Human DCP1 is crucial for mRNA decapping and possesses paralog-specific gene regulating functions

  1. Institute of Bioinformatics and Systems Biology, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
  2. Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu, 30068, Taiwan
  3. Center For Intelligent Drug Systems and Smart Bio-devices (IDS2B), National Yang Ming Chiao Tung University, Hsinchu, Taiwan
  4. Department of Pharmacy, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
  5. Department of Biochemistry, Max Planck Institute for Developmental Biology, Max-Planck-Ring 5, 72076 Tübingen, Germany
  6. Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei 112, Taiwan

Editors

  • Reviewing Editor
    Paul Donlin-Asp
    University of Edinburgh, Edinburgh, United Kingdom
  • Senior Editor
    Jonathan Cooper
    Fred Hutchinson Cancer Research Center, Seattle, United States of America

Reviewer #1 (Public Review):

Summary & Assessment:

The catalytic core of the eukaryotic decapping complex consists of the decapping enzyme DCP2 and its key activator DCP1. In humans, there are two paralogs of DCP1, DCP1a, and DCP1b, that are known to interact with DCP2 and recruit additional cofactors or coactivators to the decapping complex; however, the mechanisms by which DCP1 activates decapping and the specific roles of DCP1a versus DCP1b, remain poorly defined. In this manuscript, the authors used CRISPR/Cas9-generated DCP1a/b knockout cells to begin to unravel some of the differential roles of human DCP1a and DCP1b in mRNA decapping, gene regulation, and cellular metabolism. While this manuscript presents some new and interesting observations on human DCP1 (e.g. human DCP1a/b KO cells are viable and can be used to investigate DCP1 function; only the EVH1 domain, and not its disordered C-terminal region which recruits many decapping cofactors, is apparently required for efficient decapping in cells; DCP1a and b target different subsets of mRNAs for decay and may regulate different aspects of metabolism), there are several major issues that undercut some of the main conclusions of the paper, and some key claims that are incompletely or inconsistently supported by the presented data.

Strengths & well-supported claims:

• Through in vivo tethering assays in CRISPR/Cas9-generated DCP1a/b knockout cells, the authors show that DCP1 depletion leads to significant defects in decapping and the accumulation of capped, deadenylated mRNA decay intermediates.

• DCP1 truncation experiments reveal that only the EVH1 domain of DCP1 is necessary to rescue decapping defects in DCP1a/b KO cells.

• RNA and protein immunoprecipitation experiments suggest that DCP1 acts as a scaffold to help recruit multiple decapping cofactors to the decapping complex (e.g. EDC3, DDX6, PATL1 PNRC1, and PNRC2), but that none of these cofactors are essential for DCP2-mediated decapping in cells.

• The authors investigated the differential roles of DCP1a and DCP1b in gene regulation through transcriptomic and metabolomic analysis and found that these DCP1 paralogs target different mRNA transcripts for decapping and have different roles in cellular metabolism and their apparent links to human cancers. (Although I will note that I can't comment on the experimental details and/or rigor of the transcriptomic and metabolomic analyses, as these are outside my expertise.)

Weaknesses & incompletely supported claims:

  1. A central mechanistic claim of the paper is that "DCP1a can regulate DCP2's cellular decapping activity by enhancing DCP2's affinity to RNA, in addition to bridging the interactions of DCP2 with other decapping factors. This represents a pivotal molecular mechanism by which DCP1a exerts its regulatory control over the mRNA decapping process." Similar versions of this claim are repeated in the abstract and discussion sections. However, this appears to be entirely at odds with the observation from in vitro decapping assays with immunoprecipitated DCP2 that showed DCP1 knockout does not significantly affect the enzymatic activity of DCP2 (Figures 2B-D; I note that there may be a very small change in DCP2 activity shown in panel C, but this may be due to slightly different amounts of immunoprecipitated DCP2 used in the assay, as suggested by panel D). If DCP1 pivotally regulates decapping activity by enhancing RNA binding to DCP2, why is no difference in decapping activity observed in the absence of DCP1? Furthermore, the authors show only weak changes in relative RNA levels immunoprecipitated by DCP2 with versus without DCP1 (~2-3 fold change; consistent with the Valkov 2016 NSMB paper, which shows what looks like only modest changes in RNA binding affinity for yeast Dcp2 +/- Dcp1). Is the argument that only a 2-3 fold change in RNA binding affinity is responsible for the sizable decapping defects and significant accumulation of deadenylated intermediates observed in cells upon Dcp1 depletion? (and if so, why is this the case for in-cell data, but not the immunoprecipitated in vitro data?)

