Insect metamorphosis is regulated differently between sexes by members of a microRNA cluster

  1. School of Life Sciences, Simon F.S. Li Marine Science Laboratory, State Key Laboratory of Agrobiotechnology, Institute of Environment, Energy and Sustainability, The Chinese University of Hong Kong, Hong Kong SAR, China
  2. Institute of Agro-food Standard and Testing Technology, Shanghai Academy of Agricultural Sciences, China
  3. School of Biological Sciences, The University of Hong Kong, Hong Kong SAR, China
  4. Department of Cell and System Biology, University of Toronto, Canada
  5. Department of Biology, Queen’s University, Canada
  6. Shanghai Institute of Technology, Shanghai, China

Peer review process

Not revised: This Reviewed Preprint includes the authors’ original preprint (without revision), an eLife assessment, and public reviews.

Read more about eLife’s peer review process.

Editors

  • Reviewing Editor
    Virginie Courtier-Orgogozo
    CNRS - Université Paris Cité, Paris, France
  • Senior Editor
    Albert Cardona
    University of Cambridge, Cambridge, United Kingdom

Reviewer #1 (Public Review):

Summary:

In this paper, Li and colleagues have found mircoRNAs that affect levels of metamorphosis-regulating genes that can also affect levels of sesquiterpenoids (juvenile hormone and related compounds) and ecdysteriods, which regulate the timing and stages of insects, respectively. They first compared the transcriptomes of Drosophila at the third larval instar and at the white pre-pupa stage. They found thousands of differences in gene transcript levels between males and females, and between the two different stages. Among those genes that were differentially regulated they saw that genes involved in insect hormone biosynthesis were disproportionately represented. Many of the differentially regulated genes were involved in the insect hormone biosynthesis pathway and ascorbate and alderete metabolism. MicroRNAs were also differentially expressed during metamorphosis and were separately identified. The authors then considered genes and whether the differentially expressed microRNAs might regulate transcripts known to be involved in sesquiterpenoid production. In silico analysis of microRNAs predicted a list of 17 microRNAs that can regulate transcripts of sesquiterpenoid biosynthesis genes. The authors then used an in vitro luciferase assay to validate the binding and downregulation of 10 of the microRNAs to genes involved with sesquiterpenoid production in S2 cells.

Li and colleagues then focus on two genes they found were bound by microRNAs that have established roles in metamorphosis. The microRNAs miR-34 and miR-277 bind transcripts of two protein-coding genes that regulate metamorphosis Kr-h1, which encodes a transcription factor that is a JH-inducible transcription factor, and Allatostatin C Receptor 1, (AstC-R1), a G-protein coupled receptor that regulates the corpora allatum, the gland that produces sesquiterpenoids. Using a LAMP assay, one of the microRNAs, miR-277 was shown to bind to both AstC-R1 and Kr-h1 in in vivo whole-animal extracts. There is no mention of binding between either protein-coding transcript and the miR-34 microRNA. Temporal expression of all four transcripts shows that their abundance is anti-correlated; stages of high miR-34 or miR-277 expression correlate with low AstC-R1 or Kr-h1 expression. Homozygous deletions of both mircroRNAs result in 23% lethality, five days after adult eclosion. The authors also generated specific mutants in miR-34 or miR-277 and find differences in the expression of AstC-R1 and Kr-h1 and sex-specific differences in both sesquiterpenoids and ecdysteroids in the knock-out lines. If there were phenotypes associated with the specific knock-outs, those were not mentioned. Next, the authors examined the transcriptomes of the miR-3277 and miR-34 mutants and found several other GO-terms enriched among the differentially expressed genes. However, the sesquiterpenoid pathway and ascorbate and alderete metabolism are not listed.

Strengths:

This is an interesting manuscript that could make an important contribution to our understanding of the roles of micro RNAs at metamorphosis, and potentially of how sex-specific differences arise during metamorphosis. Strengths of the paper include the functional validation of microRNA binding, in vitro and in vivo-, as well as the characterization of sesquiterpenoid and ecdysteroid titers. The authors have also used CRISPR to generate specific knock-outs of miR-34 and miR-277. The transcriptomes will be a resource for future work to mine for differences in gene expression during metamorphosis.

