Sex-dependent gastrointestinal colonization resistance to MRSA is microbiome and Th17 dependent

  1. Department of Microbiology, New York University School of Medicine, New York, NY, 10016, USA
  2. Department of Medicine, Division of Infectious Diseases, New York University School of Medicine, New York, NY 10016, USA
  3. Department of Medicine, Division of Gastroenterology and Hepatology, New York University Langone Health, New York, NY, 10016, USA
  4. Antimicrobial-Resistant Pathogens Program, New York University School of Medicine, United States
  5. NYU-Regeneron Veterinary Postdoctoral Training Program in Laboratory Animal Medicine, Division of Comparative Medicine, New York University School of Medicine, New York, NY 10016, USA
  6. Department of Host-Microbe Interactions, St. Jude Children’s Research Hospital, Memphis, TN, 38105, USA
  7. Department of Medicine, Division of Gastroenterology and Hepatology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
  8. Department of Pathobiology, University of Pennsylvania Perelman School of Veterinary Medicine, Philadelphia, PA, 19104, USA

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
    Kiyoshi Takeda
    Osaka University, Osaka, Japan
  • Senior Editor
    Wendy Garrett
    Harvard T.H. Chan School of Public Health, Boston, United States of America

Reviewer #1 (Public review):

Summary:

Lejeune et al. demonstrated sex-dependent differences in the susceptibility to MRSA infection. The authors demonstrated the role of the microbiota and sex hormones as potential determinants of susceptibility. Moreover, the authors showed that Th17 cells and neutrophils contribute to sex hormone-dependent protection in female mice.

Strengths:

The role of microbiota was examined in various models (gnotobiotic, co-housing, microbiota transplantation). The identification of responsible immune cells was achieved using several genetic knockouts and cell-specific depletion models. The involvement of sex hormones was clarified using ovariectomy and the FCG model.

Weaknesses:

The mechanisms by which specific microbiota confer female-specific protection remain unclear.

Reviewer #2 (Public review):

The current study by Lejeune et al. investigates factors that allow for persistent MRSA infection in the GI tract. They developed an intriguing model of intestinal MRSA infection that does not use the traditional antibiotic approach, thereby allowing for a more natural infection that includes the normal intestinal microbiota. This model is more akin to what might be expected to be observed in a healthy human host. They find that biological sex plays a clear role in bacterial persistence during infection but only in mice bred at an NYU Facility and not those acquired from Jackson Labs. This clearly indicates a role for the intestinal microbiome in affecting female bacterial persistence but not male persistence which was unaffected by the origin of the mice and thus the microbiome. Through a series of clever microbiome-specific transfer experiments, they determine that the NYU-specific microbiome plays a role in this sexual dimorphism but is not solely responsible. Additional experiments indicate that Th17 cells, estrogen, and neutrophils also participate in the resistance to persistent infection. Notably, they assess the role of sex chromosomes (X/Y) using the established four core genotype model and find that these chromosomes appear to play little role in bacterial persistence.

Overall, the paper nicely adds to the growing body of literature investigating how biological sex impacts the immune system and the burden of infectious disease. The conclusions are mostly supported by the data although there are some aspects of the data that could be better addressed and clarified.

(1) There is something of a disconnect between the initial microbiome data and the later data that analyzes sex hormones and chromosomes. While there are clearly differences in microbial species across the two sites (NYU and JAX) how these bacterial species might directly interact with immune cells to induce female-specific responses is left unexplored. At the very least it would help to try and link these two distinct pieces of data to try and inform the reader how the microbiome is regulating the sex-specific response. Indeed, the reader is left with no clear exploration of the microbiota's role in the persistence of the infection and thus is left wanting.

(2) While the authors make a reasonable case that Th17 T cells are important for controlling infection (using RORgt knockout mice that cannot produce Th17 cells), it is not clear how these cells even arise during infection since the authors make most of the observations 2 days post-infection which is longer before a normal adaptive immune response would be expected to arise. The authors acknowledge this, but their explanation is incomplete. The increase in Th17 cells they observe is predicated on mitogenic stimulation, so they are not specific (at least in this study) for MRSA. It would be helpful to see a specific restimulation of these cells with MRSA antigens to determine if there are pre-existing, cross-reactive Th17 cells specific for MRSA and microbiota species which could then link these two as mentioned above.

(3) The ovariectomy experiment demonstrates a role for ovarian hormones; however, it lacks a control of adding back ovarian hormones (or at least estrogen) so it is not entirely obvious what is causing the persistence in this experiment. This is especially important considering the experiments demonstrating no role for sex chromosomes thus demonstrating that hormonal effects are highly important. Here it leaves the reader without a conclusive outcome as to the exact hormonal mechanism.

(4) The discussion is underdeveloped and is mostly a rehash of the results. It would greatly enhance the manuscript if the authors would more carefully place the results in the context of the current state of the field including a more enhanced discussion of the role of estrogen, microbiome, and T cells and how the field might predict these all interact and how they might be interacting in the current study as well.

