Volume: 1 | Issue: 2 | View as PDF | Cite
  • OPEN ACCESS

A Microsoft-Excel Template for Identifying Mouse Myeloid Cell Types in the Central Nervous System Based on Single-cell RNA Sequencing Data

  • Xin-Yi Lyu1,
  • Jing-Lu Li2,3,
  • Shu-Qin Ding2,3,
  • Jian-Guo Hu2,3 and
  • He-Zuo Lü2,3,* 
 Author information
Nature Cell and Science   2023;1(2):53-65

doi: 10.61474/ncs.2023.00004

Abstract

Keywords

Excel template, Mouse, Myeloid cell types, Central nervous system, Single-cell RNA sequencing, Clusters

Introduction

Myeloid cells play vital roles in the health and disease of the central nervous system (CNS).1,2 The cell composition of myeloid cells in CNS mainly includes microglia, monocytes, macrophages, dendritic cells, and granulocytes.3 In the healthy CNS parenchyma, monocytes, and granulocytes are absent. They are rather localized in the leptomeninges.3 However, in CNS pathologies, various myeloid cells, such as microglia, monocytes, macrophages, dendritic cells, and granulocytes, can appear and be active in the pathological CNS parenchyma.3 Although there have been many studies on these cells, how to clearly distinguish them is still a difficult problem.

Morphology, immunohistochemistry, and flow cytometry are frequently used to identify these cells.4–6 Morphology mainly relies on conventional staining to identify cells through characteristic morphology under the microscope, which is very subjective. Moreover, their morphologies are very similar under pathological conditions, therefore conventional morphology has been unable to distinguish them.7 Immunohistochemistry and flow cytometry can identify myeloid cells by labeling their markers with a panel of antibodies. Combining these two methods, we can both quantify and locate, which seems to be a perfect scheme. However, in practical application, there are often the same or cross markers among myeloid cells, which seriously affects the accuracy of analysis.8 Therefore, it is necessary to select an effective method to distinguish the myeloid cells in the CNS.

Single-cell RNA sequencing (scRNA-Seq) can sequence thousands of cells at the single-cell level, and then divide the cells into different clusters according to the similarity of gene expression.9 However, it is still difficult to further define these cell clusters because collecting the cell markers is a knotty problem for researchers.10 At present, there are three main methods for cell type identification based on single-cell transcriptome data. First, comparing the upregulated genes with the marker genes in the database, such as CellMarker (http://xteam.xbio.top/CellMarker/ ),10 PanglaoDB (https://panglaodb.se/ ),11 and the Mouse Cell Atlas (http://bis.zju.edu.cn/MCA/gallery.html ),12 and then identify the cell types in combination with their expression. In addition, we can collect marker genes of certain cell types in the literature. Second, the expression profiles of genes in unknown cell clusters and known cell types are used for similarity analysis. If the similarity was high, it would be identified as this kind of cell.13,14 For example, the R package (SingleR) can complete this analysis.15 Third, using the expression profiles of known cell types to construct classifiers as the training sets, and the gene expression profiles of unknown cell clusters are input for classification and identification.13,14 For example, the R package (Garnett) can be used for this analysis.16 Although more and more automatic cell type annotation tools have been developed, it is difficult to ensure that an automatic cell type identification tool is suitable for all cell types.17 Therefore, researchers should select one of the defined results as a reference, and name the corresponding cell clusters in combination with manual annotation and relevant knowledge background. In any case, the specific marker genes are still the basis for defining cell clusters.13,14 Generally, specific marker genes are selected according to the discipline’s background knowledge, literature, and databases. However, distinguishing a variety of myeloid cells in the CNS is not easy, because of the cross and instability of these cell markers.8 For example, adgre1 (F4/80), the established marker for macrophages,18,19 is also expressed in monocytes, microglia, and dendritic cells.20P2ry12 and Tmem119, which are microglia markers, are often downregulated or even negative under the conditions of CNS injury, inflammation, and degeneration.21,23 So, establishing a simple and practical cell type identification method (CTIM) to distinguish these cell populations is of great significance.

Material and methods

Excel template design for CTIM

Based on CellMarker (http://xteam.xbio.top/CellMarker/ ),10 PanglaoDB (https://panglaodb.se/ ),11 Mouse Cell Atlas (http://bis.zju.edu.cn/MCA/gallery.html ), combining with the recent pieces of literature,2–4,6,8,19,23–34 a simple Excel template for CTIM was designed, in which a panel of gene makers corresponding to the myeloid cells, lymphocytes, common CNS cells, and proliferative cells were included (Fig. 1 and Table S1). Here, myeloid cells included monocytes (MNCs), macrophages (MACs), microglia (MG), granulocytes (mainly neutrophils, NEUTs), and dendritic cells (DCs). To minimize the effects of lymphocytes on myeloid cell identities, T, B, and natural killer cell (referred to as NK)-specific gene markers were also listed in the table.

Excel template design for cell type definitions.
Fig. 1  Excel template design for cell type definitions.

A panel of gene markers corresponding to the myeloid cells and lymphocytes were included in the template. B, B lymphocyte; DC, dendritic cell; MAC, macrophage; MNC, monocyte; MG, microglia; NK, natural killer cell; NK/T, natural killer T cell; NEUT, neutrophil; T, T lymphocyte. P and N indicate positive and negative the gene markers, respectively. If the markers could be either positive or negative, we defined them as P/N.

