Spatial Transcriptomics of Meningeal Inflammation Reveals Variable Penetrance of Inflammatory Gene Signatures into Adjacent Brain Parenchyma

  1. Division of Neuroimmunology, Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
  2. Solomon Snyder, Department of Neuroscience Johns Hopkins University School of Medicine, Baltimore, MD, USA

Peer review process

Revised: This Reviewed Preprint has been revised by the authors in response to the previous round of peer review; the eLife assessment and the public reviews have been updated where necessary by the editors and peer reviewers.

Read more about eLife’s peer review process.

Editors

  • Reviewing Editor
    Irene Salinas
    University of New Mexico, Albuquerque, United States of America
  • Senior Editor
    Satyajit Rath
    Indian Institute of Science Education and Research (IISER), Pune, India

Reviewer 1 (Public Review):

Multiple sclerosis (MS) is a debilitating autoimmune disease that causes loss of myelin in neurons of the central nervous system. MS is characterized by the presence of inflammatory immune cells in several brain regions as well as the brain barriers (meninges). This study aims to understand the local immune hallmarks in regions of the brain parenchyma that are adjacent to the leptomeninges in a mouse model of MS. The leptomeninges are known to be a foci of inflammation in MS and perhaps "bleed" inflammatory cells and molecules to adjacent brain parenchyma regions. To do so, they use novel technology called spatial transcriptomics so that the spatial relationships between the two regions remain intact. The study identifies canonical inflammatory genes and gene sets such as complement and B cells enriched in the parenchyma in close proximity to the leptomeninges in the mouse model of MS but not control. The manuscript is very well written and easy to follow. The results will become a useful resource to others working in the field and can be followed by time series experiments where the same technology can be applied to the different stages of the disease.

Comments on revised version:

I agree that the authors successfully addressed most of my comments/critiques.
However, the fact that the control mice were not injected with CFA is somewhat concerning, because it will be hard to interpret the cause of the transcriptomic readouts described in this study. Some of the described effects might be due to CFA (which was used in the EAE but not the "naive" group), and not necessarily to the relapsing-remitting EAE immune features recapitulated in this mouse model. Moreover, this caveat associated with the "naive" control group is not being clearly stated throughout the manuscript and might go unnoticed to readers.
The authors should clearly state, in the methods section (in the section "Induction of SJL EAE"), that the naive control group was not injected with CFA.
Additionally, this potential confounder, of not using a control group injected with the same CFA regimen of the EAE group, should be mentioned in paragraph two of the discussion alongside the other limitations of the study already highlighted by the authors (or in another section of the discussion).

Reviewer 2 (Public Review):

Accumulating data suggests that the presence of immune cell infiltrates in the meninges of the multiple sclerosis brain contributes to the tissue damage in the underlying cortical grey matter by the release of inflammatory and cytotoxic factors that diffuse into the brain parenchyma. However, little is known about the identity and direct and indirect effects of these mediators at a molecular level. This study addresses the vital link between an adaptive immune response in the CSF space and the molecular mechanisms of tissue damage that drive clinical progression. In this short report the authors use a spatial transcriptomics approach using Visium Gene Expression technology from 10x Genomics, to identify gene expression signatures in the meninges and the underlying brain parenchyma, and their interrelationship, in the PLP-induced EAE model of MS in the SJL mouse. MRI imaging using a high field strength (11.7T) scanner was used to identify areas of meningeal infiltration for further study. They report, as might be expected, the upregulation of genes associated with the complement cascade, immune cell infiltration, antigen presentation, and astrocyte activation. Pathway analysis revealed the presence of TNF, JAK-STAT and NFkB signaling, amongst others, close to sites of meningeal inflammation in the EAE animals, although the spatial resolution is insufficient to indicate whether this is in the meninges, grey matter, or both.

UMAP clustering illuminated a major distinct cluster of upregulated genes in the meninges and smaller clusters associated with the grey matter parenchyma underlying the infiltrates. The meningeal cluster contained genes associated with immune cell functions and interactions, cytokine production, and action. The parenchymal clusters included genes and pathways related to glial activation, but also adaptive/B-cell mediated immunity and antigen presentation. This again suggests a technical inability to resolve fully between the compartments as immune cells do not penetrate the pial surface in this model or in MS. Finally, a trajectory analysis based on distance from the meningeal gene cluster successfully demonstrated descending and ascending gradients of gene expression, in particular a decline in pathway enrichment for immune processes with distance from the meninges.

