Ly6G+Granulocytes-derived IL-17 limits protective host responses and promotes tuberculosis pathogenesis

  1. Cellular Immunology Group, International Centre for Genetic Engineering and Biotechnology, Aruna Asaf Ali Marg, New Delhi, India-110067
  2. National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, India-110067
  3. Department of Pathology, All India Institute of Medical Sciences, New Delhi, India-110012
  4. Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore
  5. Infectious Diseases Labs (ID labs), Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore
  6. Department of Immunology, ICMR-National Institute for Research in Tuberculosis, Chennai, India – 600031
  7. Prof. M. Viswanathan Diabetes Research Center, Chennai, India
  8. Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
  9. NIH-International Center of Excellence in Research, Chennai, India
  10. Laboratory of Parasitic Diseases, NIAID-NIH, Bethesda, Maryland, USA
  11. Centre for Cellular and Molecular Biology, Hyderabad, India
  12. Lee Kong Chian School of Medicine, Nanyang Technological University (NTU), Singapore, Republic of Singapore

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
    Bavesh Kana
    University of the Witwatersrand, Johannesburg, South Africa
  • Senior Editor
    Bavesh Kana
    University of the Witwatersrand, Johannesburg, South Africa

Reviewer #1 (Public review):

Summary:

Recruitment of neutrophils to the lungs is known to drive susceptibility to infection with M. tuberculosis. In this study, the authors present data in support of the hypothesis that neutrophil production of the cytokine IL-17 underlies the detrimental effect of neutrophils on disease. They claim that neutrophils harbor a large fraction of Mtb during infection, and are a major source of IL-17. To explore the effects of blocking IL-17 signaling during primary infection, they use IL-17 blocking antibodies, SR221 (an inverse agonist of TH17 differentiation), and celecoxib, which they claim blocks Th17 differentiation, and observe modest improvements in bacterial burdens in both WT and IFN-γ deficient mice using the combination of IL-17 blockade with celecoxib during primary infection. Celecoxib enhances control of infection after BCG vaccination.

Strengths:

The most novel finding in the paper is that treatment with celecoxib significantly enhances control of infection in BCG-vaccinated mice that have been challenged with Mtb. It was already known that NSAID treatments can improve primary infection with Mtb.

Weaknesses:

The major claim of the manuscript - that neutrophils produce IL-17 that is detrimental to the host - is not strongly supported by the data. Data demonstrating neutrophil production of IL-17 lacks rigor. The experiments examining the effects of inhibitors of IL-17 on the outcome of infection are very difficult to interpret. First, treatment with IL-17 inhibitors alone has no impact on bacterial burdens in the lung, either in WT or IFN-γ KO mice. This suggests that IL-17 does not play a detrimental role during infection. Modest effects are observed using the combination of IL-17 blocking drugs and celecoxib, however, the interpretation of these results mechanistically is complicated. Celecoxib is not a specific inhibitor of Th17. Indeed, it affects levels of PGE2, which is known to have numerous impacts on Mtb infection separate from any effect on IL-17 production, as well as other eicosanoids. Finally, the human data simply demonstrates that neutrophils and IL-17 both are higher in patients who experience relapse after treatment for TB, which is expected and does not support their specific hypothesis. The use of genetic ablation of IL-17 production specifically in neutrophils and/or IL-17R in mice would greatly enhance the rigor of this study. The authors do not address the fact that numerous studies have shown that IL-17 has a protective effect in the mouse model of TB in the context of vaccination. Finally, whether and how many times each animal experiment was repeated is unclear.

Reviewer #2 (Public review):

Summary:

In this study, Sharma et al. demonstrated that Ly6G+ granulocytes (Gra cells) serve as the primary reservoirs for intracellular Mtb in infected wild-type mice and that excessive infiltration of these cells is associated with severe bacteremia in genetically susceptible IFNγ-/- mice. Notably, neutralizing IL-17 or inhibiting COX2 reversed the excessive infiltration of Ly6G+Gra cells, mitigated the associated pathology, and improved survival in these susceptible mice. Additionally, Ly6G+Gra cells were identified as a major source of IL-17 in both wild-type and IFNγ-/- mice. Inhibition of RORγt or COX2 further reduced the intracellular bacterial burden in Ly6G+Gra cells and improved lung pathology.

