Skip to main content

ORIGINAL RESEARCH article

Front. Genet., 08 March 2021
Sec. Computational Genomics
This article is part of the Research Topic Integrative Analysis of Genome-Wide Association Studies and Single-Cell Sequencing Studies View all 10 articles

Leveraging Single-Cell RNA-seq Data to Uncover the Association Between Cell Type and Chronic Liver Diseases

\r\nXiangyu Ye&#x;Xiangyu Ye1†Julong Wei&#x;Julong Wei2†Ming YueMing Yue3Yan WangYan Wang1Hongbo ChenHongbo Chen4Yongfeng ZhangYongfeng Zhang4Yifan WangYifan Wang4Meiling ZhangMeiling Zhang4Peng Huang*Peng Huang1*Rongbin Yu*Rongbin Yu1*
  • 1Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
  • 2Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, MI, United States
  • 3Department of Infectious Diseases, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
  • 4Department of Infectious Disease, Jurong Hospital Affiliated to Jiangsu University, Jurong, China

Background: Components of liver microenvironment is complex, which makes it difficult to clarify pathogenesis of chronic liver diseases (CLD). Genome-wide association studies (GWASs) have greatly revealed the role of host genetic background in CLD pathogenesis and prognosis, while single-cell RNA sequencing (scRNA-seq) enables interrogation of the cellular diversity and function of liver tissue at unprecedented resolution. Here, we made integrative analysis on the GWAS and scRNA-seq data of CLD to uncover CLD-related cell types and provide clues for understanding on the pathogenesis.

Methods: We downloaded three GWAS summary data and three scRNA-seq data on CLD. After defining the cell types for each scRNA-seq data, we used RolyPoly and LDSC-cts to integrate the GWAS and scRNA-seq. In addition, we analyzed one scRNA-seq data without association to CLD to validate the specificity of our findings.

Results: After processing the scRNA-seq data, we obtain about 19,002–32,200 cells and identified 10–17 cell types. For the HCC analysis, we identified the association between B cell and HCC in two datasets. RolyPoly also identified the association, when we integrated the two scRNA-seq datasets. In addition, we also identified natural killer (NK) cell as HCC-associated cell type in one dataset. In specificity analysis, we identified no significant cell type associated with HCC. As for the cirrhosis analysis, we obtained no significant related cell type.

Conclusion: In this integrative analysis, we identified B cell and NK cell as HCC-related cell type. More attention and verification should be paid to them in future research.

Introduction

Chronic liver disease (CLD) is a public health topic of global concern. As estimated, about 844 million people worldwide are suffering from CLD and 2 million deaths each year (Asrani et al., 2019). Starting with diverse etiology-related chronic hepatitis, CLD might develop into cirrhosis and hepatocellular carcinoma after repetitive liver damage (Gadd et al., 2020). Environment risk factors associated with CLD are virus, diet, drug, and autoimmune (Marcellin and Kutala, 2018). With the development of molecular biology, the role of host genetic background in CLD has also gained wide attention (Anstee et al., 2020). Genome-wide association studies (GWASs) have contributed greatly to our understanding of the genetic roles in CLD pathogenesis and prognosis (Matsuura et al., 2017). A number of associated polymorphisms, including variants on CDK14, SH2B3, CARD10, TLL1, PNPLA3, and HLA, have been reported (De Boer et al., 2014; Sudlow et al., 2015; Matsuura et al., 2017; Nicoletti et al., 2017; Li et al., 2018; Ishigaki et al., 2020; Schwantes-An et al., 2020). Nevertheless, the current understanding of CLD is far from enough, and it is still of great significance to further clarify the pathological process of CLD and explore new treatment strategy for CLD patients (Marcellin and Kutala, 2018).

As the largest internal organ of the body, the liver consists of many cell types, including not only epithelial cells and some non-parenchymal cells (e.g., endothelial and mesenchymal cells) but also a variety of immune cells (MacParland et al., 2018; Aizarani et al., 2019; Ramachandran et al., 2019; Sharma et al., 2020). Different cell types vary greatly in abundance and function, leading to their completely distinct roles in the physiological and pathophysiological processes of liver diseases (Ramachandran et al., 2020). Single-cell genomics technologies are transforming our understanding on diseases like CLD, enabling interrogation of cellular diversity and function at unprecedented resolution, and adding a new dimension to traditional bulk transcriptomic techniques (Giladi and Amit, 2018). Single-cell RNA sequencing (scRNA-seq) has been used to feature the fundamental liver biology and the cellular mechanisms underpinning liver regeneration (Aizarani et al., 2019). It also has been used to uncover the pathophysiological changes of hepatic fibrosis and hepatocellular carcinoma, where the heterogeneity and changes of T cells (Zheng C. et al., 2017), macrophages (Ramachandran et al., 2019), and endothelial cells (Sharma et al., 2020) residing within the liver tissue may be critical in driving disease states.

Both GWAS and scRNA-seq have thrown light on the way to indepthly understand the pathogenesis of CLD and further laid a foundation for the development of precision treatment strategy (Saviano et al., 2020). Integrating GWAS summary data and scRNA-seq data to identify the cell types associated to CLD might provide new clues for understanding the pathogenesis of CLD (Calderon et al., 2017; Finucane et al., 2018; Hao et al., 2020). Here, we used RolyPoly and LDSC-cts to ensure the robustness and confidence of the result. Especially, we first processed the scRNA-seq data to derive averaged expression vector and differential expression gene (DEG) list of each cell type for RolyPoly and LDSC-cts, respectively. Then, we used the Ensembl database to obtain the position relationship between SNPs and gene (Yates et al., 2019). Finally, with GWAS data, scRNA-seq data and block annotation in place, as well as accounting for linkage disequilibrium (LD) of related population, we applied RolyPoly and LDSC-cts to identify and prioritize CLD-relevant cell types.