Comments on revised version:

The authors have addressed all of my comments regarding the lack of spatial resolution between the grey matter and the overlying meninges and also concerning the difficulties in extrapolating from this mouse model to MS itself.
I am however very concerned about the lack of the correct control group. Immunization of rodents with complete freunds adjuvant (albeit with pertussis toxin) gives rise to widespread microglial activation, some immune cell infiltration and also structural changes to axons, particularly at nodes of Ranvier (https://doi.org/10.1097/NEN.0b013e3181f3a5b1). This will inevitably make it difficult to interpret the transcriptomics results, depending on whether these changes are reversible or not and the time frame of the reversal. In the C57Bl6 EAE models adjuvant induced microglial activation becomes chronic, whereas the axonal changes do reverse by 10 weeks. Whether this is the same in SJL EAE model using CFA alone is not clear.

Author response:

The following is the authors’ response to the original reviews.

Reviewer #1 (Recommendations For The Authors):

This study is very well framed and the writing is very clear. The manuscript is well organized and easy to follow and overall the previous state of the art of the field is taken into account. I only have a couple of minor comments

(1) There is a preprint that uses single nuclei RNA-Seq and ST on human MS subcortical white matter lesions doi: https://doi.org/10.1101/2022.11.03.514906. This work needs to be included in the discussion of the results.

(1.1) We appreciate the reviewer bringing up this important preprint, and we have referenced it in the Discussion section of our updated manuscript.

(2) The discussion should include the overall limitations of the study and how much it can be translated to human MS. Specifically, the current work uses EAE and therefore different disease stages are not captured in this study. This point is also raised by other reviewers.

(1.2) We thank the reviewer for raising this important point, and we have included additional discussion about the limitations of EAE and its disease relevance to MS.

Reviewer #2 (Recommendations For The Authors):

The authors state that this EAE model is better for studying cortical gradients because previous models "such as directly injecting inflammatory cytokines into the meninges/cortex" cause a traumatic injury. It needs to be discussed that these models have now been superseded by more refined models involving long-term overexpression of pro-inflammatory cytokines in the sub-arachnoid space, thereby avoiding traumatic injury. The current results should be discussed in light of these newer models (James et al, 2020; 2022), which are more similar to MS cortical pathology and do exhibit lymphoid-like structures.

(2.1) We thank the reviewer for pointing out these relevant studies, and we agree they describe non-traumatic and more MS-relevant models of leptomeningeal inflammation. We have included discussion of these works in the updated manuscript.

  • The study will be substantially improved if some of the ST data is validated at least partially with some RNAscope or other in situ hybridization using a subset of probes that capture the take-home message of the paper.

(2.2) We agree with the reviewer that validation of transcriptomics results is important to support our conclusions. In the updated manuscript Figure 5 and Supplemental Figure 6 we have added RNAscope results for relevant genes. In agreement with the trends noted in the manuscript, expression of genes related to antigen processing and presentation such as B2m decreases gradually with distance from LMI. We also have included a reference to a newly published manuscript from our group (Gupta et al., 2023, J. Neuroinflammation) that characterizes meningeal inflammation and sub-pial changes in the SJL EAE model. In that manuscript, IHC is used to show accumulation of B cells and T cells in the leptomeningeal space, increased microglial and astrocyte reactivity adjacent to leptomeningeal inflammation, and reduction of neuronal markers adjacent to leptomeningeal inflammation.

  • The lack of change in signaling pathways involved in B-cell/T-cell interaction and cytokine/chemokine signaling, which would be expected in areas of immune cell aggregation in the meninges, needs discussion.