Of particular interest, COX2 inhibition in wild-type mice also enhanced the efficacy of the BCG vaccine by targeting the Ly6G+Gra-resident Mtb population.

Strengths:

The experimental results showing improved BCG-mediated protective immunity through targeting IL-17-producing Ly6G+ cells and COX2 are compelling and will likely generate significant interest in the field. Overall, this study presents important findings, suggesting that the IL-17-COX2 axis could be a critical target for designing innovative vaccination strategies for TB.

Weaknesses:

However, I have the following concerns regarding some of the conclusions drawn from the experiments, which require additional experimental evidence to support and strengthen the overall study.

Major Concerns:

(1) Ly6G+ Granulocytes as a Source of IL-17: The authors assert that Ly6G+ granulocytes are the major source of IL-17 in wild-type and IFN-γ KO mice based on colocalization studies of Ly6G and IL-17. In Figure 3D, they report approximately 500 Ly6G+ cells expressing IL-17 in the Mtb-infected WT lung. Are these low numbers sufficient to drive inflammatory pathology? Additionally, have the authors evaluated these numbers in IFN-γ KO mice?

(2) Role of IL-17-Producing Ly6G Granulocytes in Pathology: The authors suggest that IL-17-producing Ly6G granulocytes drive pathology in WT and IFN-γ KO mice. However, the data presented only demonstrate an association between IL-17+ Ly6G cells and disease pathology. To strengthen their conclusion, the authors should deplete neutrophils in these mice to show that IL-17 expression, and consequently the pathology, is reduced.

(3) IL-17 Secretion by Mtb-Infected Neutrophils: Do Mtb-infected neutrophils secrete IL-17 into the supernatants? This would serve as confirmation of neutrophil-derived IL-17. Additionally, are Ly6G+ cells producing IL-17 and serving as pathogenic agents exclusively in vivo? The authors should provide comments on this.

(4) Characterization of IL-17-Producing Ly6G+ Granulocytes: Are the IL-17-producing Ly6G+ granulocytes a mixed population of neutrophils and eosinophils, or are they exclusively neutrophils? Sorting these cells followed by Giemsa or eosin staining could clarify this.

Reviewer #3 (Public review):

Summary:

The authors examine how distinct cellular environments differentially control Mtb following BCG vaccination. The key findings are that IL17-producing PMNs harbor a significant Mtb load in both wild-type and IFNg-/- mice. Targeting IL17 and Cox2 improved disease and enhanced BCG efficacy over 12 weeks and neutrophils/IL17 are associated with treatment failure in humans. The authors suggest that targeting these pathways, especially in MSMD patients may improve disease outcomes.

Strengths:

The experimental approach is generally sound and consists of low-dose aerosol infections with distinct readouts including cell sorting followed by CFU, histopathology, and RNA sequencing analysis. By combining genetic approaches and chemical/antibody treatments, the authors can probe these pathways effectively.

Understanding how distinct inflammatory pathways contribute to control or worsen Mtb disease is important and thus, the results will be of great interest to the Mtb field.

Weaknesses:

A major limitation of the current study is overlooking the role of non-hematopoietic cells in the IFNg/IL17/neutrophil response. Chimera studies from Ernst and colleagues (PMCID: PMC2807991) previously described this IDO-dependent pathway following the loss of IFNg through an increased IL17 response. This study is not cited nor discussed even though it may alter the interpretation of several experiments.

Several of the key findings in mice have previously been shown (albeit with less sophisticated experimentation) and human disease and neutrophils are well described - thus the real new finding is how intracellular Mtb in neutrophils are more refractory to BCG-mediated control. However, given there are already high levels of Mtb in PMNs compared to other cell types, and there is a decrease in intracellular Mtb in PMNs following BCG immunization the strength of this finding is a bit limited.

Author response:

eLife assessment

This potentially valuable study examines the role of IL17-producing Ly6G PMNs as a reservoir for Mycobacterium tuberculosis to evade host killing activated by BCG immunisation. The authors report that IL17-producing polymorphonuclear neutrophils harbour a significant bacterial load in both wild-type and IFNg-/- mice and that targeting IL17 and Cox2 improved disease outcomes whilst enhancing BCG efficacy. Although the authors suggest that targeting these pathways may improve disease outcomes in humans, the evidence as it stands is incomplete and requires additional experimentation for the study to realise its full impact.