Materials and Methods

Genome-Wide Association Studies Data

The first category of summary statistics is Asian ancestry GWAS. The datasets are from the Biobank of Japan (BBJ)1 (Ishigaki et al., 2020). We focus on the CLD-related phenotype that contain allele information and variant ID and that contain effect size and its standard error. With the two criteria, we obtained two GWAS summary statistics: cirrhosis (n = 212,453, prevalence = 1.03%) and HCC (n = 197,611, prevalence = 0.94%). Here, cirrhosis and HCC in BBJ were adjusted for age, sex, and top five genotype PCs (Ishigaki et al., 2020). The details of the two GWAS data are provided in Supplementary Table 1. Based on Asian ancestry from the 1000 Genome Project (1000 GP), we filtered out variants with minor allele frequency (MAF) < 0.01 and Hardy–Weinberg equilibrium (HWE) < 10–6 (Auton et al., 2015). After these quality control (QC) steps, we finally obtained 7,246,475 and 7,246,543 SNPs from the two datasets.

The second category of GWAS summary statistics is from European ancestry. The dataset is from GeneATLAS website2 (Canela-Xandri et al., 2018). We focus on the CLD-related phenotype that contain allele information and variant ID and that contain effect size and standard error. With the two criteria, we obtain one GWAS summary statistics: cirrhosis (n = 452,264, prevalence = 1.99%). This cirrhosis GWAS data was adjusted for sex, array batch, UK Biobank Assessment Center, age, age2 (Sudlow et al., 2015), and the top 20 genotype PCs as computed by UK Biobank. The details of these data are also provided in Supplementary Table 1. Based on European ancestry from the 1000 Genome Project, we filtered out variants with MAF < 0.01 and HWE < 10–6 (Auton et al., 2015). After these QC steps, we finally obtained 7,636,847 SNPs from this dataset.

We treated the phase 3 of the 1000 Genome Project as the reference panel (Auton et al., 2015). Here, we collected 503 European individuals and 504 East Asian individuals with 81,271,745 SNPs. We used PLINK to calculate Pearson’s r2 values of pairwise SNPs for RolyPoly with the default 1 MB window size (Chang et al., 2015). In LDSC-cts, we set the window size to 1 centiMorgan to estimate LD scores (Finucane et al., 2018).

Four Single-Cell Data

Considering the cirrhosis and HCC data acquired from GWAS, we searched the GEO database for related scRNA-seq data and obtained one data for liver cirrhosis and two for HCC, whose raw counts data are available (Barrett et al., 2012; Ramachandran et al., 2019; Losic et al., 2020). In addition, to verify the specificity of the outcomes, we also downloaded an idiopathic Parkinson’s disease (IPD) data. The details are provided in Supplementary Table 2. Following the original study, we performed QC and clustering for each scRNA-seq data. Note that scRNA-seq data usually have the potential to have its clusters continuously subdivided, but we just controlled the cell type number of each data within 10–20 depending on the features and quality of each data. The specific processing details of each data are as follows: After demultiplexing, aligning, and estimating cell-containing partitions and associated UMIs, a cirrhosis dataset (GSE136103) consisting of CD45 + and CD45-, blood and liver, healthy and cirrhosis, and human and mice samples were downloaded (Ramachandran et al., 2019). Here, we only chose nine human cirrhotic samples, including five CD45 + and four CD45- samples, for downstream analysis.

For scRNA-seq data analysis, we first removed potential doublets, and then excluded the cells that expressed fewer than 300 genes or mitochondrial gene content >30% of the total UMI count (Ramachandran et al., 2019). We also excluded genes expressed in fewer than three cells. We followed the analysis flow in Seurat (Stuart et al., 2019): (1) used SCTransform, a new strategy to remove the influence of technical characteristics while preserving biological heterogeneity via regularized negative binomial regression, to normalize and scale scRNA-seq data (Hafemeister and Satija, 2019); (2) used default setting of IntegrateData to remove the batch effect (Butler et al., 2018); (3) performed unsupervised clustering and differential gene expression analyses on the integrated data; (4) used principal component analysis (PCA) for linear dimension reduction, and then used shared nearest neighbor (SNN) graph-based clustering, in which the graph was constructed using the top 30 principal components; and (5) used UMAP to visualize by the same number of principal components (PCs) as the associated clustering, with perplexity ranging from 30 to 300 according to the number of cells in the dataset or lineage. The details of data processing are shown in Figure 1.

FIGURE 1
www.frontiersin.org

Figure 1. General procedure for scRNA-data processing.

In cell type definition, we referred to marker genes that are widely recognized and those from the original research. We used BuildClusterTree to assess cluster similarity by constructing the phylogenetic tree (Stuart et al., 2019). Totally, we identified 20 clusters on 23,184 cells (Supplementary Table 2 and Figure 2). Marker genes used for cell type definition are shown in Supplementary Table 3.

FIGURE 2
www.frontiersin.org

Figure 2. Cell types inferred from expression of marker gene signatures in GSE136103. NKT, natural killer T cells; Pdc, plasmacytoid dendritic cell; Treg, regulatory T cell; LEC, lymphatic endothelial cell; MDM, monocyte-derived macrophage; NK, natural killer cell; LSEC, liver sinusoids endothelial cell; DC, dendritic cell.

The first HCC dataset (GSE149614) contains 21 primary tumor, portal vein tumor thrombus (PVTT), metastatic lymph node, and non-tumor liver samples from 10 HCC patients. We downloaded the raw count data, which have been processed and aligned by Cell Ranger, and chose only 10 primary tumor samples for downstream analysis (Zheng G.X.Y. et al., 2017). After processing and clustering, we totally identified 14 cell types on 30,983 cells in this dataset (Supplementary Table 2).