Excel template design for gene markers and expression extraction

To perform the cell identification of a cluster, four Excel sheets: cell definition (Figs. 1 and 2e), cluster data (Fig. 2a), avg_logFC extraction (Fig. 2b and d), and gene extraction (Fig. 2c). In cluster data table, column A was the genes in a cluster, and column B was avg_logFC (average Log2 fold change), it was the ratio of the normalized mean gene counts in each cluster relative to all other clusters for comparison. The reason was that the count, transcripts per million, or fragments per kilobase of exon model per million mapped fragments were usually used, the gene expression value must be non-negative, and the value of fold change must be positive. When gene A expression was lower than gene B, the fold change of B on A was >1, and the fold change of log2 was >0; On the contrary, the fold change of log2 was <0. Based on this, we could display the upregulated (red) or downregulated (green) gene expression with different colors in the Excel template. In some reports, the average value of gene expression was also used. In the avg_logFC extraction table, the data in columns A and B should come from the corresponding columns of the cluster data table, column C extracted genes from column C of the gene extraction table, and column D extracted values from column C using the Excel command: VLOOKUP(Cn, A:B,2,0). In the gene extraction table, the data in column A were the gene markers from column B of the cell definition table, column B was the genes from column A of the avg_logFC extraction table, and column C was extracted values from column A using Excel command: IF(COUNTIF(B:B,An)>0,An,"").

Excel template and CTIM workflow.
Fig. 2  Excel template and CTIM workflow.

(a) Cluster data to be analyzed. (b) avg_logFC extraction. (c) Gene extraction. (d) Value extraction. (e) Cell definition: Column L, M and N mean any column (such as C1, C2, and Cn).

CTIM workflow

The workflow of CTIM included the following steps: (1) Copy columns A and B from the cluster data table, and paste them to the corresponding columns A and B of avg_logFC extraction table; (2) Copy column A from avg_logFC extraction table, and paste it to the column B of gene extraction table, then the extracted genes will be obtained from gene markers (column A); (3) Copy column C from gene extraction table, and paste as values to the column C of avg_logFC extraction table, then the extracted values will be shown in column D; (4) Copy column D from avg_logFC extraction table, and paste as values to any column you like (such as C1, C2, and Cn) in the cell definition table; (5) In cell definition table, the cell identities can be performed by comparing the extracted values (upregulated and downregulated genes are shown as red and green, respectively) to the cell types (column A) and gene markers (column B). Finally, the cell types were identified based on the upregulated markers (Fig. 2).

Data

Normalized and clustered data used in this study were obtained from previous studies.12,35–37 The reason for choosing these data was they could be directly downloaded, which allowed the authors to compare their analysis with the original reports. The data are shown in Table 1 and as an Excel worksheet in Figure 2a.

Table 1

Sources of the gene expression data used in this study

DataMiceTissueSingle cellscRNA-SeqClusteringCluster annotation
Ximerakis et al.35C57BL/6J mice (male, 2–3 months of age, and 21–22 months of age)A total of 8 young and 8 old brainsDissociated brainChromium Single Cell 3′ Chip (10x Genomics), the sequencing was performed on NextSeq 500 instrument (Illumina)Seurat package (v.2.3) in R (v.3.3.4)Using multiple cell type-specific/enriched marker genes that have been previously described in the literature (Plac8 for MNC)
Han et al.12Wild-type C57BL/6J mice (SPF, female, 6–10 week-old)Brain, blood, and bone marrowBrain was dissociated using accutase; bone marrow was treated red blood cell lysis buffer; blood was treated red blood cell lysis buffer or Ficoll separationMicrowell-Seq, the 3′ ends of the transcripts are then enriched during library generation using PCR and sequenced using the Illumina HiSeq platformSeurat was used for dimension reduction, clustering, and differential gene expression analysisSingle cell MCA (scMCA) analysis built by authors (Fig. 7A)
Sankowski et al.36 embj20211
08605-sup-0008-datasetev1SPF and GF C57BL/6J mice (mixed sex, 6–10 weeks old)The brain parenchyma, choroid plexus, leptomeninges, and perivascular space (20 mice per group)Parenchyma and perivascular space cells were isolated using Percoll gradient. The choroid plexuses and leptomeninges were treated by mechanical dissociation through a 70 micron cell strainer. Viable CD11b+CD45+CD3
B220Ly6Gcells were FACS-isolatedHigh-throughput scRNA-Seq using the high-sensitivity method mCEL-Seq2, the sequencing was performed on Illumina HiSeq 3000 sequencing system (pair-end multiplexing run) at a depth of 130,000–200,000 reads per cellSeurat version 3Generating maps for the myeloid cell populations based on published signature genes (Jordao et al.33). Fig. 1B
Mimouna et al.37C57BL/6 mice (mixed sex, 6–10 weeks old)EAE mouse spinal cordCNS-infiltrating cells were isolated using Percoll density gradient. F4/80+CD11b+CD45+ cells were sorted using FACSChromium Single Cell 3′ Chip (10x Genomics), The sequencing was performed on the Illumina NovaSeq system using a 28-8-98 paired-end cycleR version 4.0.1 software (R Core Team, 2019), fastMNN implementation, Louvain graph-based community clusteringCluster-specific markers were searched using the Wilcoxon rank-sum test. An automated cell type assignment was performed with singleR using training sets derived from the Immunological Genome Project database. PanglaoDB was used to identify putative cell and/or activation state for each individual Louvain cluster. The cell type and cell activation state transitions were identified by performing trajectory analysis with slingshot

Statistical analysis

To test the consistency of this CTIM with previous reports, the identification results were divided into three grades, excellent, satisfactory, and poor (Table 2). Bowker’s test and kappa symmetric measures were used to test the difference and consistency of the paired data between the two groups. For Bowker’s test, p < 0.05 was considered to be a statistically significant difference. For kappa symmetric measures, kappa ≥ 0.75 indicated good consistency, 0.4 ≤ kappa < 0.75 indicated general consistency and kappa < 0.4 indicated poor consistency. Data were analyzed with SPSS software v.26 (IBM Corp., Armonk, NY, USA).

