Genome-wide mapping of native co-localized G4s and R-loops in living cells

  1. Key Laboratory of Birth Defects and Related Disease of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, Sichuan, 610041, China
  2. Department of Hematology and Institute of Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China

Peer review process

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

Read more about eLife’s peer review process.

Editors

  • Reviewing Editor
    Andrés Aguilera
    CABIMER, Universidad de Sevilla, Seville, Spain
  • Senior Editor
    Murim Choi
    Seoul National University, Seoul, Korea, the Republic of

Reviewer #1 (Public Review):

Summary:

Non-B DNA structures such as G4s and R-loops have the potential to impact genome stability, gene transcription, and cell differentiation. This study investigates the distribution of G4s and R-loops in human and mouse cells using some interesting technical modifications of existing Tn5-based approaches. This work confirms that the helicase DHX9 could regulate the formation and/or stability of both structures in mouse embryonic stem cells (mESCs). It also provides evidence that the lack of DHX9 in mESCs interferes with their ability to differentiate.

Strengths:

HepG4-seq, the new antibody-free strategy to map G4s based on the ability of Hemin to act as a peroxidase when complexed to G4s, is interesting. This study also provides more evidence that the distribution pattern of G4s and R-loops might vary substantially from one cell type to another.

Weaknesses:

This study is essentially descriptive and does not provide conclusive evidence that lack of DHX9 does interfere with the ability of mESCs to differentiate by regulating directly the stability of either G4 or R-loops. In the end, it does not substantially improve our understanding of DHX9's mode of action.

There is no in-depth comparison of the newly generated data with existing datasets and no rigorous control was presented to test the specificity of the hemin-G4 interaction (a lot of the hemin-dependent signal seems to occur in the cytoplasm, which is unexpected).

The authors talk about co-occurrence between G4 and R-loops but their data does not actually demonstrate co-occurrence in time. If the same loci could form alternatively either R-loops or G4 and if DHX9 was somehow involved in determining the balance between G4s and R-loops, the authors would probably obtain the same distribution pattern. To manipulate R-loop levels in vivo and test how this affects HEPG4-seq signals would have been helpful.

This study relies exclusively on Tn5-based mapping strategies. This is a problem as global changes in DNA accessibility might strongly skew the results. It is unclear at this stage whether the lack of DHX9, BLM, or WRN has an impact on DNA accessibility, which might underlie the differences that were observed. Moreover, Tn5 cleaves DNA at a nearby accessible site, which might be at an unknown distance away from the site of interest. The spatial accuracy of Tn5-based methods is therefore debatable, which is a problem when trying to demonstrate spatial co-occurrence. Alternative mapping methods would have been helpful.

Reviewer #2 (Public Review):

Summary:

In this study, Liu et al. explore the interplay between G-quadruplexes (G4s) and R-loops. The authors developed novel techniques, HepG4-seq and HBD-seq, to capture and map these nucleic acid structures genome-wide in human HEK293 cells and mouse embryonic stem cells (mESCs). They identified dynamic, cell-type-specific distributions of co-localized G4s and R-loops, which predominantly localize at active promoters and enhancers of transcriptionally active genes. Furthermore, they assessed the role of helicase Dhx9 in regulating these structures and their impact on gene expression and cellular functions.

The manuscript provides a detailed catalogue of the genome-wide distribution of G4s and R-loops. However, the conceptual advance and the physiological relevance of the findings are not obvious. Overall, the impact of the work on the field is limited to the utility of the presented methods and datasets.

Strengths:

(1) The development and optimization of HepG4-seq and HBD-seq offer novel methods to map native G4s and R-loops.

(2) The study provides extensive data on the distribution of G4s and R-loops, highlighting their co-localization in human and mouse cells.

(3) The study consolidates the role of Dhx9 in modulating these structures and explores its impact on mESC self-renewal and differentiation.

Weaknesses:

(1) The specificity of the biotinylation process and potential off-target effects are not addressed. The authors should provide more data to validate the specificity of the G4-hemin.

(2) Other methods exploring a catalytic dead RNAseH or the HBD to pull down R-loops have been described before. The superior quality of the presented methods in comparison to existing ones is not established. A clear comparison with other methods (BG4 CUT&Tag-seq, DRIP-seq, R-CHIP, etc) should be provided.

(3) Although the study demonstrates Dhx9's role in regulating co-localized G4s and R-loops, additional functional experiments (e.g., rescue experiments) are needed to confirm these findings.

