Aurora kinase A promotes trained immunity via regulation of endogenous S-adenosylmethionine metabolism

  1. MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
  2. State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
  3. Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
  4. Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China
  5. Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, >Guangzhou, China
  6. Metabolic Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
  7. Medical Research Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
  8. Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, 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
    Boris Novakovic
    Murdoch Childrens Research Institute, Melbourne, Australia
  • Senior Editor
    Carla Rothlin
    Yale University, New Haven, United States of America

Reviewer #1 (Public review):

This work regards the role of Aurora Kinase A (AurA) in trained immunity. The authors claim that AurA is essential to the induction of trained immunity. The paper starts with a series of experiments showing the effects of suppressing AurA on beta-glucan-trained immunity. This is followed by an account of how AurA inhibition changes the epigenetic and metabolic reprogramming that are characteristic of trained immunity. The authors then zoom in on specific metabolic and epigenetic processes (regulation of S-adenocylmethionine metabolism & histone methylation). Finally, an inhibitor of AurA is used to reduce beta-glucan's anti-tumour effects in a subcutaneous MC-38 model.

Strengths:

With the exception of my confusion around the methods used for relative gene expression measurements, the experimental methods are generally well-described. I appreciate the authors' broad approach to studying different key aspects of trained immunity (from comprehensive transcriptome/chromatin accessibility measurements to detailed mechanistic experiments). Approaching the hypothesis from many different angles inspires confidence in the results (although not completely - see weaknesses section). Furthermore, the large drug-screening panel is a valuable tool as these drugs are readily available for translational drug-repurposing research.

Weaknesses

(1) The manuscript contains factual inaccuracies such as:
(a) Intro: the claim that trained cells display a shift from OXPHOS to glycolysis based on the paper by Cheng et al. in 2014; this was later shown to be dependent on the dose of stimulation and actually both glycolysis and OXPHOS are generally upregulated in trained cells (pmid 32320649)
(b) Discussion: Trained immunity was first described as such in 2011, not decades ago.

(2) The authors approach their hypothesis from different angles, which inspires a degree of confidence in the results. However, the statistical methods and reporting are underwhelming.
(a) Graphs depict mean +/- SEM, whereas mean +/- SD is almost always more informative.
(b) The use of 1-tailed tests is dubious in this scenario. Furthermore, in many experiments/figures the case could be made that the comparisons should be considered paired (the responses of cells from the same animal are inherently not independent due to their shared genetic background and, up until cell isolation, the same host factors like serum composition/microbiome/systemic inflammation etc).
(c) It could be explained a little more clearly how multiple testing correction was done and why specific tests were chosen in each instance.
(d) Most experiments are done with n = 3, some experiments are done with n = 5. This is not a lot. While I don't think power analyses should be required for simple in vitro experiments, I would be wary of drawing conclusions based on n = 3. It is also not indicated if the data points were acquired in independent experiments. ATAC-seq/RNA-seq was, judging by the figures, done on only 2 mice per group. No power calculations were done for the in vivo tumor model.
(e) Furthermore, the data spread in many experiments (particularly BMDM experiments) is extremely small. I wonder if these are true biological replicates, meaning each point represents BMDMs from a different animal? (disclaimer: I work with human materials where the spread is of course always much larger than in animal experiments, so I might be misjudging this.).

(3) Maybe the authors are reserving this for a separate paper, but it would be fantastic if the authors would report the outcomes of the entire drug screening instead of only a selected few. The field would benefit from this as it would save needless repeat experiments. The list of drugs contains several known inhibitors of training (e.g. mTOR inhibitors) so there must have been more 'hits' than the reported 8 Aurora inhibitors.

(4) Relating to the drug screen and subsequent experiments: it is unclear to me in supplementary figure 1B which concentrations belong to secondary screens #1/#2 - the methods mention 5 µM for the primary screen and "0.2 and 1 µM" for secondary screens, is it in this order or in order of descending concentration?
(a) It is unclear if the drug screen was performed with technical replicates or not - the supplementary figure 1B suggests no replicates and quite a large spread (in some cases lower concentration works better?)

(5) The methods for (presumably) qPCR for measuring gene expression in Figure 1C are missing. Which reference gene was used and is this a suitably stable gene?

(6) From the complete unedited blot image of Figure 1D it appears that the p-Aurora and total Aurora are not from the same gel (discordant number of lanes and positioning). This could be alright if there are no/only slight technical errors, but I find it misleading as it is presented as if the actin (loading control to account for aforementioned technical errors!) counts for the entire figure.

