in the honey bee studies, which were not designed to assess matters related to bee health. Our study compares the phageomes of stingless bees from the same species sampled simultaneously from the same location, but differing in their health status. We only detected Caudoviricetes phages in diseased bees, leading us to reason that these phages might be implicated in their health. One hypothesis is that the virulent tailed phages opportunistically become more abundant in diseased M. quadrifasciata due to an overall increase in the amount of bacteria in the gut. Indeed, in our previous study we found that bacterial counts estimated by 16S qPCR increased between January and March, but we did not find significant differences between diseased and healthy bees (Haag et al., 2023Haag KL, Caesar L, da Silveira Regueira-Neto M, de Sousa DR, Montenegro Marcelino V, de Queiroz Balbino V and Torres Carvalho A (2023) Temporal changes in gut microbiota composition and pollen diet associated with colony weakness of a stingless bee. Microb Ecol 85:1514-1526.). An alternative but not mutually exclusive hypothesis is that, due to host specificity, tailed viruses disrupt the equilibrium of the bacterial community, and that microbiome dysbiosis negatively affects host homeostasis. We did find in previous studies of the M. quadrifasciata microbiome based on metabarcoding that the Lactobacillales reached their lowest relative abundance during the outbreak period, but again there were no significant differences between diseased and healthy bees (Haag et al., 2023Haag KL, Caesar L, da Silveira Regueira-Neto M, de Sousa DR, Montenegro Marcelino V, de Queiroz Balbino V and Torres Carvalho A (2023) Temporal changes in gut microbiota composition and pollen diet associated with colony weakness of a stingless bee. Microb Ecol 85:1514-1526.).

It is possible that the dynamics of the microbiome, including bacteriophages, have a longer-term effect on bee health that we did not bring to light by looking at a single outbreak. Indeed, by studying two successive outbreaks (2014 and 2015), we showed a significant interaction effect of sampling year and health status on microbiome composition (Díaz et al., 2017Díaz S, de Souza Urbano S, Caesar L, Blochtein B, Sattler A, Zuge V and Haag KL (2017) Report on the microbiota of Melipona quadrifasciata affected by a recurrent disease. J Invertebr Pathol 143:35-39.), implying that the microbiome profile of diseased bees is not the same for every outbreak. To obtain a more realistic view about the role of bacteriophages in the M. quadrifasciata microbiome, and its relationship with the annual syndrome, we need to work with deeper sequencing efforts. It would be possible to use a metagenomic strategy to characterize the phageomes and bacteriomes simultaneously in successive years. Nevertheless, to our knowledge, this is the first report of bacteriophages from a stingless bee microbiome. We expect that, in the near future, with proper sampling and deep sequencing, we will be able to clarify how phages control bacterial densities in the M. quadrifasciata gut and ultimately influence their health.

Acknowledgements

Our work is being supported by Fundação de Amparo à Pesquisa do Estado do Rio Grande do Sul FAPERGS PQG 19/2551-0001860-6.