Another HCC dataset (GSE112271) contains three and four tumor samples coming from different regions of two different individuals, and we included all seven samples for downstream analysis. After data processing, we totally identified 13 clusters on 32,200 cells in this dataset (Losic et al., 2020; Supplementary Table 2).

We downloaded the processed and aligned IPD dataset (GSE157783), which contains samples from six control and five idiopathic Parkinson’s disease cases. We chose only five disease samples for downstream analysis and totally identified 12 clusters on 19,002 cells following our procedure (Supplementary Table 2).

Defining the Specific Cell Types Associated With Cirrhosis and HCC

We used RolyPoly and LDSC-cts to define the specific cell types associated with cirrhosis and HCC (Calderon et al., 2017; Finucane et al., 2018). Based on polygenic model, RolyPoly treats the variance of each gene as the linear combination of each cell type and estimates the coefficients by method-of-moment. Then, RolyPoly uses block bootstrap to estimate the variance for the cell type effects, then construct t-statistics to test them (Efron and Tibshirani, 1986). By utilizing GWAS summary statistics for all SNPs near protein-coding genes, the model performed joint analysis with gene expression of a variety of cell types simultaneously, to define prioritized trait-relevant cell types (Calderon et al., 2017). We extracted the log-normalized matrix from each processed data and averaged the expression across each identified cell-type classes. We also scaled the expression data, and then took the absolute expression values, so as to form the input of RolyPoly (Calderon et al., 2017). We referred to the Ensembl database (GRCh37) and defined a 10-kb window center around the transcription start site (TSS) of a gene as its transcribed region, to construct a block annotation as recommended that could link the location of GWAS variants with related genes. Of note, we only retained genes on autosomes (Calderon et al., 2017). We used the default parameters and set 1,000 times bootstrap to obtain robust standard errors.

Based on partition heritability, LDSC-cts needs the top upregulated genes list of each cell type rather than the expression data (Finucane et al., 2018). Here, we used Wilcoxon rank sum test embedded in Seurat to find the DEGs for each cell type with all remaining clusters as control. Following Finucane et al. (2018), we extracted the top 10% upregulated genes ranked by P value from each cell type. DEGs were identified as genes expressed in at least 0.1% total cells and with log-transformed fold change above 0 in the target cluster under comparison, so as to ensure a sufficient number of genes could be obtained from each cluster. DEGs lists of each scRNA-seq data used for LDSC-cts analysis are summarized in Supplementary Tables 4,8. We referred to the Ensembl database (GRCh37) and defined the region from the TSS to the transcription end sites (TES) of a gene as its transcribed region (Yates et al., 2019). We also added 100-kb windows on either side of the transcribed region of each gene. Finally, we applied LDSC-cts by jointly modeling the annotation that corresponded to each cell type, a common annotation that included all of the genes, and the 52 annotations in the default “baseline model,” to identify CLD-specific cell types (Finucane et al., 2018).

We also made a sensitivity analysis. Specifically, we changed the resolution used in clustering to obtain a coarser cell type list for analysis. In particular, since LDSC-cts is sensitive to the gene list used for analysis, we simultaneously changed the number of genes included in LDSC-cts to the top 5% upregulated ones.

Bonferroni correction was used for multiple tests (P < 0.1/n, where n = 4 or three is the number of cell type groups, including epithelial cell, non-parenchymal cell, lymphatic immune cell, myeloid immune cell for liver tissue, or gliocyte, neuron, and vascular cell for the brain tissue, Supplementary Table 9) (Hao et al., 2020).

Statistical Software

We used scDblFinder package (version 1.4.0), Seurat package (version 1.4.0), biomaRt package (version 2.45.6), and RolyPoly package (version 0.1.0) in R software (version 3.6.3) (R Core Team, 2020). We used PLINK (version 2.0) (Chang et al., 2015) to analyze GWAS data. We also used LDSC-cts (version 1.0.1) in python software (version 2.7.18) (Van Rossum and De Boer, 1991).

Results

HCC Datasets Analysis

For the HCC GWAS data from BBJ, we totally retained 7,246,543 variants with HWE < 10–6 and MAF > 0.01, as well as their annotation. For the scRNA-seq data (GSE149614), we identified 14 cell types on 30,983 cells (Supplementary Table 2 and Supplementary Figures 1,2). We further excluded cluster with less than 100 cells (63 mast cells) to avoid the interference of their unstable signal on the results. We also excluded the circulating cluster (2,510 cells), since it usually contains various immune cells from the circulation and may represent a mixed signal. Finally, we retained a total of 28,410 cells from 12 cell types. After integrative analysis, we identified B cell (β = 2.956 × 10–4, se = 1.442 × 10–4, P = 0.0228) as cell type associated with HCC in RolyPoly (Figure 3), whereas natural killer cell (NK), monocyte, CD4 + T cell, plasma, macrophage, hepatocyte, regulatory T cell (Treg), endotheliocyte, mesenchymal cell, CD8 + T cell, and dendritic cell (DC) showed no significance (P > 0.05). In LDSC-cts analysis, we also obtained B cell (β = 2.475 × 10–9, se = 1.116 × 10–9, P = 0.0133) as the significant cell type.

FIGURE 3
www.frontiersin.org

Figure 3. Association between HCC scRNA-seq data and HCC GWAS data from RolyPoly and LDSC-cts. Using RolyPoly and LDSC-cts to detect the association of cell types in GSE149614, GSE112271, and their combined HCC scRNA-seq data with EAS HCC GWAS data. Dashed lines in each panel represent a threshold of P = 0.1/4. NK, natural killer cell; Treg, regulatory T cell; DC, dendritic cell; MDM, monocyte-derived macrophage; LEC, lymphatic endothelial cell.