(7) Figure 2: This figure highlights results that are by far not the strongest ones - I think the 'top hits' deserve some more glory. A small explanation on why the highlighted results were selected would have been fitting.

(8) Figure 3 incl supplement: the carbon tracing experiments show more glucose-carbon going into TCA cycle (suggesting upregulated oxidative metabolism), but no mito stress test was performed on the seahorse.

(9) Inconsistent use of an 'alisertib-alone' control in addition to 'medium', 'b-glucan', 'b-glucan + alisertib'. This control would be of great added value in many cases, in my opinion.

(10) Figure 4A: looking at the unedited blot images, the blot for H3K36me3 appears in its original orientation, whereas other images appear horizontally mirrored. Please note, I don't think there is any malicious intent but this is quite sloppy and the authors should explain why/how this happened (are they different gels and the loading sequence was reversed?)

(11) For many figures, for example prominently figure 5, the text describes 'beta-glucan training' whereas the figures actually depict acute stimulation with beta-glucan. While this is partially a semantic issue (technically, the stimulation is 'the training-phase' of the experiment), this could confuse the reader.

(12) Figure 6: Cytokines, especially IL-6 and IL-1β, can be excreted by tumour cells and have pro-tumoral functions. This is not likely in the context of the other results in this case, but since there is flow cytometry data from the tumour material it would have been nice to see also intracellular cytokine staining to pinpoint the source of these cytokines.

Reviewer #2 (Public review):

Summary:

This manuscript investigates the inhibition of Aurora A and its impact on β-glucan-induced trained immunity via the FOXO3/GNMT pathway. The study demonstrates that inhibition of Aurora A leads to overconsumption of SAM, which subsequently impairs the epigenetic reprogramming of H3K4me3 and H3K36me3, effectively abolishing the training effect.

Strengths:

The authors identify the role of Aurora A through small molecule screening and validation using a variety of molecular and biochemical approaches. Overall, the findings are interesting and shed light on the previously underexplored role of Aurora A in the induction of β-glucan-driven epigenetic change.

Weaknesses:

Given the established role of histone methylations, such as H3K4me3, in trained immunity, it is not surprising that depletion of the methyl donor SAM impairs the training response. Nonetheless, this study provides solid evidence supporting the role of Aurora A in β-glucan-induced trained immunity in murine macrophages. The part of in vivo trained immunity antitumor effect is insufficient to support the final claim as using Alisertib could inhibits Aurora A other cell types other than myeloid cells.

Author response:

Reviewer #1 (Public review):

This work regards the role of Aurora Kinase A (AurA) in trained immunity. The authors claim that AurA is essential to the induction of trained immunity. The paper starts with a series of experiments showing the effects of suppressing AurA on beta-glucan-trained immunity. This is followed by an account of how AurA inhibition changes the epigenetic and metabolic reprogramming that are characteristic of trained immunity. The authors then zoom in on specific metabolic and epigenetic processes (regulation of S-adenosylmethionine metabolism & histone methylation). Finally, an inhibitor of AurA is used to reduce beta-glucan's anti-tumour effects in a subcutaneous MC-38 model.

Strengths:

With the exception of my confusion around the methods used for relative gene expression measurements, the experimental methods are generally well-described. I appreciate the authors' broad approach to studying different key aspects of trained immunity (from comprehensive transcriptome/chromatin accessibility measurements to detailed mechanistic experiments). Approaching the hypothesis from many different angles inspires confidence in the results (although not completely - see weaknesses section). Furthermore, the large drug-screening panel is a valuable tool as these drugs are readily available for translational drug-repurposing research.

We thank the reviewer for the positive and encouraging comments.

Weaknesses:

(1) The manuscript contains factual inaccuracies such as: (a) Intro: the claim that trained cells display a shift from OXPHOS to glycolysis based on the paper by Cheng et al. in 2014; this was later shown to be dependent on the dose of stimulation and actually both glycolysis and OXPHOS are generally upregulated in trained cells (pmid 32320649).

We appreciate the reviewer for pointing out this inaccuracy, and we will revise our statement to ensure accurate and updated description. We are aware that trained immunity involves different metabolic pathways, including both glycolysis and oxidative phosphorylation[1, 2]. We also detected Oxygen Consumption Rate (OCR, as detailed in comment#8) but observed no increase of oxygen consumption in trained BMDMs while previous study reported decreased oxidative phosphorylation[3]. We will discuss the potential reasons underlying such different results.