The authors acknowledge this apparent discrepancy between the in vitro DCP2 decapping assays and in-cell decapping data, writing: "this observation could be attributed to the inherent constraints of in vitro assays, which often fall short of faithfully replicating the complexity of the cellular environment where multiple factors and cofactors are at play. To determine the underlying cause, we postulated that the observed cellular decapping defect in DCP1a/b knockout cells might be attributed to DCP1 functioning as a scaffold." This is fair. They next show that DCP1 acts as a scaffold to recruit multiple factors to DCP2 in cells (EDC3, DDX6, PatL1, and PNRC1 and 2). However, while DCP1 is shown to recruit multiple cofactors to DCP2 (consistent with other studies in the decapping field, and primarily through motifs in the Dcp1 C-terminal tail), the authors ultimately show that *none* of these cofactors are actually essential for DCP2-mediated decapping in cells (Figures 3A-F). More specifically, the authors showed that the EVH1 domain was sufficient to rescue decapping defects in DCP1a/b knockout cells, that PNRC1 and PNRC2 were the only cofactors that interact with the EVH1 domain, and finally that shRNA-mediated PNRC1 or PNCR2 knockdown has no effect on in-cell decapping (Figures 3E and F). Therefore, based on the presented data, while DCP1 certainly does act as a scaffold, it doesn't seem to be the case that the major cellular decapping defect observed in DCP1a/b knockout is due to DCP1's ability to recruit specific cofactors to DCP2.

So as far as I can tell, the discrepancy between the in vitro (DCP1 not required) and in-cell (DCP1 required) decapping data, remains entirely unresolved. Therefore, I don't think that the conclusions that DCP1 regulates decapping by (a) changing RNA binding affinity (authors show this doesn't matter in vitro, and that the change in RNA binding affinity is very small) or (b) by bridging interactions of cofactors with DCP2 (authors show all tested cofactors are dispensable for robust in-cell decapping activity), are supported by the evidence presented in the paper (or convincingly supported by previous structural and functional studies of the decapping complex).

  1. Related to the RNA binding claims mentioned above, are the differences shown in Figure 3H statistically significant? Why are there no error bars shown for the MBP control? (I understand this was normalized to 1, but presumably, there were 3 biological replicates here that have some spread of values?). The individual data points for each replicate should be displayed for each bar so that readers can better assess the spread of data and the significance of the observed differences. I've listed these points as major because of the key mechanistic claim that DCP1 enhances RNA binding to DCP2 hinges in large part on this data.

  2. Also related to point (1) above, the kinetic analysis presented in Figure 2C shows that the large majority of transcript is mostly decapped at the first 5-minute timepoint; it may be that DCP2-mediated decapping activity is actually different in vitro with or without DCP1, but that this is being missed because the reaction is basically done in less than 5 minutes under the conditions being assayed (i.e. these are basically endpoint assays under these conditions). It may be that if kinetics were done under conditions to slow down the reaction somewhat (e.g. lower Dcp2 concentration, lower temperatures), so that more of the kinetic behavior is captured, the apparent discrepancy between in vitro and in-cell data would be much less. Indeed, previous studies have shown that in yeast, Dcp1 strongly activates the catalytic step (kcat) of decapping by ~10-fold, and reduces the KM by only ~2 fold (Floor et al, NSMB 2010). It might be beneficial to use purified proteins here (only a Western blot is used in Figure 2D to show the presence of DCP2 and/or DCP1, but do these complexes have other, and different, components immunoprecipitated along with them?), if possible, to better control reaction conditions.