Weaknesses:

(1) Spatial Expression of miR-34 and miR-277. If miR-34 and miR-277 regulate AstC-R1 and Kr-h1, then they must be expressed in the same cells. Although the authors show that the microRNAs do bind to the transcripts of AstC-R1 and Kr-h1 in S2 cells, and miR-277 binds AstC-R1 and Kr-h1 in vivo whole-animal homogenates, we do not know if the microRNAs are ever in the cells where AstC-R1 or Kr-h1 are expressed. AstC-R1 is only expressed in a few cells in the brain, so it is not at all certain that it is co-expressed with either microRNA. The creation of enhancer lines or in situ hybridization in Drosophila is straightforward and would sort this out.

(2) Phenotypes. Although a double deletion was used and specific knock-outs of both miR-34 and miR-277 were generated, the analysis of the mutants is very superficial. For the homozygous deletion of both microRNAs miR-34 and miR-277, only a decrease in survivorship was observed a full six days after adult eclosion - after the end of metamorphosis. No phenotype for either miR-34KO or miR-277-KO was given. The authors cite the work of others who have found specific phenotypes after manipulation of sesquiterpenoids or ecdysteroids, like Riddiford and Ashburner, but do not use any of these many studies to help them characterize the phenotype. If the loss of miR-34 and miR-277 affects so many pathways (including MAPK signaling, TGF-beta signaling, FoxO signaling, and Wnt signaling), as well as global titers of metamorphic hormones, then there shouldn't there be something different in the development to discuss?

(3) I think the reliance on GO term enrichment is getting in the way of biology. For instance, I would not describe Kr-h1 as a sesquiterpenoid biosynthesis pathway gene. Yet the authors say they were motivated to examine microRNA regulation of Kr-h1 because they saw differences in levels of the sesquiterpenoid biosynthesis pathway between WL3 and WPP, a period which also saw differences in expression of some microRNAs. I understand that Kr-h1 expression is regulated by JH, a sesquiterpenoid, but it is not directly involved with JH production, so relying on GO term enrichment has made the decision to focus on Kr-h1 feel arbitrary.

(4) The transcriptomes of miR-34 and miR-277 should have revealed genes encoding members of the sesquiterpenoid biosynthesis pathway as well as AstC-R1 and Kr-h1, but neither was mentioned. The functional tests of miR-34 and miR-277 were performed because they were shown to affect the levels of expression of genes in the sesquiterpenoid biosynthesis pathway. Figure 2 shows a significant decrease in AstC-R1 and Kr-h1 transcripts after the loss of miR-34 and miR-277. However, the results do not mention either (Lines 250-264). Instead, there is a list of 10 different GO terms (like arginine and proline metabolism or fatty acid degradation) that were enriched in miR-34 and miR-277 transcriptomes. If any of those ten types have any relationship to Kr-h1, AstC-R1, or metamorphosis, that has not been explained.

(5) Not enough care was taken in describing the stages. The methods describe wandering larvae (WL3) and white pre-pupa (WPP) for the transcriptomes, but in the text, different terms are used, like "larva", "pupa" and "L3 larvae instars" "early pupae" "late L3". Also, it seems like the small RNA libraries for sequencing were taken from "L3 larvae", but the stage of the L3 larvae was not mentioned. Staging is important, especially during metamorphosis, since differences in expression are expected to exist between different stages of L3, between early vs late wandering, and between WPP and early pupal stages.

Reviewer #2 (Public Review):

Summary:

This study proposes that the microRNA cluster miR-277/34 controls the generation of sexual dimorphism in Drosophila melanogaster during metamorphosis by acting on specific hormonal and developmental gene pathways.

Strengths:

Using a combination of mRNA and small RNA sequencing together with genome-wide in silico and in vitro analyses the authors identified a microRNA cluster that may be involved in metamorphosis and the generation of sexual dimorphism in Drosophila melanogaster.

Weaknesses:

Biological validation of the identified sexually dimorphic genes and a detailed understanding of how the microRNA cluster miR-277/34 might be involved in the regulation of sesquiterpenoids are needed.