(4) The manuscript would benefit from a more detailed discussion of the broader implications of co-localized G4s and R-loops.

(5) The manuscript lacks appropriate statistical analyses to support the major conclusions.

(6) The discussion could be expanded to address potential limitations and alternative explanations for the results.

Reviewer #3 (Public Review):

Summary:

The authors developed and optimized the methods for detecting G4s and R-loops independent of BG4 and S9.6 antibody, and mapped genomic native G4s and R-loops by HepG4-seq and HBD-seq, revealing that co-localized G4s and R-loops participate in regulating transcription and affecting the self-renewal and differentiation capabilities of mESCs.

Strengths:

By utilizing the peroxidase activity of G4-hemin complex and combining proximity labeling technology, the authors developed HepG4-seq (high throughput sequencing of hemin-induced proximal labelled G4s) , which can detect the dynamics of G4s in vivo. Meanwhile, the "GST-His6-2xHBD"-mediated CUT&Tag protocol (Wang et al., 2021) was optimized by replacing fusion protein and tag, the optimized HBD-seq avoids the generation of GST fusion protein aggregates and can reflect the genome-wide distribution of R-loops in vivo.

The authors employed HepG4-seq and HBD-seq to establish comprehensive maps of native co-localized G4s and R-loops in human HEK293 cells and mouse embryonic stem cells (mESCs). The data indicate that co-localized G4s and R-loops are dynamically altered in a cell type-dependent manner and are largely localized at active promoters and enhancers of transcriptionally active genes.

Combined with Dhx9 ChIP-seq and co-localized G4s and R-loops data in wild-type and dhx9KO mESCs, the authors confirm that the helicase Dhx9 is a direct and major regulator that regulates the formation and resolution of co-localized G4s and R-loops.

Depletion of Dhx9 impaired the self-renewal and differentiation capacities of mESCs by altering the transcription of co-localized G4s and R-loops-associated genes.

In conclusion, the authors provide an approach to studying the interplay between G4s and R-loops, shedding light on the important roles of co-localized G4s and R-loops in development and disease by regulating the transcription of related genes.

Weaknesses:

As we know, there are at least two structure data of S9.6 antibody very recently, and the questions about the specificity of the S9.6 antibody on RNA:DNA hybrids should be finished. The authors referred to (Hartono et al., 2018; Konig et al., 2017; Phillips et al., 2013) need to be updated, and the authors' bias against S9.6 antibodies needs also to be changed. However, as the authors had questioned the specificity of the S9.6 antibody, they should compare it in parallel with the data they have and the data generated by the widely used S9.6 antibody.

Although HepG4-seq is an effective G4s detection technique, and the authors have also verified its reliability to some extent, given the strong link between ROS homeostasis and G4s formation, and hemin's affinity for different types of G4s, whether HepG4-seq reflects the dynamics of G4s in vivo more accurately than existing detection techniques still needs to be more carefully corroborated.

Author response:

eLife assessment

This useful study describes an antibody-free method to map G-quadruplexes (G4s) in vertebrate cells. While the method might have potential, the current analysis is primarily descriptive and does not add substantial new insights beyond existing data (e.g., PMID:34792172). While the datasets provided might constitute a good starting point for future functional studies, additional data and analyses would be needed to fully support the major conclusions and, at the same time, clarify the advantage of this method over other methods. Specifically, the strength of the evidence for DHX9 interfering with the ability of mESCs to differentiate by regulating directly the stability of either G4s or R-loops is still incomplete.

We thank the editors for their helpful comments.

Given that antibody-based methods have been reported to leave open the possibility of recognizing partially folded G4s and promoting their folding, we have employed the peroxidase activity of the G4-hemin complex to develop a new method for capturing endogenous G4s that significantly reduces the risk of capturing partially folded G4s. We will be happy to clarify the advantage of our method.

In the Fig. 7, we applied the Dhx9 CUT&Tag assay to identify the G4s and R-loops directly bound by Dhx9 and further characterized the differential Dhx9-bound G4s and R-loops in the absence of Dhx9. Dhx9 is a versatile helicase capable of directly resolving R-loops and G4s or promoting R-loop formation (PMID: 21561811, 30341290, 29742442, 32541651, 35905379, 34316718). Furthermore, we showed that depletion of Dhx9 significantly altered the levels of G4s or R-loops around the TSS or gene bodies of several key regulators of mESC and embryonic development, such as Nanog, Lin28a, Bmp4, Wnt8a, Gata2, and Lef1, and also their RNA levels (Fig.7 I). The above evidence is sufficient to support the transcriptional regulation of mESCs cell fate by directly modulating the G4s or R-loops within the key regulators of mESCs.