(b) Discussion: Trained immunity was first described as such in 2011, not decades ago.

We are sorry for the inaccurate description, and we will correct the statement in our revised manuscript as “Despite the fact that the concept of “trained immunity” has been proposed since 2011, the mechanisms that regulate trained immunity are still not completely understood.”

(2) The authors approach their hypothesis from different angles, which inspires a degree of confidence in the results. However, the statistical methods and reporting are underwhelming.

(a) Graphs depict mean +/- SEM, whereas mean +/- SD is almost always more informative. (b) The use of 1-tailed tests is dubious in this scenario. Furthermore, in many experiments/figures the case could be made that the comparisons should be considered paired (the responses of cells from the same animal are inherently not independent due to their shared genetic background and, up until cell isolation, the same host factors like serum composition/microbiome/systemic inflammation etc). (c) It could be explained a little more clearly how multiple testing correction was done and why specific tests were chosen in each instance.

Thank you for the suggestions and we will revise all data presented as mean ± SEM in the manuscript to mean ± SD, and provide a detailed description of how multiple comparisons were performed and explain the rationale behind the different comparison methods used. Previous studies have shown that knockdown of GNMT increases intracellular SAM level and knockdown of GNMT is commonly used as a method to upregulate SAM[4-6]. Thus we used 1-tailed test in Figure 3J.

(d) Most experiments are done with n = 3, some experiments are done with n = 5. This is not a lot. While I don't think power analyses should be required for simple in vitro experiments, I would be wary of drawing conclusions based on n = 3. It is also not indicated if the data points were acquired in independent experiments. ATAC-seq/RNA-seq was, judging by the figures, done on only 2 mice per group. No power calculations were done for the in vivo tumor model.

We are sorry for the confusion in our description in figure legends. As for in vitro studies, we performed at least three independent experiments (BMs isolated from different mice) but we only display technical replicates data from one experiment in our manuscript. As for seq data, we acknowledge the reviewer's concern regarding the small sample size (n=2) in our RNA-seq/ATAC-seq experiment. We consider the sequencing experiment mainly as an exploratory approach, and performed rigorous quality control and normalization of the sequencing data to ensure the reliability of our findings. While we understand that a larger sample size would be ideal for drawing more definitive conclusions, we believe that the current data offer valuable preliminary insights that can inform future studies with larger cohorts. As a complementary method, we conducted ChIP PCR for detecting active histone modification enrichment in Il6 and Tnf region to further verify the increased accessibility of trained immunity induced inflammatory genes and reliability of our conclusions despite the small sample size. We hope this clarifies our approach, and we would be happy to further acknowledge and discuss the limitations of the current study.

For the in vivo experiment, we determined the sample size by referring to the animal numbers used for similar experiments in literatures. And according to a reported resource equation approach for calculating sample size in animal studies[7], n=5-7 is suitable for most of our mouse experiments. We will describe the details in the revised methods part.

(e) Furthermore, the data spread in many experiments (particularly BMDM experiments) is extremely small. I wonder if these are true biological replicates, meaning each point represents BMDMs from a different animal? (disclaimer: I work with human materials where the spread is of course always much larger than in animal experiments, so I might be misjudging this.).

We are sorry for the confusion in our description in figure legends. In vivo experiments represent individual mice as biological replicates, the exact values of n are reported in figure legends and each point represents data from a different animal (Figure 1I, and Figure 6). The in vitro cell assay was performed in triplicates, each experiment was independently replicated at least three times and points represents technical replicates.

(3) Maybe the authors are reserving this for a separate paper, but it would be fantastic if the authors would report the outcomes of the entire drug screening instead of only a selected few. The field would benefit from this as it would save needless repeat experiments. The list of drugs contains several known inhibitors of training (e.g. mTOR inhibitors) so there must have been more 'hits' than the reported 8 Aurora inhibitors.

Thank you for your suggestion and we will report the outcomes of the entire drug screening in the revised manuscript.

(4) Relating to the drug screen and subsequent experiments: it is unclear to me in supplementary figure 1B which concentrations belong to secondary screens #1/#2 - the methods mention 5 µM for the primary screen and "0.2 and 1 µM" for secondary screens, is it in this order or in order of descending concentration?