We used another HCC scRNA-seq data from GEO for verification. Totally, we recognized 12 cell types on 30,931 cells from the GSE112271 data with one circulating (1,192 cells) and one small cluster (77 liver sinusoids endothelial cells) excluded (Supplementary Table 2 and Supplementary Figures 3,4). We identified monocyte-derived macrophage (MDM, β = 1.665 × 10–4, se = 6.098 × 10–5, P = 0.0031), T cell (β = 1.732 × 10–4, se = 7.170 × 10–5, P = 0.0076), and natural killer cell (NK, β = 1.458 × 10–4, se = 6.976 × 10–5, P = 0.0191) as cell types significantly associated with HCC in RolyPoly (Figure 3), whereas the obtained NK (β = 2.331 × 10–9, se = 1.118 × 10–9, P = 0.0186) and B cell (β = 2.255 × 10–9, se = 1.134 × 10–9, P = 0.0234) as the significant cell types in LDSC-cts analysis.

We also integrated the two HCC scRNA-seq data and obtained a combined data consisting of 60,120 cells and 13 cell types for further analysis (Supplementary Figures 5,6). The RolyPoly analysis showed that B cell (β = 2.451 × 10–4, se = 9.240 × 10–5, P = 0.0040) was significantly associated with HCC (Figure 3), whereas the LDSC-cts identified no significant cell type.

HCC Dataset Specificity and Sensitivity Analysis

We used scRNA-seq data from other disease to verify the specificity of our findings. To be specific, we downloaded one IPD (GSE157783) scRNA-seq data, and identified 12 cell types on 19,002 cells (Supplementary Table 2 and Supplementary Figures 7,8). After excluding clusters with too few cells (47 fibroblasts and 26 T cells), we identified no cell type significantly associated with HCC in either RolyPoly or LDSC-cts analysis (Figure 4).

FIGURE 4
www.frontiersin.org

Figure 4. Association of IPD scRNA-seq data with HCC GWAS data form RolyPoly and LDSC-cts. Using RolyPoly and LDSC-cts to detect the association of cell types in GSE157783 IPD scRNA-seq data with EAS HCC GWAS data. Dashed lines in each panel represent a threshold of P = 0.1/3. OPC, Oligodendrocyte precursor cell; GABA, GABAergic neurons.

We also made a sensitivity analysis by changing the resolution used in clustering and got nine, eight, and nine cell types for GSE149614, GSE112271, and their integrated data, respectively. Sensitivity analysis showed that B cell was still significantly associated with HCC in RolyPoly analysis on GSE149614 and the integrated data, as well as in LDSC-cts analysis on the integrated data. It also showed nominal significance (P < 0.1) in LDSC-cts analysis on GSE112271, and was the top cell type (P = 0.119) in the analysis on GSE149614 (Supplementary Figure 9).

Cirrhosis Data Analysis

For the cirrhosis GWAS data from BBJ of East Asian population, we totally retained 7,246,475 variants with their annotation. For the scRNA-seq data (GSE136103), we identified 20 cell types on 23,184 cells (Supplementary Table 2; Figure 2; Supplementary Figure 10), but further excluded circulating cluster (309 cells) and clusters with less than 100 cells (56 CLEC9A + dendritic cells and 31 mast cells). Finally, we retained a gene expression data of 17 cell types. RolyPoly showed that CD4 + T cell (β = 2.278 × 10–4, se = 1.149 × 10–4, P = 0.0259) was significantly associated with cirrhosis, whereas LDSC-cts identified no significant cell type (Figure 5).

FIGURE 5
www.frontiersin.org

Figure 5. Association between cirrhosis scRNA-seq data and cirrhosis GWAS data from RolyPoly and LDSC-cts. Using RolyPoly and LDSC-cts to detect the association of cell types in GSE136103 cirrhosis scRNA-seq data with EAS and EUR cirrhosis GWAS data. Dashed lines in each panel represent a threshold of P = 0.1/4. NKT, natural killer T cell; pDC, plasmacytoid dendritic cell; Treg, regulatory T cell; LEC, lymphatic endothelial cell; MDM, monocyte-derived macrophage; NK, natural killer cell; LSEC, liver sinusoids endothelial cell; DC, dendritic cell.

We also used a cirrhosis GWAS summary data of European population from GeneATLAS website to verify the stability of our outcomes, in which a total of 7,636,847 variants was retained after QC. We identified natural killer T cell (NKT, β = 6.535 × 10–10, se = 2.423 × 10–10–1.110 × 10–9, P = 0.0038) and hepatocyte (β = 2.891 × 10–10, se = 1.364 × 10–10, P = 0.0149) as cell types significantly associated with cirrhosis in RolyPoly, while we obtained no significant cell type in the LDSC-cts analysis (Figure 5).

Discussion

Identifying disease-specific cell types has important implications to understand the mechanisms of disease, to guide research, and to develop more precise therapies (Calderon et al., 2017). In this study, using two separate methods and based on available data, we explored the CLD-related cell types through an integrative analysis on GWAS and scRNA-seq data.