Reviewer #3 (Public review):

Summary:

Using a mouse model of Staphylococcus aureus gut colonization, Lejeune et al. demonstrate that the microbiome, immune system, and sex are important contributing factors for whether this important human pathogen persists in the gut. The work begins by describing differential gut clearance of S. aureus in female B6 mice bred at NYU compared to those from Jackson Laboratories (JAX). NYU female mice cleared S. aureus from the gut but NYU male mice and mice of both sexes from JAX exhibited persistent gut colonization. Further experimentation demonstrated that differences between staphylococcal gut clearance in NYU and JAX female mice were attributed to the microbiome. However, NYU male and female mice harbor similar microbiomes, supporting the conclusion that the microbiome cannot account for the observed sex-dependent clearance of S. aureus gut colonization. To identify factors responsible for female clearance of S. aureus, the authors performed RNAseq on intestinal epithelial cells and cells enriched within the lamina propria. This analysis revealed sex-dependent transcriptional responses in both tissues. Genes associated with immune cell function and migration were distinctly expressed between the sexes. To determine which immune cell types contribute to S. aureus clearance Lejeune et al employed genetic and antibody-mediated immune cell depletion. This experiment demonstrated that CD4+ IL17+ cells and neutrophils promote the elimination of S. aureus from the gut. Subsequent experiments, including the use of the 'four core genotype model' were conducted to discern between the roles of sex chromosomes and sex hormones. This work demonstrated that sex-chromosome-linked genes are not responsible for clearance, increasing the likelihood that hormones play a dominant role in controlling S. aureus gut colonization.

Strengths:

A strength of the work is the rigorous experimental design. Appropriate controls were executed and, in most cases, multiple approaches were conducted to strengthen the authors' conclusions. The conclusions are supported by the data.

The following suggestions are offered to improve an already strong piece of scholarship.

Weaknesses:

The correlation between female sex hormones and the elimination of S. aureus from the gut could be further validated by quantifying sex hormones produced in the four core genotype mice in response to colonization. Additionally, and this may not be feasible, but according to the proposed model administering female sex hormones to male mice should decrease colonization. Finally, knowing whether the quantity of IL-17a CD4+ cells change in the OVX mice has the potential to discern whether abundance/migration of the cells or their activation is promoted by female sex hormones.

In the Discussion, the authors highlight previous work establishing a link between immune cells and sex hormone receptors, but whether the estrogen (and progesterone) receptor is differentially expressed in response to S. aureus colonization could be assessed in the RNAseq dataset. Differential expression of known X and Y chromosome-linked genes were discussed but specific sex hormones or sex hormone receptors, like the estrogen receptor, were not. This potential result could be highlighted.

Author response:

Reviewer #1 (Public review):

Summary:

Lejeune et al. demonstrated sex-dependent differences in the susceptibility to MRSA infection. The authors demonstrated the role of the microbiota and sex hormones as potential determinants of susceptibility. Moreover, the authors showed that Th17 cells and neutrophils contribute to sex hormone-dependent protection in female mice.

Strengths:

The role of microbiota was examined in various models (gnotobiotic, co-housing, microbiota transplantation). The identification of responsible immune cells was achieved using several genetic knockouts and cell-specific depletion models. The involvement of sex hormones was clarified using ovariectomy and the FCG model.

Weaknesses:

The mechanisms by which specific microbiota confer female-specific protection remain unclear.

We thank the reviewer for highlighting the strength of the manuscript including the models and techniques we employ. We agree that the relationship between the microbiota and sex-dependent protection is less developed compared with other aspects of the study. In preparation of a revised manuscript, we intend on performing a more thorough comparison of male vs. female microbiota, along with quantification of sex hormones and downstream Th17 function (neutrophil recruitment and activation).

Reviewer #2 (Public review):

Overall, the paper nicely adds to the growing body of literature investigating how biological sex impacts the immune system and the burden of infectious disease. The conclusions are mostly supported by the data although there are some aspects of the data that could be better addressed and clarified.

We thank the reviewer for appreciating our contribution. We intend on performing experiments to fill-in gaps and text revisions to increase clarity and acknowledge limitations.

(1) There is something of a disconnect between the initial microbiome data and the later data that analyzes sex hormones and chromosomes. While there are clearly differences in microbial species across the two sites (NYU and JAX) how these bacterial species might directly interact with immune cells to induce female-specific responses is left unexplored. At the very least it would help to try and link these two distinct pieces of data to try and inform the reader how the microbiome is regulating the sex-specific response. Indeed, the reader is left with no clear exploration of the microbiota's role in the persistence of the infection and thus is left wanting.

We agree. This comment is similar to Reviewer #1’s feedback. As mentioned above, we anticipate clarifying the association between sex differences and the microbiota. We will attempt to investigate specific bacteria, although some aspects of microbiota characterization may be outside the timeframe of the revision.

(2) While the authors make a reasonable case that Th17 T cells are important for controlling infection (using RORgt knockout mice that cannot produce Th17 cells), it is not clear how these cells even arise during infection since the authors make most of the observations 2 days post-infection which is longer before a normal adaptive immune response would be expected to arise. The authors acknowledge this, but their explanation is incomplete. The increase in Th17 cells they observe is predicated on mitogenic stimulation, so they are not specific (at least in this study) for MRSA. It would be helpful to see a specific restimulation of these cells with MRSA antigens to determine if there are pre-existing, cross-reactive Th17 cells specific for MRSA and microbiota species which could then link these two as mentioned above.