Thank you for your comments and we are sorry for unclear labelled results in supplementary 1B. We performed secondary drug screen at two concentrations, and drug concentrations corresponding to secondary screen#1 and #2 are 0.2, 1 μM respectively. That is to say, it is just in this order, not in an order of descending concentration.

(a) It is unclear if the drug screen was performed with technical replicates or not - the supplementary figure 1B suggests no replicates and quite a large spread (in some cases lower concentration works better?)

Thank you for your question. The drug screen was performed without technical replicates. Actually, we observed s a lower concentration works better in some cases. This might be due to the fact that the drug's effect correlates positively with its concentration only within a specific range (as seen in comment#4).

(5) The methods for (presumably) qPCR for measuring gene expression in Figure 1C are missing. Which reference gene was used and is this a suitably stable gene?

We are sorry for the omission for the qPCR method. The mRNA expression of Il6 and Tnf in trained BMDMs was normalized to untrained BMDMs and β-actin served as a reference gene. And we will describe in detail in our revised manuscript.

(6) From the complete unedited blot image of Figure 1D it appears that the p-Aurora and total Aurora are not from the same gel (discordant number of lanes and positioning). This could be alright if there are no/only slight technical errors, but I find it misleading as it is presented as if the actin (loading control to account for aforementioned technical errors!) counts for the entire figure.

Thanks for this comment. In the original data, p-Aurora and total Aurora were from different gels. In this experiment the membrane stripping/reprobing after p-Aurora antibody did now work well, so we couldn’t get all results from one gel, and we had to run another gel using the same samples to blot with anti-aurora antibody. Yes we should have provided separated actin blots as loading controls for this experiment. We will repeat the experiment and provide original data of three biological replicates to confirm the experiment result.

Figure 2: This figure highlights results that are by far not the strongest ones - I think the 'top hits' deserve some more glory. A small explanation on why the highlighted results were selected would have been fitting.

We appreciate the valuable suggestion. We will make a discussion in our revised manuscript.

(7) Figure 3 incl supplement: the carbon tracing experiments show more glucose-carbon going into TCA cycle (suggesting upregulated oxidative metabolism), but no mito stress test was performed on the seahorse.

We appreciate this question raised by the reviewer. We previously performed seahorse XF analyze to measure mito stress in β-glucan trained BMDMs in combination with alisertib (data not shown in our submitted manuscript). The results showed no increase in oxidative phosphorylation under β-glucan stimulation.

Author response image 1.

(8) Inconsistent use of an 'alisertib-alone' control in addition to 'medium', 'b-glucan', 'b-glucan + alisertib'. This control would be of great added value in many cases, in my opinion.

Thank you for your comment. We appreciate that including “alisertib-alone” group throughout all the experiments may add more value to the findings. We set the aim of the current study to investigate the role of Aurora kinase A in trained immunity. Therefore, in most settings, we did not focus on the role of aurora kinase A without β-glucan stimulation. Initially, we showed in Figure 1B and 1C that alisertib alone in a concentration lower than 1μM (included) does not affect the response to secondary stimulus. In a previous report, the authors showed that Aurora A inhibitor alone did not affect trained immunity[8]. Thus, we did not include this control group in all of the experiments.

(9) Figure 4A: looking at the unedited blot images, the blot for H3K36me3 appears in its original orientation, whereas other images appear horizontally mirrored. Please note, I don't think there is any malicious intent but this is quite sloppy and the authors should explain why/how this happened (are they different gels and the loading sequence was reversed?)

Thank you for pointing out this error. After checking the original data, we found that we indeed misassembled the orientation of several blots. We went through the assembling process and figured out that some orientations were assembled according to the loading sequences but not saved, so that the orientations in Figure 4A were not consistent with the unedited blot image. We are sorry for the careless mistake, and we will double check to make sure all the blots are correctly assembled in the revised manuscript.

(10) For many figures, for example prominently figure 5, the text describes 'beta-glucan training' whereas the figures actually depict acute stimulation with beta-glucan. While this is partially a semantic issue (technically, the stimulation is 'the training-phase' of the experiment), this could confuse the reader.

Thanks for the reviewer’s suggestion and we will reorganize our language to ensure clarity and avoid any inconsistencies that might lead to misunderstanding.

(11) Figure 6: Cytokines, especially IL-6 and IL-1β, can be excreted by tumour cells and have pro-tumoral functions. This is not likely in the context of the other results in this case, but since there is flow cytometry data from the tumour material it would have been nice to see also intracellular cytokine staining to pinpoint the source of these cytokines.