This contradiction between the in vitro and in-cell decapping data undercuts one of the main mechanistic takeaways from the first half of the paper. This needs to be addressed/resolved with further experiments to better define the role of DCP1-mediated activation, or the mechanistic conclusions significantly changed or removed.

  1. The second half of the paper compares the transcriptomic and metabolic profiles of DCP1a versus DCP1b knockouts to reveal that these target a different subset of mRNAs for degradation and have different levels of cellular metabolites. This is a great application of the DCP1a/b KO cells developed in this paper and provides new information about DCP1a vs b function in metazoans, which to my knowledge has not really been explored at all. However, the analysis of DCP1 function/expression levels in human cancer seems superficial and inconclusive: for example, the authors conclude that "...these findings indicate that DCP1a and DCP1b likely have distinct and non-redundant roles in the development and progression of cancer", but what is the evidence for this? I see that DCP1a and b levels vary in different cancer cell types, but is there any evidence that these changes are actually linked to cancer development, progression, or tumorigenesis? If not, these broader conclusions should be removed.

  2. The authors used CRISPR-Cas9 to introduce frameshift mutations that result in premature termination codons in DCP1a/b knockout cells (verified by Sanger sequencing). They then use Western blotting with DCP1a or DCP1b antibodies to confirm the absence of DCP1 in the knockout cell lines. However, the DCP1a antibody used in this study (Sigma D5444) is targeted to the C-terminal end of DCP1a. Can the authors conclusively rule out that the CRISPR/Cas-generated mutations do not result in the production of truncated DCP1a that is just unable to be detected by the C-terminally targeted antibody? While it is likely the introduced premature termination codon in the DCP1a gene results in nonsense-mediated decay of the resulting transcript, this outcome is indeed supported by the knockout results showing large defects in cellular decapping which can be rescued by the addition of the EVH1 domain, it would be better to carefully validate the success of the DCP1a knockout and conclusively show no truncated DCP1a is produced by using N-terminally targeted DCP1a antibodies (as was the case for DCP1b).

Some additional minor comments:

• More information would be helpful on the choice of DCP1 truncation boundaries; why was 1-254 chosen as one of the truncations?
• Figure S2D is a pretty important experiment because it suggests that the observed deadenylated intermediates are in fact still capped; can a positive control be added to these experiments to show that removal of cap results in rapid terminator-mediated degradation?

Reviewer #2 (Public Review):

Summary:

Chen et al., investigate the role of DCP1 paralogs in regulating RNA decay in human tissue culture cells. They assess the impact of the absence of DCP1a and/or DCP1b on the interaction of DCP2 with mRNA and other members of the decapping complex. In vitro RNA decay assays were performed to demonstrate that DCP1a/b plays a minor role in DCP2-mediated decapping and decay. The impacts of DCP1a and/or DCP1b knockout on the transcriptome and metabolome were determined.

Strengths:

Analysis of RNA abundance and metabolite differences in human tissue culture cells lacking DCP1a and/or DCP1b was performed.

The protein-protein interactions between DCP2 and other members of the decapping machinery mediated by DCP1a and/or DCP1b were assessed.

The functional role of DCP1a and/or DCP1b in mediating mRNA decapping/decay in human tissue culture cell extracts was determined.

Human tissue culture cells lacking DCP1a and/or DCP1b appear to have altered metabolomes, however, the significance and meaning of these differences are not clear.

Weaknesses:

The direct targets of DCP1a and/or DCP1b were not determined as the analysis was restricted to RNA-seq to assess RNA abundance, which can be a result of direct or indirect regulation by DCP1a/b.

P-bodies appear to be larger in human cells lacking DCP1a and DCP1b but a lack of image quantification prevents this conclusion from being drawn.

The lack of details in the methodology and figure legends limit reader understanding.

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