Thank you for evaluating our manuscript. We understand the concern related to the direct role of Ly6G+Gra-derived IL17 in TB pathogenesis. For the revised manuscript, we will provide additional experimental evidence through direct regulation of IL-17 production in Mtb-infected mice and its impact on improving BCG efficacy.

Public Reviews:

Reviewer #1 (Public review):

Summary:

Recruitment of neutrophils to the lungs is known to drive susceptibility to infection with M. tuberculosis. In this study, the authors present data in support of the hypothesis that neutrophil production of the cytokine IL-17 underlies the detrimental effect of neutrophils on disease. They claim that neutrophils harbor a large fraction of Mtb during infection, and are a major source of IL-17. To explore the effects of blocking IL-17 signaling during primary infection, they use IL-17 blocking antibodies, SR221 (an inverse agonist of TH17 differentiation), and celecoxib, which they claim blocks Th17 differentiation, and observe modest improvements in bacterial burdens in both WT and IFN-γ deficient mice using the combination of IL-17 blockade with celecoxib during primary infection. Celecoxib enhances control of infection after BCG vaccination.

Thank you for the summary.

Strengths:

The most novel finding in the paper is that treatment with celecoxib significantly enhances control of infection in BCG-vaccinated mice that have been challenged with Mtb. It was already known that NSAID treatments can improve primary infection with Mtb.

Thank you.

Weaknesses:

The major claim of the manuscript - that neutrophils produce IL-17 that is detrimental to the host - is not strongly supported by the data. Data demonstrating neutrophil production of IL17 lacks rigor.

Our response: Neutrophil production of IL-17 is supported by two independent methods/ techniques in the current version:

(1) Through Flow cytometry- a large fraction of Ly6G+CD11b+ cells from the lungs of Mtb-infected mice were also positive for IL-17 (Fig. 3C).

(2) IFA co-staining of Ly6G + cells with IL-17 in the lung sections from Mtb-infected mice (Fig. 3 E_G and Fig. 4H, Fig. 5I).

However, to further strengthen this observation, we plan to analyse sorted Ly6G+Gra from the lungs of infected mice using IL-17 ELISPOT assay. This will unequivocally prove the Ly6+Gra production of IL-17. Several publications support the production of IL-17 by neutrophils (Li et al. 2010; Katayama et al. 2013; Lin et al. 2011). For example, neutrophils have been identified as a source of IL-17 in human psoriatic lesions (Lin et al. 2011), in neuroinflammation induced by traumatic brain injury (Xu et al. 2023) and in several mouse models of infectious and autoimmune inflammation (Ferretti et al. 2003; Hoshino et al. 2008) (Li et al. 2010). However, ours is the first study reporting neutrophil IL-17 production during Mtb pathology.

The experiments examining the effects of inhibitors of IL-17 on the outcome of infection are very difficult to interpret. First, treatment with IL-17 inhibitors alone has no impact on bacterial burdens in the lung, either in WT or IFN-γ KO mice. This suggests that IL-17 does not play a detrimental role during infection. Modest effects are observed using the combination of IL-17 blocking drugs and celecoxib, however, the interpretation of these results mechanistically is complicated. Celecoxib is not a specific inhibitor of Th17. Indeed, it affects levels of PGE2, which is known to have numerous impacts on Mtb infection separate from any effect on IL-17 production, as well as other eicosanoids.

The reviewer correctly says that Celecoxib is not a specific inhibitor of Th17. However, COX-2 inhibition does have an effect on IL-17 levels, and numerous reports support this observation (Paulissen et al. 2013; Napolitani et al. 2009; Lemos et al. 2009). We elaborate on the results below for better clarity.

Firstly, in the WT mice, Celecoxib treatment led to a complete loss of IL-17 production in the lungs of Mtb-infected mice (Fig. 5D). Interestingly, IL-17 production independent of IL-23 is known to require PGE2 (Paulissen et al. 2013; Polese et al. 2021). In the WT or IFNγ KO mice, we rather noted a decline in IL-23 levels post-infection, suggesting a possible role of PGE2 in IL-17 production. However, in the lung homogenates of Mtb-infected IFNγ KO mice, Celecoxib had no effect on IL-17 levels in the lung homogenates. Thus, celecoxib controls IL-17 levels only in the Mtb-infected WT mice. Including celecoxib with anti-IL17 in the IFNγ KO mice controls pathology and extends its survival.