Table 2

Grade evaluation criterion of cell type identities

ConsistencyAccuracyGrade
ConsistentBoth completely accurateBoth excellent (A)
Both partially accurateBoth satisfactory (B)
Neither is accurateBoth poor (C)
NonconsistentOne is completely accurateExcellent (A)
One is partially accurateSatisfactory (B)
One is not accuratePoor (C)

Results

Descriptive comparison of the CTIM with the literature in CNS myeloid cells

Using the CTIM, CNS myeloid cells in four data sources reported in the literature were identified (Table 1).12,35–37 In supplementary Table 3 of Ximerakis et al.,35 the authors listed the most discriminating genes per cell type. From that table, MNCs, MACs, MG, NEUTs, DCs, neuronal-restricted precursors (NRPs), immature neurons, mature neurons, astrocyte-restricted precursors, astrocytes, oligodendrocyte precursor cells, oligodendrocytes, ependymocytes, and hypendymal cells were chosen as gold standard cells to test the CTIM. As shown in Figure 3, Table 3, and Figure S1, of the 14 cell clusters, MNCs were identified as mixed with a few NEUTs and DCs, and NRPs as proliferative cells. The other 12 cell clusters were completely consistent.

Table 3

Comparison of cell types identified with data from Ximerakis et al35

ClusterReported cell typeOur cell typeConsistencyReason
MNCMNCMNC (mixed with a few NEUT and DC)PartPlac8 is also expressed in NEUT and DC
MACMACMACYesNR
MGMGMGYesNR
NEUTNEUTNEUTYesNR
DCDCDCYesNR
NRPNRPProliferative cellsNANot within the scope of our evaluation.
ImmNImmNNeuronYesNR
mNEURmNEURNeuronYesNR
ARPARPASTYesNR
ASTASTASTYesNR
OPCOPCOPCYesNR
OLOLOLYesNR
EPCEPCEpendymalYesNR
HypEPCHypEPCEpendymalYesNR
Representative results and heatmap of cell type identification by CTIM.
Fig. 3  Representative results and heatmap of cell type identification by CTIM.

MNC, MAC, MG, NEUT, DC, NRP, ImmN, mNEUR, ARP, AST, OPC, OLs, EPC, and HypEPC by Ximerakis, et al.35 were used to test cell type identification Excel template and seurat package. Of the 14 cell clusters, MNC was identified as MNC (mixed with a few NEUTs and DCs), and NRP as proliferative cells. The other 12 cell clusters were completely consistent. The gene expression levels were showed as Log2 Fold Change. Upregulated genes are shown in red (>0), and downregulated genes in green (<0). The depth of color respectively indicates the extent of up or downregulation. If the genes were not found in Cluster data, they would be shown as “N/A”. ARP, astrocyte-restricted precursor; AST, astrocyte; CTIM, cell type identification method; DC, dendritic cell; EPC, ependymocyte; HypEPC, hypendymal cell; ImmN, immature neuron; MAC, macrophage; MG, microglia; MNC, monocyte; mNEUR, mature neuron; NEUT, neutrophil; NRP, neuronal-restricted precursor; OL, oligodendrocyte; OPC, oligodendrocyte precursor cell.

Table 4 shows the results of the comparison of cell types identified in adult mouse brains. Fifteen clusters of adult mouse brains from Han et al.12 were identified. In the 15 cell clusters, pan-GABAergic and Schwann cells were not in the CTIM, the reported cluster 4 (Macrophage_Klf2 high) was mixed with a few MG, and the other 12 cell clusters were completely consistent. The CD11b+CD45+CD3B220-Ly6G cells isolated using fluorescence-activated cell sorting from adult mouse brain parenchyma, choroid plexus, leptomeninges, and perivascular space (embj2021108605-sup-0008-datasetev1) by Sankowski et al.36 were compared. As shown in Table 5, in the 17 cell clusters, 14 were completely consistent. The nonconsistent clusters included cluster 15 because it included stromal cells, which was not in our table. The reported cluster 6 (CNS-associated macrophages, CAMs) may have been Kolmer epiplexus cells that are reported to express microglial markers, and cluster 9 (CAMs), genes expressed in MACs were not increased.34

Table 4

Comparison of the cell type identified in adult brain with data from Han et al12

ClusterReported cell typeOur cell typeConsistencyReason
1Myelinating oligodendrocyteOLYesNR
2MicrogliaMGYesNR
3Astrocyte_Mfe8 highASTYesNR
4Macrophage_Klf2 highMAC/MGPartThe reported cluster 4 was mixed with a few MG
5Astrocyte_Atp1b2 highASTYesNR
6Oligodendrocyte precursor cellOPCYesNR
7NeuronNeuronYesNR
8Macrophage_Lyz2 highMACYesNR
9Astroglial cell (Bergman glia)ASTYesNR
10Pan-GABAergicProliferative cellsNANot within the scope of our evaluation.
11Astrocyte_Pla2g7 highASTYesNR
12Schwann cellUnknownNANot within the scope of our evaluation.
13Granulocyte_Il33 highNEUTYesNR
14Hypothalamic ependymal cellEpendymal cellsYesNR
15Granulocyte_Ngp highNEUTYesNR
Table 5