Public Reviews:

Reviewer #1 (Public Review):

Summary:

Non-B DNA structures such as G4s and R-loops have the potential to impact genome stability, gene transcription, and cell differentiation. This study investigates the distribution of G4s and R-loops in human and mouse cells using some interesting technical modifications of existing Tn5-based approaches. This work confirms that the helicase DHX9 could regulate the formation and/or stability of both structures in mouse embryonic stem cells (mESCs). It also provides evidence that the lack of DHX9 in mESCs interferes with their ability to differentiate.

Strengths:

HepG4-seq, the new antibody-free strategy to map G4s based on the ability of Hemin to act as a peroxidase when complexed to G4s, is interesting. This study also provides more evidence that the distribution pattern of G4s and R-loops might vary substantially from one cell type to another.

We appreciate your valuable points.

Weaknesses:

This study is essentially descriptive and does not provide conclusive evidence that lack of DHX9 does interfere with the ability of mESCs to differentiate by regulating directly the stability of either G4 or R-loops. In the end, it does not substantially improve our understanding of DHX9's mode of action.

In this study, we aimed to report new methods for capturing endogenous G4s and R-loops in living cells. Dhx9 has been reported to directly unwind R-loops and G4s or promote R-loop formation (PMID: 21561811, 30341290, 29742442, 32541651, 35905379, 34316718). To understand the direct Dhx9-bound G4s and R-loops, we performed the Dhx9 CUT&Tag assay and analyzed the co-localization of Dhx9-binding sites and G4s or R-loops. We found that 47,857 co-localized G4s and R-loops are directly bound by Dhx9 in the wild-type mESCs and 4,060 of them display significantly differential signals in absence of Dhx9, suggesting that redundant regulators exist as well. We showed that depletion of Dhx9 significantly altered the RNA levels of several key regulators of mESC and embryonic development, such as Nanog, Lin28a, Bmp4, Wnt8a, Gata2, and Lef1, which coincides with the significantly differential levels of G4s or R-loops around the TSS or gene bodies of these genes (Fig.7). The comprehensive molecular mechanism of Dhx9 action is indeed not the focus of this study. We will work on it in the future studies. Thank you for the comments.

There is no in-depth comparison of the newly generated data with existing datasets and no rigorous control was presented to test the specificity of the hemin-G4 interaction (a lot of the hemin-dependent signal seems to occur in the cytoplasm, which is unexpected).

The specificity of hemin-G4-induced peroxidase activity and self-biotinylation has been well demonstrated in previous studies (PMID: 19618960, 22106035, 28973477, 32329781). In the Fig.1A, we compared the hemin-G4-induced biotinylation levels in different conditions. Cells treated with hemin and Bio-An exhibited a robust fluorescence signal, while the absence of either hemin or Bio-An almost completely abolished the biotinylation signals, suggesting a specific and active biotinylation activity. To identify the specific signals, we have included the non-label control and used this control to call confident HepG4 peaks in all HepG4-seq assays.

The hemin-RNA G4 complex has also been reported to have mimic peroxidase activity and trigger similar self-biotinylation signals as DNA G4s (PMID: 32329781, 31257395, 27422869). Therefore, it is not surprising to observe hemin-dependent signals in the cytoplasm generated by cytoplasmic RNA G4s.

In the revised version, we will include careful comparison between our data and previous datasets.

The authors talk about co-occurrence between G4 and R-loops but their data does not actually demonstrate co-occurrence in time. If the same loci could form alternatively either R-loops or G4 and if DHX9 was somehow involved in determining the balance between G4s and R-loops, the authors would probably obtain the same distribution pattern. To manipulate R-loop levels in vivo and test how this affects HEPG4-seq signals would have been helpful.