In the analysis of HCC, both RolyPoly and LDSC-cts identified B cell as significant associated with HCC (P = 0.0228 and P = 0.0133, respectively). B cell mainly exerts its humoral immunity function through the antibody production and antigen presentation, and can also regulate T cells and innate immune responses (Tsou et al., 2016). Recently, the regulation role of resident B cell in tumor has been investigated (Garaud et al., 2018; Lechner et al., 2019; Wang et al., 2019). The balance between B cells in different states and their activities may have the potential to affect pro- or anti-tumor functions (Largeot et al., 2019; Liu et al., 2019). A similar phenomenon has also been observed in liver disease. In a Hras12V HCC mouse models, B cells were found to have a potential role in suppressing hepatic tumorigenesis (Wang et al., 2017), whereas in another mouse model with inflammation-associated HCC, infiltrating B cells was correlated with increased tumor aggressiveness and mortality (Faggioli et al., 2018). In addition, activated FcγRIIlow/ B cells from HCC tumor may also suppress host anti-tumor immune response via IL-10 signals (Ouyang et al., 2016; Jin et al., 2017). Nevertheless, the depth of research on tumor-associated B cells and their subsets is far less than that of T cells. As for the liver diseases, existing several unbiased scRNAseq research on CLD have not revealed major alterations in the composition or transcriptional profile of liver B cells in disease state (MacParland et al., 2018; Ramachandran et al., 2019; Losic et al., 2020; Sharma et al., 2020). Separate single-cell research has not been conducted specifically on the relationship between B cells and liver disease. However, with the development of single-cell technology, the combination of single-cell transcriptomics and immunomics (B cell receptor) is expected to further reveal the exact role of B cells in HCC and other CLD, and explore B cell-based immunotherapy (Setliff et al., 2019).

We also used another HCC-related scRNA-seq data to verify our findings. RolyPoly identified MDM, T cell, and NK cell, rather than B cell, as significant cell types, whereas B cell remained significant together with NK cell in LDSC-cts analysis. This might have resulted from LDSC-cts using DEGs, which may be conserved but more robust among different studies for a specific disease. Although we have averaged the expression for each identified cell type and taken a scale on the averaged data, differences in data structure arising from the different angles of the two original studies may also be a probable interpretation (Losic et al., 2020). Therefore, we further integrated the two data and repeated these analyses, and found that B cell regained its significance in the integrated data under RolyPoly method. In addition, we used the IPD scRNA-seq data (GSE157783) from brain tissue to make specificity analysis, and found that neither RolyPoly nor LDSC-cts method identified significant cell types. The above results jointly indicated that B cells may be a significant cell type for HCC, and more attention should be paid to them in future research.

Of note, outcomes from the second HCC data also suggested that NK cells might be HCC-related cells, which was significant in both RolyPoly and LDSC-cts analysis. Although this result has not been verified in our analysis, a previous study has identified the contribution of NK cell in liver injury (Luci et al., 2019), NK cell composition alteration and an interaction with other clusters was also observed in HCC (Zhang et al., 2019). Thus, it is also of meaning to further explore the relationship between NK cell and HCC.

As for the analysis on cirrhosis, we have not obtained an overlap cell type in the two methods, with CD4 + T cell significant in RolyPoly analysis using the GWAS data on East Asian population, while NKT and hepatocyte are significant in RolyPoly analysis on European population. That might be caused by the different linkage disequilibrium and minor allele frequency (MAF) for different ancestry, cross-population correlations of causal SNP effects, and heritability (Mather and Thalamuthu, 2020; Wang et al., 2020; Yang and Zhou, 2020). For example, there are 1,558 SNPs and 76 SNPs with P < 10–6 in EAS and EUR datasets, respectively (Supplementary Table 10).

Certainly, several limitations remain in our study. First, all data used came from public databases, and external experiments were not conducted to verify our findings; but alternatively, we used other available GWAS and scRNA-seq data to make verification as well as specificity analysis, which would also ensure the reliability of our results to some extent. Second, SCTransform is a relative powerful normalization method, which may weaken the heterogeneity among samples when used for integration (Butler et al., 2018; Tran et al., 2020). Since we were aimed to apply similar cell type definition strategy in different samples and focused mainly on the similarity rather than heterogeneity, it may offer more help than interference to our analysis. In addition, since current research advances have limited ability in cell type definition and explanation, we applied a relative conservation cell subdivided strategy in the current study. With the in-depth research on various cell subtypes and the development of single-cell technology, similar research is expected be carried out in a larger sample with a higher resolution and precision, and more novel findings with biological explanation would be obtained.

In summary, we performed integrative analysis on GWAS summary data and single scRNA-seq data of CLD, and identified B cell as a potential HCC-related cell type. Since we have made verification from multiple angles, our outcomes are of relative reliability. In addition, as the single-cell atlas of different tissues and diseases has been completed, more targeted researches are expected, and our study would provide valuable clues for further research on CLD.

Code Availability

Code used for data processing, integrated analysis, and plotting could be found at (https://github.com/XiangyuYe/CLD-specific-celltype-identification).

Data Availability Statement

Asian ancestry CLD GWAS summary data was downloaded from BBJ (http://jenger.riken.jp/en/). European ancestry CLD GWAS summary data was downloaded from GeneATLAS website (http://geneatlas.roslin.ed.ac.uk/). ScRNA-seq datasets used (GSE136103, GSE149614, GSE112271, and GSE157783) were downloaded from the GEO database (https://www.ncbi.nlm.nih.gov/geo/).

Author Contributions

RBY and PH designed the study. JLW, YFW, and MLZ performed the datasets quality control. XYY and YW performed the data analysis. PH, HBC, and YFZ interpreted the analysis results. XYY and JLW wrote the draft manuscript. RBY, PH, and MY revised the article. All authors accepted the final manuscript.

Funding

This work was supported by the Natural Science Foundation of Jiangsu Province (Nos. BK20171054 and BK20190106), National Natural Science Foundation of China (No. 81703273), Key Research and Development Program of Zhenjiang, China (Nos. SH2019035 and SH2018058), and People’s Livelihood, Science and Technology Program of Jurong, China (Nos. SF2018453282, SF2019758956, and 2020SA00107).

Conflict of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Acknowledgments

We acknowledge the participants and investigators of Biobank Japan Project and GeneATLAS for making the summary data publicly available for us.