(2.3) While we detected significant upregulation in antigen presentation, complement activation, and humoral immune signaling, areas of meningeal inflammation identified as cluster 11 showed upregulation of numerous other GO gene sets associated with immune cell interaction and cytokine signaling, as described in supplementary table 3. These include T-cell receptor binding, CCR chemokine receptor binding, interleukin 8 production, response to interleukin 1, positive regulation of interleukin-6 production, tumor necrosis factor production, leukocyte cell-cell adhesion. Overall, we believe that the collection of enriched gene sets is consistent with peripheral myeloid and lymphoid infiltration and cytokine production, with the most prominent cytokine / pathways being interferon ɣ/antigen processing and presentation, complement, and humoral inflammation.

  • Fig 4 subclusters includes T-cell activation, pos regulation of neuronal death, cellular response to IFNg, neg regulation of neuronal projections, Ig mediated immune response, cell killing, pos regulation of programmed cell death, pos regulation of apoptotic process, but none of these are discussed despite their obvious importance.

(2.4) We agree with the reviewer that these upregulated genesets warrant additional discussion and have added additional reference to these genesets in the results section. Also, the genesets ‘positive regulation of programmed cell death’, ‘positive regulation of apoptotic process’, and ‘positive regulation of cell death’ were erroneously included in Figure 4F in the initial manuscript, as they are actually downregulated in cluster 1_4. This has been clarified in the text.

  • Subcluster 11 appears spatially to represent the meninges, but what pathways are expressed there? 330 genes/pathways altered independent of other clusters - immune cell regulation?

(2.5) We refer the reviewer to Supplementary Table 3, which contains a complete list of GO genesets enriched within cluster 11 spots.

  • The surprising lack of immunoglobulin genes upregulated in the meninges of the mice, considering these are the genes most upregulated in the MS meninges. Should be pointed out and discussed.

(2.6) We appreciate the reviewer bringing up immunoglobulin genes, which previous publications have shown are elevated in MS meninges and cortical grey matter lesions. Consistent with this, several immunoglobulin genes are elevated in cluster 11, including genes encoding IgG2b, IgA, and IgM. While these results were available within the original submission in Supplementary Table 2, we have included the graph in the updated Supplementary Figure 3.

  • Meningeal signature may be poorly represented given the individual slices shown in suppl 3A, which suggests that only 3 of the EAE slices had significant meningeal infiltrates, indicated by cluster 11 genes.

(2.7) There was heterogeneity in the location and extent of meningeal infiltrate / cluster 11 in the EAE slices, as the reviewer points out. 2 slices had severe inflammation, 2 had moderate inflammation, and 2 had relatively mild inflammation, but all EAE slices were enriched in inflammation relative to naïve as demonstrated not only through clustering, but also through enriched marker analysis between EAE and Naive and Progeny analysis.

  • The ST is not resolving the meningeal tissue and the immediate underlying grey matter, as demonstrated by a high signal for both CXCL13 and GFAP in cluster 11.

(2.8) We agree that the spatial transcriptomics strategy applied here is inadequate to precisely delineate between meningeal inflammation and the underlying brain parenchyma, and that the elevation of markers such as GFAP in cluster 11 indicates some ‘contamination’ of parenchymal cells into cluster 11. We have clarified this in the text and discussed the limitation of the spatial transcriptomics method used.

  • More information is required concerning how many animals were used in this study, to meet the requirements for complying with the 3Rs.

(2.9) A total of 4 mice were used per group. In the naïve group one mouse contributed two slices, for a total of 5 naïve slices. In the EAE group two mice contributed two slices, for a total of 6 EAE slices. We have clarified this in the methods section of the updated manuscript.

Reviewer #3 (Recommendations For The Authors):

The authors should provide a more thorough description of the methodology, and there are a few minor concerns about experimental details, data presentation, and description that need to be addressed. In the next few lines, I will highlight a few important aspects that need to be addressed, propose some changes to the main manuscript, and suggest some additional experiments that, if successful, could confirm/support/further strengthen the conclusions that are at this point purely based on transcriptomic data.

Major comments/suggestions:

  • The main gene expression changes between the control and EAE groups obtained via spatial transcriptomics need to be validated with another technique, at least partially. I suggest performing RNAscope or immunofluorescence imaging using brain sections from a new and independent cohort of animals, where cell-specific markers can also be tested. This type of assessment would work as a validation method and could also inform about the cell-specific contribution to the observed transcriptomic changes.