Comparison of the cell type identifies with data from Sankowski et al36

ClusterReported cell typeOur cell typeConsistencyReason
C0MGMGYesNR
C1CAMsMACYesNR
C2MGMGYesNR
C3CAMsMACYesNR
C4CAMsMACYesNR
C5MGMGYesNR
C6CAMsMGNoThe expression of typical genes of MAC including Mrc1, Cd163, Lyve1, Pf4, Ms4a7, Stab1, and Cbr2 were not elevated. In contrast, MG-specific markers Hex, Olfml3, and Sparc were significantly elevated. This might be Kolmer perplexes cells that are reported to express “microglial markers” (Van Hove et al., 2019)34
.C7CAMsMACYesNR
C8Ly6clow monocytesMNCYesNR
C9CAMsUnknownNAThe expression of typical genes of MAC including Mrc1, Cd163, Lyve1, Pf4, Ms4a7, Stab1, and Cbr2 were not elevated. The other genes were not within the scope of our evaluation.
C10MGMGYesNR
C11Ly6chi monocytesMNCYesNR
C12DCsDCYesNR
C13CAMsMACYesNR
C14Proliferating. cellsProliferating cellsYesNR
C15Stromal cellsUnknownNANot within the scope of our evaluation.
C16LymphocytesNKYesNR

We encountered some thorny problems when analyzing the data of Mimouna et al.37 In that data source, Louvain graph-based community clustering was used to divide the cells into clusters, and PanglaoDB was used to identify putative cell and/or activation state for each individual Louvain cluster. The cell types identified using CTIM are shown in Table 6. Although the results were basically consistent, the cell types were mixed, which indicated that the cell clustering for this data was not perfect.

Table 6

Comparison of the cell type analysis with data from Mimouna et al37

ClusterReported cell typeOur cell typeConsistencyReason
C1MAC/MG/othersMAC/MG/othersYesCell clustering was not ideal.
C2MAC/MG/NEUTMAC/MG/NEUTYesCell clustering was not ideal
C3MNC/MAC/MGMAC/MG/NEUTPartCell clustering was not ideal
C4MAC/MG/NEUTMAC/MG/NEUTYesCell clustering was not ideal
C5MNC/MACMAC/MG/NEUTPartCell clustering was not ideal
C6NEUTMAC/MG/NEUTPartCell clustering was not ideal
C7MAC/MG/othersMAC/MG/NEUTYesCell clustering was not ideal
C8T/othersMAC/MG/NEUTPartCell clustering was not ideal
C9MNC/MACMAC/MG/NEUTPartCell clustering was not ideal

Comparison of the CTIM with the literature in peripheral blood and bone marrow myeloid cells

To test the identification of non-CNS myeloid cells by CTIM, 21 peripheral blood cell clusters and 17 bone marrow cell clusters of adult mice from Han et al.12 were employed. Table 7 shows the peripheral blood results. Of the 21 cell clusters, cluster 14 (Erythroblast_Car2 high), cluster 20 (B cell_Igha high), and cluster 21 (Erythroblast_Hba-a2 high) were not in the table. The reported cluster 18 (Macrophage_Pf4 high) included a few NEUTs, the other 17 cell clusters were completely consistent. The bone marrow results are shown in Table 8. Of the 17 cell clusters, cluster 3 (neutrophil progenitors), cluster 8 (hematopoietic stem progenitor cells), cluster 9 (erythroblasts), and cluster 15 (mast cells) were not in the table, the other 14 cell clusters were completely consistent.

Table 7

Comparison of the cell type identified in peripheral blood with data from Han et al12

ClusterReported cell typeOur cell typeConsistencyReason
1T cell_Trbc2 highTYesNR
2B cell_Ly6d highBYesNR
3Macrophage_S100a4 highMACYesNR
4Neutrophil_Retnlg highNEUTYesNR
5Neutrophil_Ltf highNEUTYesNR
6Neutrophil_Camp highNEUTYesNR
7Neutrophil_Il1b highNEUTYesNR
8NK cell_Gzma highNKYesNR
9Macrophage_Ace highMACYesNR
10Monocyte_Elane highMNCYesNR
11B cell_Vpreb3 highBYesNR
12Monocyte_F13a1 highMNCYesNR
13T cell_Gm14303 highTYesNR
14Erythroblast_Car2 highProliferative cellsNANot within the scope of our evaluation.
15B cell_Rps27rt highBYesNR
16Dendritic cell_Siglech highDCYesNR
17Basophil_Prss34 highUnknownNANA
18Macrophage_Pf4 highMAC/NEUTPartThe reported cluster 18 was mixed with a few NEUT.
19B cell_Igha highUnknownNANot within the scope of our evaluation.
20Macrophage_Flt-ps1 highMACYesNR
21Erythroblast_Hba-a2 highUnknownNANot within the scope of our evaluation.
Table 8

Comparison of the cell type identified in bone marrow with data from Han et al12

ClusterReported cell typeOur cell typeConsistencyReason
1Neutrophil_Cebpe highNEUTYesNR
2Neutrophil_Mmp8 highNEUTYesNR
3Neutrophil progenitorMNC/MAC/NEUTNANot within the scope of our evaluation.
4Monocyte_Prtn3 highMNCYesNR
5Macrophage_Ms4a6c highMACYesNR
6Neutrophil_Ngp highNEUTYesNR
7Prepro B cellBYesNR
8Hematopoietic stem progenitor cellUnknownNANot within the scope of our evaluation.
9ErythroblastProliferative unknown cellNANot within the scope of our evaluation.
10Neutrophil_Fcnb highNEUTYesNR
11B cell_Igkc highBYesNR
12Macrophage_S100a4 highMACYesNR
13T cell_Ms4a4b highTYesNR
14Dendritic cell_Siglech highDCYesNR
15Mast cellUnknownNANot within the scope of our evaluation.
16Dendritic cell_H2-Eb1 highDCYesNR
17Monocyte_Mif highMNCYesNR