Supplementary Material

The Supplementary Material for this article can be found online at: https://www.frontiersin.org/articles/10.3389/fgene.2021.637322/full#supplementary-material

Footnotes

  1. ^ http://jenger.riken.jp/en/
  2. ^ http://geneatlas.roslin.ed.ac.uk/

References

Aizarani, N., Saviano, A., Sagar, Mailly, L., Durand, S., Herman, J. S., et al. (2019). A human liver cell atlas reveals heterogeneity and epithelial progenitors. Nature 572, 199–204. doi: 10.1038/s41586-019-1373-2

PubMed Abstract | CrossRef Full Text | Google Scholar

Anstee, Q. M., Darlay, R., Cockell, S., Meroni, M., Govaere, O., Tiniakos, D., et al. (2020). Genome-wide association study of non-alcoholic fatty liver and steatohepatitis in a histologically characterised cohort(✩). J. Hepatol. 73, 505–515. doi: 10.1016/j.jhep.2020.04.003

PubMed Abstract | CrossRef Full Text | Google Scholar

Asrani, S. K., Devarbhavi, H., Eaton, J., and Kamath, P. S. (2019). Burden of liver diseases in the world. J. Hepatol. 70, 151–171. doi: 10.1016/j.jhep.2018.09.014

PubMed Abstract | CrossRef Full Text | Google Scholar

Auton, A., Abecasis, G. R., Altshuler, D. M., Durbin, R. M., Abecasis, G. R., Bentley, D. R., et al. (2015). A global reference for human genetic variation. Nature 526, 68–74. doi: 10.1038/nature15393

PubMed Abstract | CrossRef Full Text | Google Scholar

Barrett, T., Wilhite, S. E., Ledoux, P., Evangelista, C., Kim, I. F., Tomashevsky, M., et al. (2012). NCBI GEO: archive for functional genomics data sets—update. Nucleic Acids Res. 41, D991–D995. doi: 10.1093/nar/gks1193

PubMed Abstract | CrossRef Full Text | Google Scholar

Butler, A., Hoffman, P., Smibert, P., Papalexi, E., and Satija, R. (2018). Integrating single-cell transcriptomic data across different conditions, technologies, and species. Nature Biotechnol. 36, 411–420. doi: 10.1038/nbt.4096

PubMed Abstract | CrossRef Full Text | Google Scholar

Calderon, D., Bhaskar, A., Knowles, D. A., Golan, D., Raj, T., Fu, A. Q., et al. (2017). Inferring relevant cell types for complex traits by using single-cell gene expression. Am. J. Hum. Genet. 101, 686–699. doi: 10.1016/j.ajhg.2017.09.009

PubMed Abstract | CrossRef Full Text | Google Scholar

Canela-Xandri, O., Rawlik, K., and Tenesa, A. (2018). An atlas of genetic associations in UK Biobank. Nat. Genet. 50, 1593–1599. doi: 10.1038/s41588-018-0248-z

PubMed Abstract | CrossRef Full Text | Google Scholar

Chang, C. C., Chow, C. C., Tellier, L. C., Vattikuti, S., Purcell, S. M., and Lee, J. J. (2015). Second-generation PLINK: rising to the challenge of larger and richer datasets. GigaScience 4:7. doi: 10.1186/s13742-015-0047-8

PubMed Abstract | CrossRef Full Text | Google Scholar

De Boer, Y. S., Van Gerven, N. M. F., Zwiers, A., Verwer, B. J., Van Hoek, B., Van Erpecum, K. J., et al. (2014). Genome-Wide association study identifies variants associated with autoimmune hepatitis type 1. Gastroenterology 147, 443.e–452.e. doi: 10.1053/j.gastro.2014.04.022

PubMed Abstract | CrossRef Full Text | Google Scholar

Efron, B., and Tibshirani, R. (1986). Bootstrap methods for standard errors, confidence intervals, and other measures of statistical accuracy. Stat. Sci. 1, 54–75. doi: 10.1214/ss/1177013815

CrossRef Full Text | Google Scholar

Faggioli, F., Palagano, E., Di Tommaso, L., Donadon, M., Marrella, V., Recordati, C., et al. (2018). B lymphocytes limit senescence-driven fibrosis resolution and favor hepatocarcinogenesis in mouse liver injury. Hepatology 67, 1970–1985. doi: 10.1002/hep.29636

PubMed Abstract | CrossRef Full Text | Google Scholar

Finucane, H. K., Reshef, Y. A., Anttila, V., Slowikowski, K., Gusev, A., Byrnes, A., et al. (2018). Heritability enrichment of specifically expressed genes identifies disease-relevant tissues and cell types. Nat. Genet. 50, 621–629. doi: 10.1038/s41588-018-0081-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Gadd, V. L., Aleksieva, N., and Forbes, S. J. (2020). Epithelial plasticity during liver injury and regeneration. Cell Stem Cell 27, 557–573. doi: 10.1016/j.stem.2020.08.016

PubMed Abstract | CrossRef Full Text | Google Scholar

Garaud, S., Zayakin, P., Buisseret, L., Rulle, U., Silina, K., De Wind, A., et al. (2018). Antigen specificity and clinical significance of IgG and IgA autoantibodies produced in situ by tumor-infiltrating B cells in breast cancer. Front. Immunol. 9:2660. doi: 10.3389/fimmu.2018.02660

PubMed Abstract | CrossRef Full Text | Google Scholar

Giladi, A., and Amit, I. (2018). Single-Cell genomics: a stepping stone for future immunology discoveries. Cell 172, 14–21. doi: 10.1016/j.cell.2017.11.011