References

  • Bin Jang H, Bolduc B, Zablocki O, Kuhn JH, Roux S, Adriaenssens EM, Brister JR, Kropinski AM, Krupovic M, Lavigne R et al (2019) Taxonomic assignment of uncultivated prokaryotic virus genomes is enabled by gene-sharing networks. Nat Biotechnol 37:632-639.
  • Bonilla-Rosso G, Steiner T, Wichmann F, Bexkens E and Engel P (2020) Honey bees harbor a diverse gut virome engaging in nested strain-level interactions with the microbiota. Proc Natl Acad Sci U S A 117:7355-7362.
  • Busby TJ, Miller CR, Moran NA and Van Leuven JT (2022) Global composition of the bacteriophage community in honey bees. mSystems 7:e01195-21.
  • Caesar L, Cibulski SP, Canal CW, Blochtein B, Sattler A and Haag KL (2019) The virome of an endangered stingless bee suffering from annual mortality in southern Brazil. J Gen Virol 100:1153-1164.
  • Caesar L, Lopes AMC, Radaeski JN, Bauermann SG, Konzen ER, Pombert J-F, Sattler A, Blochtein B, Carvalho AT and Haag KL (2022) Longitudinal survey reveals delayed effects of low gene expression on stingless bee colony health. J Apic Res 61:654-663.
  • Camargo AP, Roux S, Schulz F, Babinski M, Xu Y, Hu B, Chain PSG, Nayfach S and Kyrpides NC (2023) You can move, but you can’t hide: Identification of mobile genetic elements with geNomad. bioRXiv:2023.03.05.531206.
  • Cerqueira AES, Hammer TJ, Moran NA, Santana WC, Kasuya MCM and da Silva CC (2021) Extinction of anciently associated gut bacterial symbionts in a clade of stingless bees. ISME J 15:2813-2816.
  • Deboutte W, Beller L, Yinda CK, Maes P, de Graaf DC and Matthijnssens J (2020) Honey-bee-associated prokaryotic viral communities reveal wide viral diversity and a profound metabolic coding potential. Proc Natl Acad Sci U S A 117:10511-10519.
  • Díaz S, de Souza Urbano S, Caesar L, Blochtein B, Sattler A, Zuge V and Haag KL (2017) Report on the microbiota of Melipona quadrifasciata affected by a recurrent disease. J Invertebr Pathol 143:35-39.
  • Dion MB, Plante P-L, Zufferey E, Shah SA, Corbeil J and Moineau S (2021) Streamlining CRISPR spacer-based bacterial host predictions to decipher the viral dark matter. Nucleic Acids Res 49:3127-3138.
  • Edwards RA and Rohwer F (2005) Viral metagenomics. Nat Rev Microbiol 3:504-510.
  • Fuhrman JA (1999) Marine viruses and their biogeochemical and ecological effects. Nature 399:541-548.
  • Guindon S and Gascuel O (2003) A simple, fast, and accurate algorithm to estimate large phylogenies by maximum likelihood. Syst Biol 52:696-704.
  • Guo J, Bolduc B, Zayed AA, Varsani A, Dominguez-Huerta G, Delmont TO, Pratama AA, Gazitúa MC, Vik D, Sullivan MB et al (2021) VirSorter2: a multi-classifier, expert-guided approach to detect diverse DNA and RNA viruses. Microbiome 9:37.
  • Haag KL, Caesar L, da Silveira Regueira-Neto M, de Sousa DR, Montenegro Marcelino V, de Queiroz Balbino V and Torres Carvalho A (2023) Temporal changes in gut microbiota composition and pollen diet associated with colony weakness of a stingless bee. Microb Ecol 85:1514-1526.
  • Hyatt D, Chen G-L, Locascio PF, Land ML, Larimer FW and Hauser LJ (2010) Prodigal: Prokaryotic gene recognition and translation initiation site identification. BMC Bioinformatics 11:119.
  • Katoh K and Standley DM (2013) MAFFT multiple sequence alignment software version 7: improvements in performance and usability. Mol Biol Evol 30:772-780.
  • Kieft K, Adams A, Salamzade R, Kalan L and Anantharaman K (2022) vRhyme enables binning of viral genomes from metagenomes. Nucleic Acids Res 50:e83.
  • Koskella B and Meaden S (2013) Understanding bacteriophage specificity in natural microbial communities. Viruses 5:806-823.
  • Langmead B and Salzberg SL (2012) Fast gapped-read alignment with Bowtie 2. Nat Methods 9:357-359.
  • Li H, Handsaker B, Wysoker A, Fennell T, Ruan J, Homer N, Marth G, Abecasis G, Durbin R and 1000 Genome Project Data Processing Subgroup (2009) The Sequence Alignment/Map format and SAMtools. Bioinformatics 25:2078-2079.
  • Nayfach S, Camargo AP, Schulz F, Eloe-Fadrosh E, Roux S and Kyrpides NC (2021) CheckV assesses the quality and completeness of metagenome-assembled viral genomes. Nat Biotechnol 39:578-585.
  • Olm MR, Brown CT, Brooks B and Banfield JF (2017) dRep: a tool for fast and accurate genomic comparisons that enables improved genome recovery from metagenomes through de-replication. ISME J 11:2864-2868.
  • Roux S, Adriaenssens EM, Dutilh BE, Koonin EV, Kropinski AM, Krupovic M, Kuhn JH, Lavigne R, Brister JR, Varsani A et al (2019) Minimum information about an uncultivated virus genome (MIUViG). Nat Biotechnol 37:29-37.
  • Sarton-Lohéac G, Nunes da Silva CG, Mazel F, Baud G, de Bakker V, Das S, El Chazli Y, Ellegaard K, Garcia-Garcera M, Glover N et al (2023) Deep divergence and genomic diversification of gut symbionts of Neotropical stingless bees. mBio 14:e0353822.
  • Seemann T (2014) Prokka: Rapid prokaryotic genome annotation. Bioinforma Oxf Engl 30:2068-2069.
  • Seo S-U and Kweon M-N (2019) Virome-host interactions in intestinal health and disease. Curr Opin Virol 37:63-71.
  • Shkoporov AN, Turkington CJ and Hill C (2022) Mutualistic interplay between bacteriophages and bacteria in the human gut. Nat Rev Microbiol 20:737-749.
  • Steinegger M and Söding J (2017) MMseqs2 enables sensitive protein sequence searching for the analysis of massive data sets. Nat Biotechnol 35:1026-1028.
  • Townsend EM, Kelly L, Muscatt G, Box JD, Hargraves N, Lilley D and Jameson E (2021) The Human Gut Phageome: Origins and roles in the human gut microbiome. Front Cell Infect Microbiol 11:643214.
  • Zheng H, Steele MI, Leonard SP, Motta EVS and Moran NA (2018) Honey bees as models for gut microbiota research. Lab Anim 47:317-325.
  • Zhu Y, Shang J, Peng C and Sun Y (2022) Phage family classification under Caudoviricetes: A review of current tools using the latest ICTV classification framework. Front Microbiol 13:1032186.

Internet Resources

Supplementary material

The following online material is available for this article:

Table S1 - Summary of DNA and RNA contig taxonomy. Table S2 - Metagenome coverage and contig length.

Edited by

Associate Editor:

Loreta Brandão de Freitas

Publication Dates

  • Publication in this collection
    19 Jan 2024
  • Date of issue
    2023

History

  • Received
    28 Apr 2023
  • Accepted
    06 Dec 2023
Sociedade Brasileira de Genética Rua Cap. Adelmio Norberto da Silva, 736, 14025-670 Ribeirão Preto SP Brazil, Tel.: (55 16) 3911-4130 / Fax.: (55 16) 3621-3552 - Ribeirão Preto - SP - Brazil
E-mail: editor@gmb.org.br