(3.1) Please refer to response 2.2

  • The representative qualitative spatial expression heatmaps for each gene in Fig. 1F should be accompanied by corresponding graphs with quantitative measurements. Similar to what is done regarding the data in Fig. 2B and D.

(3.2) We agree with the reviewer that quantitative graphs were missing, and we have included them in the updated Supplementary Figure 1.

  • A supplementary table discriminating all the DEGs (132 up and 70 downregulated) between cluster 11 and the other clusters has to be provided. What is the contribution of recruited encephalitogenic adaptive immune cells to this cluster 11 gene signature?

(3.3) These unfiltered results are provided in Supplementary Table 2, and to view the up and down regulated genes the reader can sort the table based on fold change and adjusted P value. We believe providing the complete table is more useful to the reader, since the fold change and

P value thresholds used to determine “significance” are arbitrary. Since the spatial transcriptomics method used in this work does not have single cell resolution, we cannot accurately estimate the contribution of encephalitogenic adaptive immune cells in cluster 11. However, given previously published work of lymphocyte infiltration into the subarachnoid space in SJL EAE (Gupta et al., 2023, J. Neuroinflammation) and the enrichment of Cd3e in cluster 11 (Log2FC 0.31, adjusted P-val 0.005) we assume some contribution of peripheral lymphocytes.

  • The authors mention that there is grey matter pathology in this relapse model, and this has been shown in a previous publication (Bhargava et al., 2021). However, the regions analyzed in the present study are different from the ones shown in the referenced paper. Is there an overexpression of genes involved in, or gene modules indicative of, neuronal stress and/or death that spatially overlap with clusters 1 and 2? If so, it would be important to provide information about those gene modules in the main figures. It would also be quite relevant to show the levels of cell stress/death proteins and of axonal stress/damage, by APP and/or nonphosphorylated SMI-32 staining, in the deep brain regions (like the thalamus), to corroborate the link between these phenomena and the gene signatures of subclusters 1_3, 1_4, and 2_6.

(3.4) We thank the review for this insightful comment. We have recently published a manuscript that histologically analyzes leptomeningeal inflammation in the SJL EAE model, specifically assessing the areas looked at in our submitted manuscript (Gupta et al., 2023, J. Neuroinflammation). In that manuscript, IHC is used to show accumulation of B cells and T cells in the leptomeningeal space, increased microglial and astrocyte reactivity adjacent to leptomeningeal inflammation, and reduction of neuronal markers adjacent to leptomeningeal inflammation. To further describe the gene modules in the inflammatory subclusters 1_3/1_4/2_6, we have now provided heatmaps of the selected genesets and their constituent genes (Supplementary Figure 5).

  • It would be important to provide heatmaps discriminating the DEGs that make the gene modules that are significantly altered in subclusters 1_3, 1_4, and 2_6. The gene ontology terms are sometimes ambiguous. For instance, it would be very informative to the reader (and to the field) to know which altered genes compose the "lysosome", "immune response", "response to stress", or "B cell meditated immunity" pathways that are altered in the EAE subcluster 1_3 (Fig. 4E). The same applies to the gene modules altered in the other subclusters of interest. Authors should also consider generating a Venn diagram with the DEGs from subclusters 1_3, 1_4, and 2_6, to complement the GO term Venn presented in Fig. 4H. Having these pieces of information readily available, either as main or supplementary figures, would be a great addition.

(3.5) We agree with the reviewer on this point and have included these heatmaps in Supplementary Figure 5.

  • The role of IFN-gamma as well as B cells (and Igs) in myelination/remyelination is mentioned in the discussion. However, there is very little evidence that these cells or their cytokines/Igs are mediating the described transcriptomic signatures at the level of the brain parenchyma of EAE mice undergoing relapse. Do the "antigen processing and presentation, cell killing, interleukin 6 production, and interferon gamma response" go terms, which better fitted the trajectory analysis, in fact include genes expressed almost exclusively by T and/or B cells? Are there genes that are downstream of IFN type I or II signaling?