Results of the CTIM compared with the published literature

According to the grading evaluation method in Table 2, the results of all data analysis (Tables 38) were evaluated. Excluding those clusters that are not within the scope of the analysis (N/A), a total of 83 valid cases were obtained. As shown in Table 9, excellent, satisfactory, and poor results in previous studies were 74, 3, and 6, respectively. Correspondingly, they were 77, 1, and 5 in the results of CTIM. The overall consistency rate was 93.98% (78/83). Bowker’s test showed that there was no significant difference between the two groups (p > 0.05). Kappa symmetric measures showed that the kappa value was 0.642 (p < 0.01), indicating general consistency.

Table 9

Bowker’s test and kappa symmetric measures of literature and our results

Studies * CTIM crosstabulation
GradingGrading (CTIM)
Total
A (excellent)B (satisfactory)C (poor)
Grading (studies)A731074
B3003
C1056
Total771583

Discussion

For the last few decades, many advanced techniques, such as immunohistochemistry, flow cytometry, etc. have been used to identify CNS myeloid cell-subtypes. However, owing to the lack of absolutely specific markers and unstable expression of biomarkers under different pathophysiological conditions, their accuracy is still not satisfactory.8 Although, scRNA-Seq is a promising new technology to solve this problem, for ordinary researchers, various programming language analysis packages for scRNA-Seq data are not an easy task, and bioinformatics experts do not necessarily know the specific markers of CNS myeloid cell-subtypes.9 Therefore, building a bridge to connect the knowledge gap between ordinary researchers and bioinformatics experts is important.

In this study, a Microsoft-Excel template was designed, in which a panel of gene makers corresponding to myeloid cells, lymphocytes, common CNS cells, and proliferative cells were included. For users, as long as the gene expression data of cell clusters are obtained, the clusters can be named directly using this Excel template. It should be emphasized that the template is mainly suitable for determining the major categories of myeloid cells. If researchers need to further distinguish the subtypes of certain cells, it is only needed to add corresponding gene markers. This Excel template is open source, and researchers can modify or add new genes based on their needs (Table S1). For the selection of gene markers, we considered not only the relative specificity but also the crossover and commonality of different cells. In the Excel template, the letters P and N mean the gene markers are positive or negative. If the markers are positive or negative, they are defined as “P/N” (Fig. 1). For example, Ptprc (the gene of CD45) is a common marker of myeloid cells and lymphocytes.38–40 It was used as a common marker of myeloid cells and lymphocytes to distinguish CNS nonmyeloid cells (astrocytes, oligodendrocytes, neurons, etc.). In addition, in theory, the protein molecule CD45 expressed by Ptprc gene is positive in many leukocytes, but in the process of collecting gene markers and drawing the Excel template, we found that Ptprc gene was not expressed in every cell cluster, so it was defined as P/N. In addition to Ptprc, there were many similar examples (see Fig. 1 and Table S1 for details). For a certain cell, although there are some relatively specific gene markers, a panel of gene markers was still used to comprehensively evaluate and then define them. This could effectively distinguish the cell types with similar or cross gene expression and ensure the accuracy of cell cluster identification. In this Excel template, there were 73 gene markers (excluding nonmyeloid CNS cells) in each panel that could be used to distinguish myeloid cell-subtypes and lymphocytes (Fig. 1). For example, MNC could express Ptprc (P/N), Cd14 (P/N), Itgam (P/N), Itgax (P/N), Csf3r (P/N), Adgre1(P/N), Ly6c1 (P/N), S100a4 (P/N), Cd68 (P), Ly86 (P/N), Ctsb (P/N), Ccr2 (P/N), Ly6c2 (P), Plac8 (P), Pf4 (P/N), Lyz1 (P), Hmox1 (P/N), F13a1(P), Lyst (P/N), Prtn3 (P/N), Elane (P/N), and Pilra (P/N). Although several molecules (Cd68, Ly6c2, Plac8 and Lyz1) are positive (P) in MNC, they are also expressed in other cells. So, there were no absolute specific markers of MNC in this template. Nevertheless, we could still determine its cell type using comparative analysis. For those cell types with their own specific gene markers, it was easy to identify cell clusters using comparative analysis. Typical examples were Ms4a7, Lyve1, Cbr2, Mrc1, and Cd163 for MAC; Hexb, Olfml3, Sparc, Tgfbr1, P2ry12, and Tmem119 for MG; Ltf, Ly6g, Mmp8, Camp, Ngp, Fcnb, Cebpe, Retnlg, S100a8, S100a9, Lcn2, G0s2, Wfdc21 for NEUT. Of course, because of limitations of knowledge background and research level, this Excel template still has some defects. For example, for DCs, the expressions of H2-Ab1, H2-Eb1, H2-Aa, Cd74, and Cd209a should be positive, but these markers can also be expressed in MAC and B cells, especially B cells, are not myeloid cells, which is easy to result in misidentification. In this template, B cell markers were also added to facilitate distinguishing B cells from DC. In addition, it should be aware of Kolmer epiplexus cells which were reported to express “microglial markers” like P2ry12 as well.34–40 Kolmer epiplexus cells, first reported by Kolmer in 1921, are a population of macrophages that attach to the ventricle-facing surface of the choroid plexus.41,42 The gene transcription of these cells is more consistent with microglia than nonparenchymal macrophages. In addition, Kolmer epiplexus cells have the same ontogenetic and self-renewal ability as microglia, so they are considered a nonparenchymal microglia subtype.34,41 Therefore, we should be careful with the interpretation and definition of microglia and macrophages when encountering suspected Kolmer epiplexus cells. For example, in the cluster 6 of Table 5, the typical gene markers of MAC, including Mrc1, Cd163, Lyve1, Pf4, Ms4a7, Stab1, and Cbr2, were not increased. In contrast, MG specific markers, Hexb, Olfml3, and Sparc, were significantly increased. This might be identified as Kolmer epiplexus cells.

