RAG suppresses group 2 innate lymphoid cells

  1. Division of Allergy and Immunology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63130, USA
  2. Immunology & Inflammation Research Therapeutic Area, Sanofi, Cambridge, MA 02141, USA
  3. Kimberly and Eric J. Waldman Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
  4. Mark Lebwohl Center for Neuroinflammation and Sensation, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA
  5. Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
  6. Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
  7. Division of Dermatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63130, USA
  8. Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63130, USA
  9. Department of Life Science, College of Natural Sciences, Hanyang University, Seoul 04763, Republic of Korea
  10. Division of Rheumatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
  11. Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Immunology and Microbial Pathogenesis Program, Graduate School of Medical Sciences, Weill Cornell Medical College, New York, NY 10065, USA
  12. Allen Discovery Center for Neuroimmune Interactions, Icahn School of Medicine at Mount Sinai 10019

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
    Gabrielle Belz
    University of Queensland, Brisbane, Australia
  • Senior Editor
    Carla Rothlin
    Yale University, New Haven, United States of America

Reviewer #1 (Public Review):

The study starts with the notion that in an AD-like disease model, ILC2s in the Rag1 knock-out were expanded and contained relatively more IL-5+ and IL-13+ ILC2s. This was confirmed in the Rag2 knock-out mouse model.

By using a chimeric mouse model in which wild-type knock-out splenocytes were injected into irradiated Rag1 knock-out mice, it was shown that even though the adaptive lymphocyte compartment was restored, there were increased AD-like symptoms and increased ILC2 expansion and activity. Moreover, in the reverse chimeric model, i.e. injecting a mix of wild-type and Rag1 knock-out splenocytes into irradiated wild-type animals, it was shown that the Rag1 knock-out ILC2s expanded more and were more active. Therefore, the authors could conclude that the RAG1 mediated effects were ILC2 cell-intrinsic.

Subsequent fate-mapping experiments using the Rag1Cre;reporter mouse model showed that there were indeed RAGnaïve and RAGexp ILC2 populations within naïve mice. Lastly, the authors performed multi-omic profiling, using single-cell RNA sequencing and ATAC-sequencing, in which a specific gene expression profile was associated with ILC2. These included well-known genes but the authors notably also found expression of Ccl1 and Ccr8 within the ILC2. The authors confirmed their earlier observations that in the RAGexp ILC2 population, the Th2 regulome was more suppressed, i.e. more closed, compared to the RAGnaïve population, indicative of the suppressive function of RAG on ILC2 activity. I do agree with the authors' notion that the main weakness was that this study lacks the mechanism by which RAG regulates these changes in ILC2s.

The manuscript is very well written and easy to follow, and the compelling conclusions are well supported by the data. The experiments are meticulously designed and presented. I wish to commend the authors for the study's quality.

Even though the study is compelling and well supported by the presented data, some additional context could increase the significance:

(1) The presence of the RAGnaïve and RAGexp ILC2 populations raises some questions on the (different?) origin of these populations. It is known that there are different waves of ILC2 origin (most notably shown in the Schneider et al Immunity 2019 publication, PMID 31128962). I believe it would be very interesting to further discuss or possibly show if there are different origins for these two ILC populations.

Several publications describe the presence and origin of ILC2s in/from the thymus (PMIDs 33432227 24155745). Could the authors discuss whether there might be a common origin for the RAGexp ILC2 and Th2 cells from a thymic lineage? If true that the two populations would be derived from different populations, e.g. being the embryonic (possibly RAGnaïve) vs. adult bone marrow/thymus (possibly RAGexp), this would show a unique functional difference between the embryonic derived ILC2 vs. adult ILC2.

(2) On line 104 & Figures 1C/G etc. the authors describe that in the RAG knock-out ILC2 are relatively more abundant in the lineage negative fraction. On line 108 they further briefly mentioned that this observation is an indication of enhanced ILC2 expansion. Since the study includes an extensive multi-omics analysis, could the authors discuss whether they have seen a correlation of RAG expression in ILC2 with regulation of genes associated with proliferation, which could explain this phenomenon?

Reviewer #2 (Public Review):

Summary:

The study by Ver Heul et al., investigates the consequences of RAG expression for type 2 innate lymphoid cell (ILC2) function. RAG expression is essential for the generation of the receptors expressed by B and T cells and their subsequent development. Innate lymphocytes, which arise from the same initial progenitor populations, are in part defined by their ability to develop in the absence of RAG expression. However, it has been described in multiple studies that a significant proportion of innate lymphocytes show a history of Rag expression. In compelling studies several years ago, members of this research team revealed that early Rag expression during the development of Natural Killer cells (Karo et al., Cell 2014), the first described innate lymphocyte, had functional consequences.

Here, the authors revisit this topic, a worthwhile endeavour given the broad history of Rag expression within all ILCs and the common use of RAG-deficient mice to specifically assess ILC function. Focusing on ILC2s and utilising state-of-the-art approaches, the authors sought to understand whether early expression of Rag during ILC2 development had consequences for activity, fitness, or function. Having identified cell-intrinsic effects in vivo, the authors investigated the causes of this, identifying epigenetic changes associated with the accessibility genes associated with core ILC2 functions.

The manuscript is well written and does an excellent job of supporting the reader through reasonably complex transcriptional and epigenetic analyses, with considerate use of explanatory diagrams. Overall I think that the conclusions are fair, the topic is thought-provoking, and the research is likely of broad immunological interest. I think that the extent of functional data and mechanistic insight is appropriate.