Second, the reviewer’s observation is only partially correct that IL-17 inhibition has a modest effect on the outcome of infection. While IL-17 neutralization and inhibition alone in the IFNγ KO mice and WT mice, respectively, did not bring down the lung CFU burden significantly, in both these cases, there was an improvement in the lung pathology. The reduced pathology coincided with reduced neutrophil recruitment and a reduced Ly6G+Graresident Mtb population in the WT mice. IL-17 neutralization alone improved IFNγ KO mice survival by ~10 days (Fig. 4F-G).

Third, regarding the SR2211 and Celecoxib combination study, we agree with the reviewer that Celecoxib has roles independent of IL-17 regulation. However, in the results presented in this study, there are three key aspects- 1) neutrophil-derived IL-17-dependent neutrophil recruitment, 2) the presence of a large proportion of intracellular Mtb in the neutrophils and 3) dissemination of Mtb to the spleen. Celecoxib treatment alone helps reduce lung Mtb burden in the WT mice. However, SR2211 fails to do so. It is evident that celecoxib is doing more than just inhibiting IL-17 production. The result shows that celecoxib blocks neutrophil recruitment (which could be an IL-17-dependent mechanism) and also controls the intraneutrophil bacterial population. Finally, either SR2211 or celecoxib could block dissemination to the spleen. The role of neutrophils in TB dissemination is only beginning to emerge (Hult et al. 2021). We will revise the description in the results and discussion section for this data to make it easier to understand.

Finally, we have also done experiments with SR2211 in BCG-vaccinated animals, which shows the direct impact of IL-17 inhibition on the BCG vaccine efficacy. We will add this result in the revised version.

Finally, the human data simply demonstrates that neutrophils and IL-17 both are higher in patients who experience relapse after treatment for TB, which is expected and does not support their specific hypothesis.

We disagree with the above statement. Why a higher IL-17 is expected in patients who show relapse, death or failed treatment outcomes? Classically, IL-17 is believed to be protective against TB, and the reviewer also points to that in the comments below. A very limited set of studies support the non-protective/pathological role of IL-17 in tuberculosis (Cruz et al. 2010). High IL-17 and neutrophilia at the baseline in the human subjects (i.e. at the time of recruitment in the study) highlight severe pathology in those subjects, which could have contributed to the failed treatment outcome. This observation in the human cohort strongly supports the overall theme and central observation in this study.

The use of genetic ablation of IL-17 production specifically in neutrophils and/or IL-17R in mice would greatly enhance the rigor of this study.

The reviewer’s point is well-taken. Having a genetic ablation of IL-17 production, specifically in the neutrophils, would be excellent. At present, however, we lack this resource, and therefore, it is not feasible to do this experiment within a defined timeline. Instead, for the revised manuscript, we will present the data with SR2211, a direct inhibitor of RORgt and, therefore, IL-17, in BCG-vaccinated mice.

The authors do not address the fact that numerous studies have shown that IL-17 has a protective effect in the mouse model of TB in the context of vaccination.

Yes, there are a few articles that talk about the protective effect of IL-17 in the mouse model of TB in the context of vaccination (Khader et al. 2007; Desel et al. 2011; Choi et al. 2020). This part was discussed in the original manuscript (in the Introduction section). For the revised manuscript, we will also provide results from the experiment where we blocked IL-17 production by inhibiting RORgt using SR2211 in BCG-vaccinated mice. The results clearly show IL-17 as a negative regulator of BCG-mediated protective immunity. We believe some of the reasons for the observed differences could be 1) in our study, we analysed IL-17 levels in the lung homogenates at late phases of infection, and 2) most published studies rely on ex vivo stimulation of immune cells to measure cytokine production, whereas we actually measured the cytokine levels in the lung homogenates. We will elaborate on these points in the revised version.

Finally, whether and how many times each animal experiment was repeated is unclear.

We will provide the details of the number of experiments in the revised version. Briefly, the BCG vaccination experiment (Figure 1) and BCG vaccination with Celecoxib treatment experiment (Figure 6) were performed twice and thrice, respectively. The IL-17 neutralization experiment (Figure 4) and the SR2211 treatment experiment (Figure 5) were done once. We will add another SR2211 experiment data in the revised version.