(3.6) Pathways including antigen processing / presentation, humoral inflammation, complement, among others were enriched in areas of meningeal inflammation and adjacent areas of parenchyma. These signaling pathways are mediated by effector molecules, many of which are produced by lymphocytes, but that can act on cells within the CNS parenchyma. The heatmaps in Supplementary Figure 5 demonstrate the significant role of MHC and complement genes, which could be expressed by leukocytes as well as glia, on many of the pathways.

  • Is the transcriptomic overlap between meningeal and brain parenchymal regions, or the appearance of signatures similar to the parenchymal subclusters 1_3, 1_4, and 2_6, prevented if the mice are treated with the murine versions of natalizumab or rituximab prior relapse?

(3.6) We appreciate the reviewers suggestion. Our future directions for this work includes testing the effects of disease modifying therapies on spatial and single-cell transcriptomic readouts of disease in SJL EAE.

  • Please clarify what control group was used in this study. Naïve mice are mentioned in the Results section, does this mean that control animals were not injected with CFA? Authors should also elaborate on the descriptive methodology employed for the analysis of the spatial

transcriptomics data - especially regarding the trajectory analysis. As is, overall, the methodology description might not favor reproducibility.

(3.7) We appreciate the need for clarification here. Our control group in this study was naïve, not having received any CFA or pertussis toxin. While often used as the control in EAE studies focused on mechanisms of autoimmunity, CFA and pertussis toxin independently induce systemic inflammation. Since in this study we were interested in neuroinflammation broadly, we chose to use a naïve comparison group to maximize our ability to find genes enriched in neuroinflammation. We have elaborated our methods section, including methods related to trajectory analysis.

Minor comments/suggestions:

In Fig. 1D the indication of the rostral to ventral axis needs to be inverted.

Addressed.

In Fig. 1E the authors should also include a representative H&E staining of the same region in a control animal.

Addressed.

There is inconsistency in the number of clusters obtained after UMAP unbiased clustering of the spatial transcriptomic data:

  • Fig. 3A-E - twelve clusters are shown (cluster 0 to 11).
  • In the Results section eleven clusters are mentioned - "we performed unbiased UMAP clustering on the spatial transcriptomic dataset and identified 11 distinct clusters".

The text was incorrect, there were 12 distinct clusters. This has been corrected.

Considering the mice strain used was SJL/J mice, the peptide used to induce EAE should be PLP139-151, as mentioned in the Methods section "Induction of SJL EAE". However, the legend of Fig. 1 mentions "post immunization with MOG 35-55". Please correct this.

Corrected.

In the Methods section it is mentioned "At 12 weeks post-immunization, animals were euthanized", however the Results section mentions that tissues were harvested at 11 weeks post-immunization - "Brain slices were collected from four naïve mice and four EAE mice 11 weeks postimmunization". Please correct this.

The Methods were incorrect, this has now been fixed.

Please clarify the number of animals used for spatial transcriptomic analysis:

  • Legend of Fig. 1 mentions "Red arrows indicate MRI time points, black arrow indicates time of tissue harvesting (N = 6)." Whilst in the Results section it states "Brain slices were collected from four naïve mice and four EAE mice".

The figure one legend has now been corrected (N = 4). Additionally, we have added clarification about the number of animals / slices used in the Methods section (see response 2.9).

Please be consistent in the way of representing DEGs in the MA plots:

  • Fig. 3F shows the upregulated genes (in red) on the right and the downregulated genes (in blue) on the left.

  • Supplemental Fig. 2K shows the upregulated genes (in red) on the left and the downregulated genes (in blue) on the right.

  • Supplemental Fig. 4 shows the upregulated genes on the right in blue, while the downregulated genes are in red.

This has been fixed.

The letters attributed to each subcluster in panels E-G of Fig. 4 are different from the respective figure legend.

This has been fixed.

Correct the legend of supplemental figure 2: o "(G-H) Representative spatial feature plots of read count (F) and UMI (G) demonstrate expected anatomic variability in transcript amount and diversity.".

This has been fixed.

In Supplemental Fig. 4G there is probably an error with the XX axis, since the significantly up and down-regulated genes are not visible.

This has been fixed.

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