Compared with the findings of Ximerakis et al.,35 only one cluster was inconsistent (Table 3). Our results showed that there were a few NEUT and DC mixed with their MNC. The possible reason was that they took Plac8 as a specific marker of MNC. In fact, Plac8 is also expressed in NEUT and DC.12 Compared with Han et al.,12 in the cell type identified of adult brain, the cluster 4 was inconsistent (Table 4). The reason may be that the reported cluster 4 was mixed with a few MG, because we could find the typical microglia markers (Hexb, Olfml3, Sparc, Tgfbr1, P2ry12, and Tmem119). Compared with the findings of Sankowski et al.,36 the clusters 6 and 9 were inconsistent (Table 5). Both clusters were identified as CAMs, however, the expression of typical genes of MACs (Mrc1, Cd163, Lyve1, Pf4, Ms4a7, Stab1, and Cbr2) was not increased in both clusters. In contrast, MG specific markers (Hexb, Olfml3, and Sparc) were significantly increased in cluster 6, while the other genes in cluster 9 were not in our table. Comparing with the cell type identified in peripheral blood and bone marrow of Han et al.,12 excepting cluster 18 of peripheral blood was mixed with a few NEUT, the others were completely consistent. These indicated that our Excel template was also very effective for the analysis of non-CNS myeloid cells.

From the above analysis, it can be deduced that the appropriate gene markers and ideal scRNA-Seq data clustering are key factors for the accuracy of cell definition. The importance of cell clustering can be understood by the following example. When the data reported by Mimouna et al.37 were analyzed, both the reported and the CTIM were not ideal. Analyzing the reasons, it was found that their data clustering methods were different from those used in other studies. The cell clustering method in this literature was Louvain graph-based community clustering, which may be the reason why the clustering was not ideal. Although this Excel template still could be used to identify the cell types based on the author’s data, the cell types in each of the nine clusters were mixed (Table 6). Therefore, the data used in this Excel template should be processed through the standard scRNA-Seq analysis process, including quality control, standardization, data correction, feature selection, and data dimensionality reduction, finally, the cells were divided into different clusters according to the similarity of gene expression.

Conclusions

The Excel template can be a bridge to span the knowledge gap between ordinary researchers and bioinformatics experts. For ordinary researchers without a foundation in computer language programming, it can easily distinguish myeloid cell-subtypes and nonmyeloid cells by using a panel of gene markers for cell clustering data of CNS. For bioinformatics experts, it is also a valuable reference for selecting gene markers. It will also encourage researchers pertaining to different fields interested in utilizing the ever-growing scRNA-Seq data to design similar templates and pipelines for their specific cell population.

Supporting information

Supplementary material for this article is available at https://doi.org/10.61474/ncs.2023.00004 .

Table S1

Excel template design for CTIM.

(XLSX)

Fig. S1

Visual gene expression heatmap of Figure 3. The results of Figure 3 were extracted and used to create a separate heatmap. Upregulated genes are shown in red (>0). Downregulated genes are shown in green (<0). Depth of color indicates the extent of upregulation or downregulation.

(TIF)

Abbreviations

ARP: 

astrocyte-restricted precursor

AST: 

astrocyte

B: 

B lymphocyte

CAM: 

CNS-associated macrophage

CNS: 

central nervous system

CTIM: 

cell type identification method

DC: 

dendritic cell

EAE: 

experimental autoimmune encephalomyelitis

EPC: 

ependymocyte

FACS: 

fluorescence-activated cell sorting

GF: 

germ-free

HypEPC: 

hypendymal cell

ImmN: 

immature neuron

MAC: 

macrophage

MG: 

microglia

MNC: 

monocyte

mNEUR: 

mature neuron

NA: 

not available

NEUT: 

neutrophil

NK: 

nature killer cell

NK/T: 

natural killer T cell

NR: 

not relevant

NRP: 

neuronal-restricted precursor

OL: 

oligodendrocyte

OPC: 

oligodendrocyte precursor cell

scMCA: 

A tool defines cell types in mouse based on single-cell digital expression

scRNA-Seq: 

single-cell RNA sequencing

SPF: 

specific pathogen free

T: 

T lymphocyte

Declarations

Funding

This study was supported by grant from the National Natural Science Foundation of China (82072416).

Conflict of interest

The manuscript was submitted during Dr. He-Zuo Lü's term as an editorial board member of Nature Cell and Science. The authors have no other conflict of interest to declare.

Authors’ contributions

Study design, data interpretation and writing (HZL, JGH) literature search, data collection, data analysis, and generation of tables and figures (XYL, JLL, SQD). All authors made a significant contribution to this study and have approved the final manuscript.