Reviewer #2 (Public review):

Summary:

In this study, Sharma et al. demonstrated that Ly6G+ granulocytes (Gra cells) serve as the primary reservoirs for intracellular Mtb in infected wild-type mice and that excessive infiltration of these cells is associated with severe bacteremia in genetically susceptible IFNγ/- mice. Notably, neutralizing IL-17 or inhibiting COX2 reversed the excessive infiltration of Ly6G+Gra cells, mitigated the associated pathology, and improved survival in these susceptible mice. Additionally, Ly6G+Gra cells were identified as a major source of IL-17 in both wild-type and IFNγ-/- mice. Inhibition of RORγt or COX2 further reduced the intracellular bacterial burden in Ly6G+Gra cells and improved lung pathology.

Of particular interest, COX2 inhibition in wild-type mice also enhanced the efficacy of the BCG vaccine by targeting the Ly6G+Gra-resident Mtb population.

Thank you for the summary.

Strengths:

The experimental results showing improved BCG-mediated protective immunity through targeting IL-17-producing Ly6G+ cells and COX2 are compelling and will likely generate significant interest in the field. Overall, this study presents important findings, suggesting that the IL-17-COX2 axis could be a critical target for designing innovative vaccination strategies for TB.

Thank you for highlighting the overall strengths of the study. Weaknesses:

However, I have the following concerns regarding some of the conclusions drawn from the experiments, which require additional experimental evidence to support and strengthen the overall study.

Major Concerns:

(1) Ly6G+ Granulocytes as a Source of IL-17: The authors assert that Ly6G+ granulocytes are the major source of IL-17 in wild-type and IFN-γ KO mice based on colocalization studies of Ly6G and IL-17. In Figure 3D, they report approximately 500 Ly6G+ cells expressing IL-17 in the Mtb-infected WT lung. Are these low numbers sufficient to drive inflammatory pathology? Additionally, have the authors evaluated these numbers in IFN-γ KO mice?

Thank you for pointing out about the numbers in Fig. 3D. It was our oversight to label the axis as No. of IL17+Ly6G+Gra/lung. For this data, only a part of the lung was used. For the revised manuscript, we will provide the number of these cells at the whole lung level from Mtb-infected WT mice. Unfortunately, we did not evaluate these numbers in IFN-γ KO mice through FACS.

For the assertion that Ly6G+Gra are the major source of IL-17 in TB, we have used two separate strategies- a) IFA and b) FACS.

However, as described above in response to the first reviewer, for the revision, we propose to perform an IL-17 ELISpot assay on the sorted Ly6G+Gra from the lungs of Mtb-infected WT mice.

(2) Role of IL-17-Producing Ly6G Granulocytes in Pathology: The authors suggest that IL17-producing Ly6G granulocytes drive pathology in WT and IFN-γ KO mice. However, the data presented only demonstrate an association between IL-17+ Ly6G cells and disease pathology. To strengthen their conclusion, the authors should deplete neutrophils in these mice to show that IL-17 expression, and consequently the pathology, is reduced.

Thank you for this suggestion. Others have done neutrophil depletion studies in TB, and so far, the outcomes remain inconclusive. In some studies, neutrophil depletion helps the pathogen (Rankin et al. 2022; Pedrosa et al. 2000; Appelberg et al. 1995), and in others, it helps the host (Lovewell et al. 2021; Mishra et al. 2017) ). One reason for this variability is the stage of infection when neutrophil depletion was done. However, another crucial factor is the heterogeneity in the neutrophil population. There are reports that suggest neutrophil subtypes with protective versus pathological trajectories (Nwongbouwoh Muefong et al. 2022; Lyadova 2017; Hellebrekers, Vrisekoop, and Koenderman 2018; Leliefeld et al. 2018). Depleting the entire population using anti-Ly6G could impact this heterogeneity and may impact the inferences drawn. A better approach would be to characterise this heterogeneous population, efforts towards which could be part of a separate study.

For the revised manuscript, we will provide results from the SR2211 experiment in BCG-vaccinated mice and other results to show the role of IL-17-producing Ly6G+Gra in TB pathology.