Strengths:

- The logical and stepwise use of mouse models to first demonstrate the impact on ILC2 function in vivo and a cell-intrinsic role. Initial analyses show enhanced cytokine production by ILC2 from RAG-deficient mice. Then through two different chimeric mice (including BM chimeras), the authors convincingly show this is cell intrinsic and not simply as a result of lymphopenia. This is important given other studies implicating enhanced ILC function in RAG-/- mice reflect altered competition for resources (e.g. cytokines).

- Use of Rag expression fate mapping to support analyses of how cells were impacted - this enables a robust platform supporting subsequent analyses of the consequences of Rag expression for ILC2.

- Use of snRNA-seq supports gene expression and chromatin accessibility studies - these reveal clear differences in the data sets consistent with altered ILC2 function.

- Convincing evidence of epigenetic changes associated with loci strongly linked to ILC2 function. This forms a detailed analysis that potentially helps explain some of the altered ILC2 functions observed in ex vivo stimulation assays.

- Provision of a wealth of expression data and bioinformatics analyses that can serve as valuable resources to the field.

Weaknesses:

- Lack of insight into precisely how early RAG expression mediates its effects, although I think this is beyond the scale of this current manuscript. Really this is the fundamental next question from the data provided here.

- The epigenetic analyses provide evidence of differences in the state of chromatin, but there is no data on what may be interacting or binding at these sites, impeding understanding of what this means mechanistically.

- Focus on ILC2 from skin-draining lymph nodes rather than the principal site of ILC2 activity itself (the skin). This may well reflect the ease at which cells can be isolated from different tissues.

- Comparison with ILC2 from other sites would have helped to substantiate findings and compensate for the reliance on data on ILC2 from skin-draining lymph nodes, which are not usually assessed amongst ILC2 populations.

- The studies of how ILC2 are impacted are a little limited, focused exclusively on IL-13 and IL-5 cytokine expression.

Reviewer #3 (Public Review):

In this study, Ver Heul et al. investigate the role of RAG expression in ILC2 functions. While RAG genes are not required for the development of ILCs, previous studies have reported a history of expression in these cells. The authors aim to determine the potential consequences of this expression in mature cells. They demonstrate that ILC2s from RAG1 or RAG2 deficient mice exhibit increased expression of IL-5 and IL-13 and suggest that these cells are expanded in the absence of RAG expression. However, it is unclear whether this effect is due to a direct impact of RAG genes or a consequence of the lack of T and B cells in this condition. This ambiguity represents a key issue with this study: distinguishing the direct effects of RAG genes from the indirect consequences of a lymphopenic environment.

The authors focus their study on ILC2s found in the skin-draining lymph nodes, omitting analysis of tissues where ILC2s are more enriched, such as the gut, lungs, and fat tissue. This approach is surprising given the goal of evaluating the role of RAG genes in ILC2s across different tissues. The study shows that ILC2s derived from RAG-/- mice are more activated than those from WT mice, and RAG-deficient mice show increased inflammation in an atopic dermatitis (AD)-like disease model. The authors use an elegant model to distinguish ILC2s with a history of RAG expression from those that never expressed RAG genes. However, this model is currently limited to transcriptional and epigenomic analyses, which suggest that RAG genes suppress the type 2 regulome at the Th2 locus in ILC2s.

The authors report a higher frequency of ILC2s in RAG-/- mice in skin-draining lymph nodes, which is expected as these mice lack T and B cells, leading to ILC expansion. Previous studies have reported hyper-activation of ILCs in RAG-deficient mice, suggesting that this is not necessarily an intrinsic phenomenon. For example, RAG-/- mice exhibit hyperphosphorylation of STAT3 in the gut, leading to hyperactivation of ILC3s. This study does not currently provide conclusive evidence of an intrinsic role of RAG genes in the hyperactivation of ILC2s. The splenocyte chimera model is artificial and does not reflect a normal environment in tissues other than the spleen. Similarly, the mixed BM model does not demonstrate an intrinsic role of RAG genes, as RAG1-/- BM cells cannot contribute to the B and T cell pool, leading to an expected expansion of ILC2s. As the data are currently presented it is expected that a proportion of IL-5-producing cells will come from the RAG1-/- BM.

Overall, the level of analysis could be improved. Total cell numbers are not presented, the response of other immune cells to IL-5 and IL-13 (except the eosinophils in the splenocyte chimera mice) is not analyzed, and the analysis is limited to skin-draining lymph nodes.

The authors have a promising model in which they can track ILC2s that have expressed RAG or not. They need to perform a comprehensive characterization of ILC2s in these mice, which develop in a normal environment with T and B cells. Approximately 50% of the ILC2s have a history of RAG expression. It would be valuable to know whether these cells differ from ILC2s that never expressed RAG, in terms of proliferation and expression of IL-5 and IL-13. These analyses should be conducted in different tissues, as ILC2s adapt their phenotype and transcriptional landscape to their environment. Additionally, the authors should perform their AD-like disease model in these mice.

The authors provide a valuable dataset of single-nuclei RNA sequencing (snRNA-seq) and ATAC sequencing (snATAC-seq) from RAGexp (RAG fate map-positive) and RAGnaïve (RAG fate map-negative) ILC2s. This elegant approach demonstrates that ILC2s with a history of RAG expression are epigenomically suppressed. However, key genes such as IL-5 and IL-13 do not appear to be differentially regulated between RAGexp and RAGnaïve ILC2s according to Table S5. Although the authors show that the regulome activity of IL-5 and IL-13 is decreased in RAGexp ILC2s, how do the authors explain that these genes are not differentially expressed between the RAGexp and RAGnaïve ILC2? I think that it is important to validate this in vivo.

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