References

  1. Croese T, Castellani G, Schwartz M. Immune cell compartmentalization for brain surveillance and protection. Nat Immunol 2021;22(9):1083-1092 View Article PubMed/NCBI
  2. Prinz M, Erny D, Hagemeyer N. Ontogeny and homeostasis of CNS myeloid cells. Nat Immunol 2017;18(4):385-392 View Article PubMed/NCBI
  3. Herz J, Filiano AJ, Wiltbank AT, Yogev N, Kipnis J. Myeloid Cells in the Central Nervous System. Immunity 2017;46(6):943-956 View Article PubMed/NCBI
  4. Ajami B, Samusik N, Wieghofer P, Ho PP, Crotti A, Bjornson Z, et al. Single-cell mass cytometry reveals distinct populations of brain myeloid cells in mouse neuroinflammation and neurodegeneration models. Nat Neurosci 2018;21(4):541-551 View Article PubMed/NCBI
  5. Manouchehri N, Hussain RZ, Cravens PD, Esaulova E, Artyomov MN, Edelson BT, et al. CD11c(+)CD88(+)CD317(+) myeloid cells are critical mediators of persistent CNS autoimmunity. Proc Natl Acad Sci U S A 2021;118(14):e2014492118 View Article PubMed/NCBI
  6. Schwabenland M, Brück W, Priller J, Stadelmann C, Lassmann H, Prinz M. Analyzing microglial phenotypes across neuropathologies: a practical guide. Acta Neuropathol 2021;142(6):923-936 View Article PubMed/NCBI
  7. David S, Greenhalgh AD, Kroner A. Macrophage and microglial plasticity in the injured spinal cord. Neuroscience 2015;307:311-318 View Article PubMed/NCBI
  8. Quintana FJ. Myeloid cells in the central nervous system: So similar, yet so different. Sci Immunol 2019;4(32):eaaw2841 View Article PubMed/NCBI
  9. Cembrowski MS. Single-cell transcriptomics as a framework and roadmap for understanding the brain. J Neurosci Methods 2019;326:108353 View Article PubMed/NCBI
  10. Zhang X, Lan Y, Xu J, Quan F, Zhao E, Deng C, et al. CellMarker: a manually curated resource of cell markers in human and mouse. Nucleic Acids Res 2019;47(D1):D721-D728 View Article PubMed/NCBI
  11. Franzén O, Gan LM, Björkegren JLM. PanglaoDB: a web server for exploration of mouse and human single-cell RNA sequencing data. Database (Oxford) 2019;2019:baz046 View Article PubMed/NCBI
  12. Han X, Wang R, Zhou Y, Fei L, Sun H, Lai S, et al. Mapping the Mouse Cell Atlas by Microwell-Seq. Cell 2018;172(5):1091-1107.e17 View Article PubMed/NCBI
  13. Zhao X, Wu S, Fang N, Sun X, Fan J. Evaluation of single-cell classifiers for single-cell RNA sequencing data sets. Brief Bioinform 2020;21(5):1581-1595 View Article PubMed/NCBI
  14. Huang Q, Liu Y, Du Y, Garmire LX. Evaluation of Cell Type Annotation R Packages on Single-cell RNA-seq Data. Genomics Proteomics Bioinformatics 2021;19(2):267-281 View Article PubMed/NCBI
  15. Aran D, Looney AP, Liu L, Wu E, Fong V, Hsu A, et al. Reference-based analysis of lung single-cell sequencing reveals a transitional profibrotic macrophage. Nat Immunol 2019;20(2):163-172 View Article PubMed/NCBI
  16. Pliner HA, Shendure J, Trapnell C. Supervised classification enables rapid annotation of cell atlases. Nat Methods 2019;16(10):983-986 View Article PubMed/NCBI
  17. Zhang AW, O’Flanagan C, Chavez EA, Lim JLP, Ceglia N, McPherson A, et al. Probabilistic cell-type assignment of single-cell RNA-seq for tumor microenvironment profiling. Nat Methods 2019;16(10):1007-1015 View Article PubMed/NCBI
  18. Lee MN, Lee Y, Wu D, Pae M. Luteolin inhibits NLRP3 inflammasome activation via blocking ASC oligomerization. J Nutr Biochem 2021;92:108614 View Article PubMed/NCBI
  19. Schulz C, Gomez Perdiguero E, Chorro L, Szabo-Rogers H, Cagnard N, Kierdorf K, et al. A lineage of myeloid cells independent of Myb and hematopoietic stem cells. Science 2012;336(6077):86-90 View Article PubMed/NCBI
  20. Summers KM, Bush SJ, Hume DA. Network analysis of transcriptomic diversity amongst resident tissue macrophages and dendritic cells in the mouse mononuclear phagocyte system. PLoS Biol 2020;18(10):e3000859 View Article PubMed/NCBI
  21. Kenkhuis B, Somarakis A, de Haan L, Dzyubachyk O, IJsselsteijn ME, de Miranda NFCC, et al. Iron loading is a prominent feature of activated microglia in Alzheimer’s disease patients. Acta Neuropathol Commun 2021;9(1):27 View Article PubMed/NCBI
  22. Zrzavy T, Hametner S, Wimmer I, Butovsky O, Weiner HL, Lassmann H. Loss of ‘homeostatic’ microglia and patterns of their activation in active multiple sclerosis. Brain 2017;140(7):1900-1913 View Article PubMed/NCBI
  23. Milich LM, Choi JS, Ryan C, Cerqueira SR, Benavides S, Yahn SL, et al. Single-cell analysis of the cellular heterogeneity and interactions in the injured mouse spinal cord. J Exp Med 2021;218(8):e20210040 View Article PubMed/NCBI