(3) IL-17 Secretion by Mtb-Infected Neutrophils: Do Mtb-infected neutrophils secrete IL-17 into the supernatants? This would serve as confirmation of neutrophil-derived IL-17. Additionally, are Ly6G+ cells producing IL-17 and serving as pathogenic agents exclusively in vivo? The authors should provide comments on this.

We have not directly measured IL-17 secretion by neutrophils in our experiments. However, Hu et al have reported IL-17 secretion by Mtb-infected neutrophils in vitro (Hu et al. 2017). Whether there are a few neutrophil roles exclusively seen under in vivo condition is an interesting proposition. We do have some observations that suggest in vitro phenotype of Mtb-infected neutrophils is different from in vivo.

(4) Characterization of IL-17-Producing Ly6G+ Granulocytes: Are the IL-17-producing Ly6G+ granulocytes a mixed population of neutrophils and eosinophils, or are they exclusively neutrophils? Sorting these cells followed by Giemsa or eosin staining could clarify this.

This is a very important point. While usually eosinophils do not express Ly6G markers in laboratory mice, under specific contexts, including infections, eosinophils can express Ly6G. Since we have not characterized these potential Ly6G+ sub-populations, that is one of the reasons we refer to the cell types as Ly6G+ granulocytes, which do not exclude Ly6G+ eosinophils. A detailed characterization of these subsets could be taken up as a separate study.

Reviewer #3 (Public review):

Summary:

The authors examine how distinct cellular environments differentially control Mtb following BCG vaccination. The key findings are that IL17-producing PMNs harbor a significant Mtb load in both wild-type and IFNg-/- mice. Targeting IL17 and Cox2 improved disease and enhanced BCG efficacy over 12 weeks and neutrophils/IL17 are associated with treatment failure in humans. The authors suggest that targeting these pathways, especially in MSMD patients may improve disease outcomes.

Thank you.

Strengths:

The experimental approach is generally sound and consists of low-dose aerosol infections with distinct readouts including cell sorting followed by CFU, histopathology, and RNA sequencing analysis. By combining genetic approaches and chemical/antibody treatments, the authors can probe these pathways effectively.

Understanding how distinct inflammatory pathways contribute to control or worsen Mtb disease is important and thus, the results will be of great interest to the Mtb field.

Thank you.

Weaknesses:

A major limitation of the current study is overlooking the role of non-hematopoietic cells in the IFNg/IL17/neutrophil response. Chimera studies from Ernst and colleagues (PMCID: PMC2807991) previously described this IDO-dependent pathway following the loss of IFNg through an increased IL17 response. This study is not cited nor discussed even though it may alter the interpretation of several experiments.

Thank you for pointing out this earlier study, which we concede we missed discussing. We disagree on the point that results from that study may alter the interpretation of several experiments in our study. On the contrary, the main observation that loss of IFNγ causes severe IL-17 levels is aligned in both studies.

IDO1 is known to alter Th cell differentiation towards Tregs and away from Th17 (Baban et al. 2009). It is absolutely feasible for the non-hematopoietic cells to regulate these events. However, that does not rule out the neutrophil production of IL-17 and the downstream pathological effect shown in this study. We will discuss and cite this study in the revised manuscript.

Several of the key findings in mice have previously been shown (albeit with less sophisticated experimentation) and human disease and neutrophils are well described - thus the real new finding is how intracellular Mtb in neutrophils are more refractory to BCGmediated control. However, given there are already high levels of Mtb in PMNs compared to other cell types, and there is a decrease in intracellular Mtb in PMNs following BCG immunization the strength of this finding is a bit limited.

The reviewer’s interpretation of the BCG-refractory Mtb population in the neutrophil is interesting. The reviewer is right that neutrophils had a higher intracellular Mtb burden, which decreased in the BCG-vaccinated animals. Thus, on that account, the reviewer rightly mentions that BCG is able to control Mtb even in neutrophils. However, BCG almost clears intracellular burden from other cell types analysed, and therefore, the remnant pool of intracellular Mtb in the lungs of BCG-vaccinated animals could be mostly those present in the neutrophils. This is a substantial novel development in the field and attracts focus towards innate immune cells for vaccine efficacy.

References:

Appelberg, R., A. G. Castro, S. Gomes, J. Pedrosa, and M. T. Silva. 1995. 'SuscepBbility of beige mice to Mycobacterium avium: role of neutrophils', Infect Immun, 63: 3381-7.