We acknowledge that this is a major limitation of our study. Although an experiment demonstrating pre-existing, cross-reactive T cells would help support our conclusion, aspects of MRSA biology may make the results of this experiment difficult to interpret. We have consulted with an expert on MRSA virulence factors, co-lead author Dr. Victor Torres, about the feasibility of this experiment. MRSA possess superantigens, such as Staphylococcal enterotoxin B, which bind directly to specific Vβ regions of T-cell receptors (TCR) and major histocompatibility complex (MHC) class II on antigen-presenting cells, resulting in hyperactivation of T lymphocytes and monocytes/macrophages. Additionally, other MRSA virulence factors, such as α-hemolysin and LukED, can induce cell death of lymphocytes. MRSA’s enterotoxins are heat stable, so heat-inactivation of the bacterium may not help in this matter. For these reasons, restimulation of lymphocytes with MRSA antigens may be difficult to interpret. We humbly suggest that addressing this aspect of the mechanism is outside the scope of this manuscript.

A study by Shao et al. provides an example of a host commensal species inducing Th17 cells with cross-reactivity against MRSA. Upon intestinal colonization, the intestinal fungus Candida albicans influences T cell polarization towards a Th17 phenotype in the spleen and peripheral lymph nodes which provided protection to the host against systemic candidemia. Interestingly, this induction of protective Th17 cells, increased IL-17 and responsiveness in circulating Ly6G+ neutrophils also protected mice from intravenous infection with MRSA, indicating that T cell activation and polarization by intestinal C. albicans leads to non-specific protective responses against extracellular pathogens.

Shao TY, Ang WXG, Jiang TT, Huang FS, Andersen H, Kinder JM, Pham G, Burg AR, Ruff B, Gonzalez T, Khurana Hershey GK, Haslam DB, Way SS. Commensal Candida albicans Positively Calibrates Systemic Th17 Immunological Responses. Cell Host & Microbe. 2019 Mar 13;25(3):404-417.e6. doi: 10.1016/j.chom.2019.02.004. PMID: 30870622; PMCID: PMC6419754.

Reviewer #3 (Public review):

Strengths:

A strength of the work is the rigorous experimental design. Appropriate controls were executed and, in most cases, multiple approaches were conducted to strengthen the authors' conclusions. The conclusions are supported by the data.

The following suggestions are offered to improve an already strong piece of scholarship.

Weaknesses:

The correlation between female sex hormones and the elimination of S. aureus from the gut could be further validated by quantifying sex hormones produced in the four core genotype mice in response to colonization. Additionally, and this may not be feasible, but according to the proposed model administering female sex hormones to male mice should decrease colonization. Finally, knowing whether the quantity of IL-17a CD4+ cells change in the OVX mice has the potential to discern whether abundance/migration of the cells or their activation is promoted by female sex hormones.

In the Discussion, the authors highlight previous work establishing a link between immune cells and sex hormone receptors, but whether the estrogen (and progesterone) receptor is differentially expressed in response to S. aureus colonization could be assessed in the RNAseq dataset. Differential expression of known X and Y chromosome-linked genes were discussed but specific sex hormones or sex hormone receptors, like the estrogen receptor, were not. This potential result could be highlighted.

We appreciate the comment on the scholarship and thank the Reviewer for the insightful suggestions to improve this manuscript. We intend on measuring hormone levels and performing the recommended (or similar) experiments based on availability of reagents and mice during the revision period. We also apologize for not including references that address some of the Reviewer’s questions. Other research groups have compared the levels of hormones between XX and XY males and females in the four core genotypes model and have found similar levels of circulating testosterone in adult XX and XY males. No difference was found in circulating estradiol levels in XX vs XY- females when tested at 4-6 or 7-9 months of age.

Karen M. Palaszynski, Deborah L. Smith, Shana Kamrava, Paul S. Burgoyne, Arthur P. Arnold, Rhonda R. Voskuhl, A Yin-Yang Effect between Sex Chromosome Complement and Sex Hormones on the Immune Response. Endocrinology, Volume 146, Issue 8, 1 August 2005, Pages 3280–3285, https://doi.org/10.1210/en.2005-0284

Sasidhar MV, Itoh N, Gold SM, Lawson GW, Voskuhl RR. The XX sex chromosome complement in mice is associated with increased spontaneous lupus compared with XY. Ann Rheum Dis. 2012 Aug;71(8):1418-22. doi: 10.1136/annrheumdis-2011-201246. Epub 2012 May 12. PMID: 22580585; PMCID: PMC4452281.

Examination of the levels of estrogen, progesterone, and androgen receptors in our cecal-colonic lamina propria RNA-seq dataset is an excellent idea. We will add these analyses to the revised manuscript. We are planning additional experiments to better understand the contributions of hormones or their receptors and anticipate including such data in either a response letter or revised manuscript.

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