  24. Niehaus JK, Taylor-Blake B, Loo L, Simon JM, Zylka MJ. Spinal macrophages resolve nociceptive hypersensitivity after peripheral injury. Neuron 2021;109(8):1274-1282.e6 View Article PubMed/NCBI
  25. Abe N, Nishihara T, Yorozuya T, Tanaka J. Microglia and Macrophages in the Pathological Central and Peripheral Nervous Systems. Cells 2020;9(9):2132 View Article PubMed/NCBI
  26. Plemel JR, Stratton JA, Michaels NJ, Rawji KS, Zhang E, Sinha S, et al. Microglia response following acute demyelination is heterogeneous and limits infiltrating macrophage dispersion. Sci Adv 2020;6(3):eaay6324 View Article PubMed/NCBI
  27. Xiao Y, Hu X, Fan S, Zhong J, Mo X, Liu X, et al. Single-Cell Transcriptome Profiling Reveals the Suppressive Role of Retinal Neurons in Microglia Activation Under Diabetes Mellitus. Front Cell Dev Biol 2021;9:680947 View Article PubMed/NCBI
  28. Mrdjen D, Pavlovic A, Hartmann FJ, Schreiner B, Utz SG, Leung BP, et al. High-Dimensional Single-Cell Mapping of Central Nervous System Immune Cells Reveals Distinct Myeloid Subsets in Health, Aging, and Disease. Immunity 2018;48(2):380-395.e6 View Article PubMed/NCBI
  29. Somebang K, Rudolph J, Imhof I, Li L, Niemi EC, Shigenaga J, et al. CCR2 deficiency alters activation of microglia subsets in traumatic brain injury. Cell Rep 2021;36(12):109727 View Article PubMed/NCBI
  30. Wahane S, Zhou X, Zhou X, Guo L, Friedl MS, Kluge M, et al. Diversified transcriptional responses of myeloid and glial cells in spinal cord injury shaped by HDAC3 activity. Sci Adv 2021;7(9):eabd8811 View Article PubMed/NCBI
  31. David S, Kroner A, Greenhalgh AD, Zarruk JG, López-Vales R. Myeloid cell responses after spinal cord injury. J Neuroimmunol 2018;321:97-108 View Article PubMed/NCBI
  32. Utz SG, See P, Mildenberger W, Thion MS, Silvin A, Lutz M, et al. Early Fate Defines Microglia and Non-parenchymal Brain Macrophage Development. Cell 2020;181(3):557-573.e18 View Article PubMed/NCBI
  33. Jordão MJC, Sankowski R, Brendecke SM, Locatelli G, Tai YH, et al. Single-cell profiling identifies myeloid cell subsets with distinct fates during neuroinflammation. Science 2019;363(6425):eaat7554 View Article PubMed/NCBI
  34. Van Hove H, Martens L, Scheyltjens I, De Vlaminck K, Pombo Antunes AR, De Prijck S, et al. A single-cell atlas of mouse brain macrophages reveals unique transcriptional identities shaped by ontogeny and tissue environment. Nat Neurosci 2019;22(6):1021-1035 View Article PubMed/NCBI
  35. Ximerakis M, Lipnick SL, Innes BT, Simmons SK, Adiconis X, Dionne D, et al. Single-cell transcriptomic profiling of the aging mouse brain. Nat Neurosci 2019;22(10):1696-1708 View Article PubMed/NCBI
  36. Sankowski R, Ahmari J, Mezö C, Hrabě de Angelis AL, Fuchs V, Utermöhlen O, et al. Commensal microbiota divergently affect myeloid subsets in the mammalian central nervous system during homeostasis and disease. EMBO J 2021;40(23):e108605 View Article PubMed/NCBI
  37. Mimouna S, Rollins DA, Shibu G, Tharmalingam B, Deochand DK, Chen X, et al. Transcription cofactor GRIP1 differentially affects myeloid cell-driven neuroinflammation and response to IFN-β therapy. J Exp Med 2021;218(1):e20192386 View Article PubMed/NCBI
  38. Hermiston ML, Xu Z, Weiss A. CD45: a critical regulator of signaling thresholds in immune cells. Annu Rev Immunol 2003;21:107-137 View Article PubMed/NCBI
  39. Thomas ML. The leukocyte common antigen family. Annu Rev Immunol 1989;7:339-369 View Article PubMed/NCBI
  40. Rosenberg AB, Roco CM, Muscat RA, Kuchina A, Sample P, Yao Z, et al. Single-cell profiling of the developing mouse brain and spinal cord with split-pool barcoding. Science 2018;360(6385):176-182 View Article PubMed/NCBI
  41. Munro DAD, Bradford BM, Mariani SA, Hampton DW, Vink CS, Chandran S, et al. CNS macrophages differentially rely on an intronic Csf1r enhancer for their development. Development 2020;147(23):dev194449 View Article PubMed/NCBI
  42. Nakamura S, Koga N, Moriyasu N. [Epiplexus cell (Kolmer cell) and its reaction against foreign bodies]. No To Shinkei 1982;34(9):895-907 PubMed/NCBI
  • Nature Cell and Science
  • 2958-695X

A Microsoft-Excel Template for Identifying Mouse Myeloid Cell Types in the Central Nervous System Based on Single-cell RNA Sequencing Data

Xin-Yi Lyu, Jing-Lu Li, Shu-Qin Ding, Jian-Guo Hu, He-Zuo Lü
  • Reset Zoom
  • Download TIFF