Baban, B., P. R. Chandler, M. D. Sharma, J. Pihkala, P. A. Koni, D. H. Munn, and A. L. Mellor. 2009. 'IDO activates regulatory T cells and blocks their conversion into Th17-like T cells', J Immunol, 183: 2475-83.

Choi, H. G., K. W. Kwon, S. Choi, Y. W. Back, H. S. Park, S. M. Kang, E. Choi, S. J. Shin, and H. J. Kim. 2020. 'AnBgen-Specific IFN-gamma/IL-17-Co-Producing CD4(+) T-Cells Are the Determinants for ProtecBve Efficacy of Tuberculosis Subunit Vaccine', Vaccines (Basel), 8.

Cruz, A., A. G. Fraga, J. J. Fountain, J. Rangel-Moreno, E. Torrado, M. Saraiva, D. R. Pereira, T. D. Randall, J. Pedrosa, A. M. Cooper, and A. G. Castro. 2010. 'Pathological role of interleukin 17 in mice subjected to repeated BCG vaccination after infection with Mycobacterium tuberculosis', J Exp Med, 207: 1609-16.

Desel, C., A. Dorhoi, S. Bandermann, L. Grode, B. Eisele, and S. H. Kaufmann. 2011. 'Recombinant BCG DeltaureC hly+ induces superior protection over parental BCG by simulating a balanced combination of type 1 and type 17 cytokine responses', J Infect Dis, 204: 1573-84.

Ferreg, S., O. Bonneau, G. R. Dubois, C. E. Jones, and A. Trifilieff. 2003. 'IL-17, produced by lymphocytes and neutrophils, is necessary for lipopolysaccharide-induced airway neutrophilia: IL-15 as a possible trigger', J Immunol, 170: 2106-12.

Hellebrekers, P., N. Vrisekoop, and L. Koenderman. 2018. 'Neutrophil phenotypes in health and disease', Eur J Clin Invest, 48 Suppl 2: e12943.

Hoshino, A., T. Nagao, N. Nagi-Miura, N. Ohno, M. Yasuhara, K. Yamamoto, T. Nakayama, and K. Suzuki. 2008. 'MPO-ANCA induces IL-17 production by activated neutrophils in vitro via classical complement pathway-dependent manner', J Autoimmun, 31: 79-89.

Hu, S., W. He, X. Du, J. Yang, Q. Wen, X. P. Zhong, and L. Ma. 2017. 'IL-17 ProducBon of Neutrophils Enhances AnBbacteria Ability but Promotes ArthriBs Development During Mycobacterium tuberculosis InfecBon', EBioMedicine, 23: 88-99.

Hult, C., J. T. Magla, H. P. Gideon, J. J. Linderman, and D. E. Kirschner. 2021. 'Neutrophil Dynamics Affect Mycobacterium tuberculosis Granuloma Outcomes and DisseminaBon', Front Immunol, 12: 712457.

Katayama, M., K. Ohmura, N. Yukawa, C. Terao, M. Hashimoto, H. Yoshifuji, D. Kawabata, T. Fujii, Y. Iwakura, and T. Mimori. 2013. 'Neutrophils are essential as a source of IL-17 in the effector phase of arthritis', PLoS One, 8: e62231.

Khader, S. A., G. K. Bell, J. E. Pearl, J. J. Fountain, J. Rangel-Moreno, G. E. Cilley, F. Shen, S. M. Eaton, S. L. Gaffen, S. L. Swain, R. M. Locksley, L. Haynes, T. D. Randall, and A. M. Cooper. 2007. 'IL-23 and IL-17 in the establishment of protective pulmonary CD4+ T cell responses after vaccination and during Mycobacterium tuberculosis challenge', Nat Immunol, 8: 369-77.

Leliefeld, P. H. C., J. Pillay, N. Vrisekoop, M. Heeres, T. Tak, M. Kox, S. H. M. Rooijakkers, T. W. Kuijpers, P. Pickkers, L. P. H. Leenen, and L. Koenderman. 2018. 'DifferenBal antibacterial control by neutrophil subsets', Blood Adv, 2: 1344-55.