PubMed Abstract | CrossRef Full Text | Google Scholar

Hafemeister, C., and Satija, R. (2019). Normalization and variance stabilization of single-cell RNA-seq data using regularized negative binomial regression. Genome Biol. 20:296. doi: 10.1186/s13059-019-1874-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Hao, X., Wang, K., Dai, C., Ding, Z., Yang, W., Wang, C., et al. (2020). Integrative analysis of scRNA-seq and GWAS data pinpoints periportal hepatocytes as the relevant liver cell types for blood lipids. Hum. Mol. Genet. 29, 3145–3153. doi: 10.1093/hmg/ddaa188

PubMed Abstract | CrossRef Full Text | Google Scholar

Ishigaki, K., Akiyama, M., Kanai, M., Takahashi, A., Kawakami, E., Sugishita, H., et al. (2020). Large-scale genome-wide association study in a Japanese population identifies novel susceptibility loci across different diseases. Nat. Genet. 52, 669–679. doi: 10.1038/s41588-020-0640-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Jin, Y., Lang, C., Tang, J., Geng, J., Song, H. K., Sun, Z., et al. (2017). CXCR5+CD8+ T cells could induce the death of tumor cells in HBV-related hepatocellular carcinoma. Int. Immunopharmacol. 53, 42–48. doi: 10.1016/j.intimp.2017.10.009

PubMed Abstract | CrossRef Full Text | Google Scholar

Largeot, A., Pagano, G., Gonder, S., Moussay, E., and Paggetti, J. (2019). The B-side of cancer immunity: the underrated tune. Cells 8:449. doi: 10.3390/cells8050449

PubMed Abstract | CrossRef Full Text | Google Scholar

Lechner, A., Schlößer, H. A., Thelen, M., Wennhold, K., Rothschild, S. I., Gilles, R., et al. (2019). Tumor-associated B cells and humoral immune response in head and neck squamous cell carcinoma. Oncoimmunology 8, 1535293–1535293. doi: 10.1080/2162402X.2018.1535293

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, Y., Zhai, Y., Song, Q., Zhang, H., Cao, P., Ping, J., et al. (2018). Genome-Wide association study identifies a new locus at 7q21.13 associated with hepatitis B virus–related hepatocellular carcinoma. Clin. Cancer Res. 24, 906–915. doi: 10.1158/1078-0432.CCR-17-2537

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, M., Sun, Q., Wang, J., Wei, F., Yang, L., and Ren, X. (2019). A new perspective: exploring future therapeutic strategies for cancer by understanding the dual role of B lymphocytes in tumor immunity. Int. J. Cancer 144, 2909–2917. doi: 10.1002/ijc.31850

PubMed Abstract | CrossRef Full Text | Google Scholar

Losic, B., Craig, A. J., Villacorta-Martin, C., Martins-Filho, S. N., Akers, N., Chen, X., et al. (2020). Intratumoral heterogeneity and clonal evolution in liver cancer. Nat. Commun. 11:291. doi: 10.1038/s41467-019-14050-z

PubMed Abstract | CrossRef Full Text | Google Scholar

Luci, C., Vieira, E., Perchet, T., Gual, P., and Golub, R. (2019). Natural killer cells and type 1 innate lymphoid cells are new actors in non-alcoholic fatty liver disease. Front. Immunol. 10:1192. doi: 10.3389/fimmu.2019.01192

PubMed Abstract | CrossRef Full Text | Google Scholar

MacParland, S. A., Liu, J. C., Ma, X.-Z., Innes, B. T., Bartczak, A. M., Gage, B. K., et al. (2018). Single cell RNA sequencing of human liver reveals distinct intrahepatic macrophage populations. Nat. Commun. 9:4383. doi: 10.1038/s41467-018-06318-7

PubMed Abstract | CrossRef Full Text | Google Scholar

Marcellin, P., and Kutala, B. K. (2018). Liver diseases: a major, neglected global public health problem requiring urgent actions and large-scale screening. Liver Int. 38(Suppl. 1), 2–6. doi: 10.1111/liv.13682

PubMed Abstract | CrossRef Full Text | Google Scholar

Mather, K. A., and Thalamuthu, A. (2020). Unraveling the genetic contributions to complex traits across different ethnic groups. Nat. Med. 26, 467–469. doi: 10.1038/s41591-020-0834-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Matsuura, K., Sawai, H., Ikeo, K., Ogawa, S., Iio, E., Isogawa, M., et al. (2017). Genome-wide association study identifies TLL1 variant associated with development of hepatocellular carcinoma after eradication of hepatitis C virus infection. Gastroenterology 152, 1383–1394. doi: 10.1053/j.gastro.2017.01.041

PubMed Abstract | CrossRef Full Text | Google Scholar

Nicoletti, P., Aithal, G. P., Bjornsson, E. S., Andrade, R. J., Sawle, A., Arrese, M., et al. (2017). Association of liver injury from specific drugs, or groups of drugs, with polymorphisms in HLA and other genes in a genome-wide association study. Gastroenterology 152, 1078–1089. doi: 10.1053/j.gastro.2016.12.016

PubMed Abstract | CrossRef Full Text | Google Scholar

Ouyang, F. Z., Wu, R. Q., Wei, Y., Liu, R. X., Yang, D., Xiao, X., et al. (2016). Dendritic cell-elicited B-cell activation fosters immune privilege via IL-10 signals in hepatocellular carcinoma. Nat. Commun. 7:13453. doi: 10.1038/ncomms13453

PubMed Abstract | CrossRef Full Text | Google Scholar

R Core Team (2020). R: A Language and Environment for Statistical Computing. Vienna: R Core Team.