Single-molecule fluorescence studies have shown the existence of a positive feedback mechanism of G4 and R-loop formation during transcription (PMID: 32810236, 32636376), suggesting that G4s and Rloops could co-localize at the same molecule. Dhx9 is a versatile helicase capable of directly resolving R-loops and G4s or promoting R-loop formation (PMID: 21561811, 30341290, 29742442, 32541651, 35905379, 34316718). Although depletion of Dhx9 resulted in 6,171 Dhx9-bound co-localized G4s and R-loops with significantly altered levels of G4s or R-loops, only 276 of them (~4.5%) harbored altered G4s and R-loops, suggesting that the interacting G4s and R-loops are rare in living cells. Nowadays, the genome-wide co-occurrence of two factors are mainly obtained by bioinformatically intersection analysis. We agreed that the heterogenous distribution between cells will give false positive co-occurrence patterns. We will carefully discuss this point in the revised version. At the same time, we will make efforts to develop a new method to map the co-localized G4 and R-loop in the same molecule in the future study.

This study relies exclusively on Tn5-based mapping strategies. This is a problem as global changes in DNA accessibility might strongly skew the results. It is unclear at this stage whether the lack of DHX9, BLM, or WRN has an impact on DNA accessibility, which might underlie the differences that were observed. Moreover, Tn5 cleaves DNA at a nearby accessible site, which might be at an unknown distance away from the site of interest. The spatial accuracy of Tn5-based methods is therefore debatable, which is a problem when trying to demonstrate spatial co-occurrence. Alternative mapping methods would have been helpful.

In this study, we used the recombinant streptavidin monomer and anti-GP41 nanobody fusion protein (mSA-scFv) to specifically recognize hemin-G4-induced biotinylated G4 and then recruit the recombinant GP41-tagged Tn5 protein to these G4s sites. Similarly, the recombinant V5-tagged N-terminal hybrid-binding domain (HBD) of RNase H1 specifically recognizes R-loops and recruit the recombinant protein G-Tn5 (pG-Tn5) with the help of anti-V5 antibody. Therefore, the spatial distance of Tn5 to the target sites is well controlled and very short, and also the recruitment of Tn5 is specifically determined by the existence of G4s in HepG4-seq and R-loops in HBD-seq.

Reviewer #2 (Public Review):

Summary:

In this study, Liu et al. explore the interplay between G-quadruplexes (G4s) and R-loops. The authors developed novel techniques, HepG4-seq and HBD-seq, to capture and map these nucleic acid structures genome-wide in human HEK293 cells and mouse embryonic stem cells (mESCs). They identified dynamic, cell-type-specific distributions of co-localized G4s and R-loops, which predominantly localize at active promoters and enhancers of transcriptionally active genes. Furthermore, they assessed the role of helicase Dhx9 in regulating these structures and their impact on gene expression and cellular functions.

The manuscript provides a detailed catalogue of the genome-wide distribution of G4s and R-loops. However, the conceptual advance and the physiological relevance of the findings are not obvious. Overall, the impact of the work on the field is limited to the utility of the presented methods and datasets.

Strengths:

(1) The development and optimization of HepG4-seq and HBD-seq offer novel methods to map native G4s and R-loops.

(2) The study provides extensive data on the distribution of G4s and R-loops, highlighting their co-localization in human and mouse cells.

(3) The study consolidates the role of Dhx9 in modulating these structures and explores its impact on mESC self-renewal and differentiation.

We appreciate your valuable points.

Weaknesses:

(1) The specificity of the biotinylation process and potential off-target effects are not addressed. The authors should provide more data to validate the specificity of the G4-hemin.

The specificity of hemin-G4-induced peroxidase activity and self-biotinylation has been well demonstrated in previous studies (PMID: 19618960, 22106035, 28973477, 32329781). In the Fig.1A, we compared the hemin-G4-induced biotinylation levels in different conditions. Cells treated with hemin and Bio-An exhibited a robust fluorescence signal, while the absence of either hemin or Bio-An almost completely abolished the biotinylation signals, suggesting a specific and active biotinylation activity.

(2) Other methods exploring a catalytic dead RNAseH or the HBD to pull down R-loops have been described before. The superior quality of the presented methods in comparison to existing ones is not established. A clear comparison with other methods (BG4 CUT&Tag-seq, DRIP-seq, R-CHIP, etc) should be provided.

Thank you for the suggestions. We will include the comparisons in the revised version.

(3) Although the study demonstrates Dhx9's role in regulating co-localized G4s and R-loops, additional functional experiments (e.g., rescue experiments) are needed to confirm these findings.

Dhx9 has been demonstrate as a versatile helicase capable of directly resolving R-loops and G4s or promoting R-loop formation in previous studies (PMID: 21561811, 30341290, 29742442, 32541651, 35905379, 34316718). We believe that the current new dataset and previous studies are enough to support the capability of Dhx9 in regulating co-localized G4s and R-loops.