Lemos, H. P., R. Grespan, S. M. Vieira, T. M. Cunha, W. A. Verri, Jr., K. S. Fernandes, F. O. Souto, I. B. McInnes, S. H. Ferreira, F. Y. Liew, and F. Q. Cunha. 2009. 'Prostaglandin mediates IL-23/IL-17induced neutrophil migraBon in inflammation by inhibiting IL-12 and IFNgamma production', Proc Natl Acad Sci U S A, 106: 5954-9.

Li, L., L. Huang, A. L. Vergis, H. Ye, A. Bajwa, V. Narayan, R. M. Strieter, D. L. Rosin, and M. D. Okusa. 2010. 'IL-17 produced by neutrophils regulates IFN-gamma-mediated neutrophil migration in mouse kidney ischemia-reperfusion injury', J Clin Invest, 120: 331-42.

Lin, A. M., C. J. Rubin, R. Khandpur, J. Y. Wang, M. Riblen, S. Yalavarthi, E. C. Villanueva, P. Shah, M. J. Kaplan, and A. T. Bruce. 2011. 'Mast cells and neutrophils release IL-17 through extracellular trap formation in psoriasis', J Immunol, 187: 490-500.

Lovewell, R. R., C. E. Baer, B. B. Mishra, C. M. Smith, and C. M. Sasseg. 2021. 'Granulocytes act as a niche for Mycobacterium tuberculosis growth', Mucosal Immunol, 14: 229-41.

Lyadova, I. V. 2017. 'Neutrophils in Tuberculosis: Heterogeneity Shapes the Way?', Mediators Inflamm, 2017: 8619307.

Mishra, B. B., R. R. Lovewell, A. J. Olive, G. Zhang, W. Wang, E. Eugenin, C. M. Smith, J. Y. Phuah, J. E. Long, M. L. Dubuke, S. G. Palace, J. D. Goguen, R. E. Baker, S. Nambi, R. Mishra, M. G. Booty, C. E. Baer, S. A. Shaffer, V. Dartois, B. A. McCormick, X. Chen, and C. M. Sasseg. 2017. 'Nitric oxide prevents a pathogen-permissive granulocytic inflammation during tuberculosis', Nat Microbiol, 2: 17072.

Napolitani, G., E. V. Acosta-Rodriguez, A. Lanzavecchia, and F. Sallusto. 2009. 'Prostaglandin E2 enhances Th17 responses via modulation of IL-17 and IFN-gamma production by memory CD4+ T cells', Eur J Immunol, 39: 1301-12.

Nwongbouwoh Muefong, C., O. Owolabi, S. Donkor, S. Charalambous, A. Bakuli, A. Rachow, C. Geldmacher, and J. S. Sutherland. 2022. 'Neutrophils Contribute to Severity of Tuberculosis Pathology and Recovery From Lung Damage Pre- and Posnreatment', Clin Infect Dis, 74: 1757-66.

Paulissen, S. M., J. P. van Hamburg, N. Davelaar, P. S. Asmawidjaja, J. M. Hazes, and E. Lubberts. 2013. 'Synovial fibroblasts directly induce Th17 pathogenicity via the cyclooxygenase/prostaglandin E2 pathway, independent of IL-23', J Immunol, 191: 1364-72.

Pedrosa, J., B. M. Saunders, R. Appelberg, I. M. Orme, M. T. Silva, and A. M. Cooper. 2000. 'Neutrophils play a protective nonphagocytic role in systemic Mycobacterium tuberculosis infection of mice', Infect Immun, 68: 577-83.

Polese, B., B. Thurairajah, H. Zhang, C. L. Soo, C. A. McMahon, G. Fontes, S. N. A. Hussain, V. Abadie, and I. L. King. 2021. 'Prostaglandin E(2) amplifies IL-17 production by gamma-delta T cells during barrier inflammation', Cell Rep, 36: 109456.

Rankin, A. N., S. V. Hendrix, S. K. Naik, and C. L. Stallings. 2022. 'Exploring the Role of Low-Density Neutrophils During Mycobacterium tuberculosis InfecBon', Front Cell Infect Microbiol, 12: 901590.

Xu, X. J., Q. Q. Ge, M. S. Yang, Y. Zhuang, B. Zhang, J. Q. Dong, F. Niu, H. Li, and B. Y. Liu. 2023. 'Neutrophil-derived interleukin-17A participates in neuroinflammation induced by traumatic brain injury', Neural Regen Res, 18: 1046-51.

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