Thanks for the reviewer’s suggestion. To address potential concerns raised by the reviewers, we will perform intracellular cytokines staining in tumor experiments with mice trained with β-glucan or in combination with alisertib followed MC38 inoculation.

Reviewer #2 (Public review):

Summary:

This manuscript investigates the inhibition of Aurora A and its impact on β-glucan-induced trained immunity via the FOXO3/GNMT pathway. The study demonstrates that inhibition of Aurora A leads to overconsumption of SAM, which subsequently impairs the epigenetic reprogramming of H3K4me3 and H3K36me3, effectively abolishing the training effect.

Strengths:

The authors identify the role of Aurora A through small molecule screening and validation using a variety of molecular and biochemical approaches. Overall, the findings are interesting and shed light on the previously underexplored role of Aurora A in the induction of β-glucan-driven epigenetic change.

We thank the reviewer for the positive and encouraging comments.

Weaknesses:

Given the established role of histone methylations, such as H3K4me3, in trained immunity, it is not surprising that depletion of the methyl donor SAM impairs the training response. Nonetheless, this study provides solid evidence supporting the role of Aurora A in β-glucan-induced trained immunity in murine macrophages. The part of in vivo trained immunity antitumor effect is insufficient to support the final claim as using Alisertib could inhibits Aurora A other cell types other than myeloid cells.

We appreciate the question raised by the reviewer. Though SAM generally acts as methyl donor, whether the epigenetic reprogram in trained immunity is directly linked to SAM metabolism is not known. In our study, we provided evidence suggesting the necessity of SAM maintenance in supporting trained immunity. As for in vivo tumor model, tumor cells were subcutaneously inoculated 24 h after oral administration of alisertib. Previous studies showed alisertib administered orally had a half-life of 10 h and 90% concentration reduction in serum after 24 h [9, 10]. Therefore, we suppose that tumor cells are more susceptible to long-term effects of drugs on the immune system rather than directly affected by alisertib. To further address the reviewer’s concern, we will perform bone marrow transplantation (trained mice as donor and naïve mice as recipient) to clarify the mechanistic contribution of trained immunity versus off-target effects.

Cited references

(1) Ferreira, A.V., et al., Metabolic Regulation in the Induction of Trained Immunity. Semin Immunopathol, 2024. 46(3-4): p. 7.

(2) Keating, S.T., et al., Rewiring of glucose metabolism defines trained immunity induced by oxidized low-density lipoprotein. J Mol Med (Berl), 2020. 98(6): p. 819-831.

(3) Li, X., et al., Maladaptive innate immune training of myelopoiesis links inflammatory comorbidities. Cell, 2022. 185(10): p. 1709-1727.e18.

(4) Luka, Z., S.H. Mudd, and C. Wagner, Glycine N-methyltransferase and regulation of S-adenosylmethionine levels. J Biol Chem, 2009. 284(34): p. 22507-11.

(5) Hughey, C.C., et al., Glycine N-methyltransferase deletion in mice diverts carbon flux from gluconeogenesis to pathways that utilize excess methionine cycle intermediates. J Biol Chem, 2018. 293(30): p. 11944-11954.

(6) Simile, M.M., et al., Nuclear localization dictates hepatocarcinogenesis suppression by glycine N-methyltransferase. Transl Oncol, 2022. 15(1): p. 101239.

(7) Arifin, W.N. and W.M. Zahiruddin, Sample Size Calculation in Animal Studies Using Resource Equation Approach. Malays J Med Sci, 2017. 24(5): p. 101-105.

(8) Benjaskulluecha, S., et al., Screening of compounds to identify novel epigenetic regulatory factors that affect innate immune memory in macrophages. Sci Rep, 2022. 12(1): p. 1912.

(9) Yang, J.J., et al., Preclinical drug metabolism and pharmacokinetics, and prediction of human pharmacokinetics and efficacious dose of the investigational Aurora A kinase inhibitor alisertib (MLN8237). Drug Metab Lett, 2014. 7(2): p. 96-104.

(10) Palani, S., et al., Preclinical pharmacokinetic/pharmacodynamic/efficacy relationships for alisertib, an investigational small-molecule inhibitor of Aurora A kinase. Cancer Chemother Pharmacol, 2013. 72(6): p. 1255-64.

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