(4) The manuscript would benefit from a more detailed discussion of the broader implications of co-localized G4s and R-loops.

Thank you for the suggestions. We will include a more detailed discussion in the revised version.

(5) The manuscript lacks appropriate statistical analyses to support the major conclusions.

We apologized for this point. Whereas we have applied careful statistical analyses in this study, lacking of some statistical details make people hard to understand some conclusions. We will carefully add details of all statistical analysis.

(6) The discussion could be expanded to address potential limitations and alternative explanations for the results.

Thank you for the suggestions. We will include a more detailed discussion about this point in the revised version.

Reviewer #3 (Public Review):

Summary:

The authors developed and optimized the methods for detecting G4s and R-loops independent of BG4 and S9.6 antibody, and mapped genomic native G4s and R-loops by HepG4-seq and HBD-seq, revealing that co-localized G4s and R-loops participate in regulating transcription and affecting the self-renewal and differentiation capabilities of mESCs.

Strengths:

By utilizing the peroxidase activity of G4-hemin complex and combining proximity labeling technology, the authors developed HepG4-seq (high throughput sequencing of hemin-induced proximal labelled G4s), which can detect the dynamics of G4s in vivo. Meanwhile, the "GST-His6-2xHBD"-mediated CUT&Tag protocol (Wang et al., 2021) was optimized by replacing fusion protein and tag, the optimized HBD-seq avoids the generation of GST fusion protein aggregates and can reflect the genome-wide distribution of R-loops in vivo.

The authors employed HepG4-seq and HBD-seq to establish comprehensive maps of native co-localized G4s and R-loops in human HEK293 cells and mouse embryonic stem cells (mESCs). The data indicate that co-localized G4s and R-loops are dynamically altered in a cell type-dependent manner and are largely localized at active promoters and enhancers of transcriptionally active genes.

Combined with Dhx9 ChIP-seq and co-localized G4s and R-loops data in wild-type and dhx9KO mESCs, the authors confirm that the helicase Dhx9 is a direct and major regulator that regulates the formation and resolution of co-localized G4s and R-loops.

Depletion of Dhx9 impaired the self-renewal and differentiation capacities of mESCs by altering the transcription of co-localized G4s and R-loops-associated genes.

In conclusion, the authors provide an approach to studying the interplay between G4s and R-loops, shedding light on the important roles of co-localized G4s and R-loops in development and disease by regulating the transcription of related genes.

We appreciate your valuable points.

Weaknesses:

As we know, there are at least two structure data of S9.6 antibody very recently, and the questions about the specificity of the S9.6 antibody on RNA:DNA hybrids should be finished. The authors referred to (Hartono et al., 2018; Konig et al., 2017; Phillips et al., 2013) need to be updated, and the authors' bias against S9.6 antibodies needs also to be changed. However, as the authors had questioned the specificity of the S9.6 antibody, they should compare it in parallel with the data they have and the data generated by the widely used S9.6 antibody.

Thank you for the updating information about the structure data of S9.6 antibody. We politely disagree the specificity of the S9.6 antibody on RNA:DNA hybrids. The structural studies of S9.6 (PMID: 35347133, 35550870) used only one RNA:DNA hybrid to show the superior specificity of S9.6 on RNA:DNA hybrid than dsRNA and dsDNA. However, Fabian K. et al has reported that the binding affinities of S9.6 on RNA:DNA hybrid exhibits obvious sequence-dependent bias from null to nanomolar range (PMID: 28594954). We will include the comparison between S9.6-derived data and our HBD-seq data in the revised version.

Although HepG4-seq is an effective G4s detection technique, and the authors have also verified its reliability to some extent, given the strong link between ROS homeostasis and G4s formation, and hemin's affinity for different types of G4s, whether HepG4-seq reflects the dynamics of G4s in vivo more accurately than existing detection techniques still needs to be more carefully corroborated.

Thank you for pointing out this issue. In the in vitro hemin-G4 induced self-biotinylation assay, parallel G4s exhibit higher peroxidase activities than anti-parallel G4s. Thus, the dynamics of G4 conformation could affect the HepG4-seq signals (PMID: 32329781). In the future, people may need to combine HepG4-seq and BG4s-eq to carefully explain the endogenous G4s. We will carefully discuss this point in the revised version.

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