Major suggestions:

(1) If AstC-R1 and Kr-h1 are targets of the miR-277/34 cluster and cause their downregulation, it is not clear why there would also be a decrease in the levels of these genes in the miR-277/34 mutants. This would suggest that the mechanism is not straightforward and that further epistatic experiments should be carried out in order to clarify this issue.

(2) The changes in the expression levels of AstC-R1 in pupae of miR-277-KO and mir-34-KO flies must be accompanied by photos of the respective larvae and pupae, as well as an analysis of the larvae-pupa transition on the mutants by gender.

(3) Biological validation of the identified sexually dimorphic genes in vivo will be necessary for the support of this work.

Reviewer #3 (Public Review):

Summary:

The authors show convincingly the complexity of gene up- and down-regulation at the outset of metamorphosis and identify substantial differences between the two sexes, even at this early time in development. The complexity of microRNA expression and the difference between the sexes are also nicely laid out. The functional significance of these differences, though, is harder to establish. The authors have focused on the roles of two families of developmental hormones, the ecdysteroids, and the juvenile hormones. The emergence of sex-specific differentiation of organs during metamorphosis is clearly downstream of the action of ecdysteroids and/or JH, but there is no evidence that the presence or lack of these hormones has any effect on the sexual identity of organ systems - i.e., that manipulations of JH or ecdysteroid result in either the masculinization or feminization of individuals or their organs. The precedence for the linkage of these hormones to sex determination is the 2002, Belgacem & Martin study, which describes the effects of JH on fly locomotion. These authors show that the number of stop/start bouts is sexually dimorphic, and removal of JH in males shifts their frequency into the female range while giving treated males exogenous JH moves it back. While this is referred to as a "feminization" of male behavior, this quantitative shift in frequency is not as compelling as would be a qualitative shift -- for example, the removal of JH causing males to show egg-laying behavior (a result that has never been seen). Also, these effects are in a fully mature system, rather than at the early metamorphic time examined in the present paper. In driving and coordination metamorphosis, JH and ecdysteroids are intimately involved in sexual differentiation, but I know of no compelling evidence that they play a role in sex determination.

While the summary of the effects or removal of specific microRNAs on the components of the biosynthetic pathway for the JHs and ecdysteroids (Figure 2E ,F) is quite compelling, I am concerned about the effects of the removal of mir-277 and mir-34 on the levels of both the JHs and 20E. My concern centers around the data from the control group (w[1118] animals in Figure 2D). These data are the first report of a marked sex difference in the titer of either JH or ecdysteroid at the start of metamorphosis in Drosophila. As expected, males show a 10-20 increase in levels of JH III, JHB3, and 20E between the L3 stage and the white puparium, but, surprisingly, the levels of these hormones in female L3 larvae are equal to or greater than that seen at pupariation! These data for females run counter to over 50 years of work on the effects of ecdysteroids in Drosophila!

As far as I can gather from the paper, the L3 data were obtained using wandering larvae. This stage lasts for about 12 hours and ends with pupariation. Larvae from this period need to be used with caution for hormone studies. Levels of both JH and ecdysteroid are low as larvae leave the food but rapidly rise to their peak levels at the white puparium stage 12 hours later. To deal with the rapidly changing hormonal landscape through this period, the researchers have used physiological markers to track this progression. Initially, it was the sage of "puffing" of the giant salivary gland chromosomes, but, for bulk collection of staged larvae, larvae are fed on food containing a blue dye, and progression is tracked by the loss of blue coloring from the gut. I could not find if the authors had any criteria for selecting larvae during the wandering period. Male and female larvae grow to different sizes. Might this difference in growth be biased when larvae were selected during their wandering phase?

The other hormone-related issue is the expression of Kr-h1 during larval stages and metamorphosis (Figure 1G). Kr-h1 is the main target of JH and Kr-h1 expression is often used as a proxy for the JH titer. The authors report that peak Kr-h1 expression occurs in the L3 (when the JH titer should be lowest!) and that it drops at wandering. This pattern is counter to that reported in the literature (e.g., FlyBase, ModEncode).

  1. Howard Hughes Medical Institute
  2. Wellcome Trust
  3. Max-Planck-Gesellschaft
  4. Knut and Alice Wallenberg Foundation