Google Scholar

Ramachandran, P., Dobie, R., Wilson-Kanamori, J. R., Dora, E. F., Henderson, B. E. P., Luu, N. T., et al. (2019). Resolving the fibrotic niche of human liver cirrhosis at single-cell level. Nature 575, 512–518. doi: 10.1038/s41586-019-1631-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Ramachandran, P., Matchett, K. P., Dobie, R., Wilson-Kanamori, J. R., and Henderson, N. C. (2020). Single-cell technologies in hepatology: new insights into liver biology and disease pathogenesis. Nat. Rev. Gastroenterol. Hepatol. 17, 457–472. doi: 10.1038/s41575-020-0304-x

PubMed Abstract | CrossRef Full Text | Google Scholar

Saviano, A., Henderson, N. C., and Baumert, T. F. (2020). Single-cell genomics and spatial transcriptomics: discovery of novel cell states and cellular interactions in liver physiology and disease biology. J. Hepatol. 73, 1219–1230. doi: 10.1016/j.jhep.2020.06.004

PubMed Abstract | CrossRef Full Text | Google Scholar

Schwantes-An, T.-H., Darlay, R., Mathurin, P., Masson, S., Liangpunsakul, S., Mueller, S., et al. (2020). Genome-wide association study and meta-analysis on alcohol-related liver cirrhosis identifies novel genetic risk factors. Hepatology [Online ahead of print] doi: 10.1002/hep.31535

PubMed Abstract | CrossRef Full Text | Google Scholar

Setliff, I., Shiakolas, A. R., Pilewski, K. A., Murji, A. A., Mapengo, R. E., Janowska, K., et al. (2019). High-throughput mapping of b cell receptor sequences to antigen specificity. Cell 179, 1636.e–1646.e. doi: 10.1016/j.cell.2019.11.003

PubMed Abstract | CrossRef Full Text | Google Scholar

Sharma, A., Seow, J. J. W., Dutertre, C.-A., Pai, R., Blériot, C., Mishra, A., et al. (2020). Onco-fetal reprogramming of endothelial cells drives immunosuppressive macrophages in hepatocellular carcinoma. Cell 183, 377.e–394.e. doi: 10.1016/j.cell.2020.08.040

PubMed Abstract | CrossRef Full Text | Google Scholar

Stuart, T., Butler, A., Hoffman, P., Hafemeister, C., Papalexi, E., and Mauck, W. M. (2019). Comprehensive integration of single-cell data. Cell 177, 1888.e–1902.e. doi: 10.1016/j.cell.2019.05.031

PubMed Abstract | CrossRef Full Text | Google Scholar

Sudlow, C., Gallacher, J., Allen, N., Beral, V., Burton, P., Danesh, J., et al. (2015). UK biobank: an open access resource for identifying the causes of a wide range of complex diseases of middle and old age. PLoS Med. 12:e1001779. doi: 10.1371/journal.pmed.1001779

PubMed Abstract | CrossRef Full Text | Google Scholar

Tran, H. T. N., Ang, K. S., Chevrier, M., Zhang, X., Lee, N. Y. S., Goh, M., et al. (2020). A benchmark of batch-effect correction methods for single-cell RNA sequencing data. Genome biol. 21, 12–12. doi: 10.1186/s13059-019-1850-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Tsou, P., Katayama, H., Ostrin, E. J., and Hanash, S. M. (2016). The emerging role of B cells in tumor immunity. Cancer Res. 76, 5597–5601. doi: 10.1158/0008-5472.CAN-16-0431

PubMed Abstract | CrossRef Full Text | Google Scholar

Van Rossum, G., and De Boer, J. (1991). Interactively testing remote servers using the Python programming language. CWI Q. 4, 283–304.

Google Scholar

Wang, K., Nie, X., Rong, Z., Fan, T., Li, J., Wang, X., et al. (2017). B lymphocytes repress hepatic tumorigenesis but not development in Hras12V transgenic mice. Int. J. Cancer 141, 1201–1214. doi: 10.1002/ijc.30823

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, S.-S., Liu, W., Ly, D., Xu, H., Qu, L., and Zhang, L. (2019). Tumor-infiltrating B cells: their role and application in anti-tumor immunity in lung cancer. Cell. Mol. Immunol. 16, 6–18. doi: 10.1038/s41423-018-0027-x

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, Y., Guo, J., Ni, G., Yang, J., Visscher, P. M., and Yengo, L. (2020). Theoretical and empirical quantification of the accuracy of polygenic scores in ancestry divergent populations. Nat. Commun. 11:3865. doi: 10.1038/s41467-020-17719-y

PubMed Abstract | CrossRef Full Text | Google Scholar

Yang, S., and Zhou, X. (2020). Accurate and scalable construction of polygenic scores in large biobank data sets. Am. J. Hum. Genet. 106, 679–693. doi: 10.1016/j.ajhg.2020.03.013

PubMed Abstract | CrossRef Full Text | Google Scholar

Yates, A. D., Achuthan, P., Akanni, W., Allen, J., Allen, J., Alvarez-Jarreta, J., et al. (2019). Ensembl 2020. Nucleic Acids Res. 48, D682–D688. doi: 10.1093/nar/gkz966

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, Q., He, Y., Luo, N., Patel, S. J., Han, Y., Gao, R., et al. (2019). Landscape and dynamics of single immune cells in hepatocellular carcinoma. Cell 179, 829.e–845.e. doi: 10.1016/j.cell.2019.10.003

PubMed Abstract | CrossRef Full Text | Google Scholar

Zheng, C., Zheng, L., Yoo, J.-K., Guo, H., Zhang, Y., Guo, X., et al. (2017). Landscape of infiltrating T cells in liver cancer revealed by single-cell sequencing. Cell 169, 1342.e–1356.e. doi: 10.1016/j.cell.2017.05.035

PubMed Abstract | CrossRef Full Text | Google Scholar

Zheng, G. X. Y., Terry, J. M., Belgrader, P., Ryvkin, P., Bent, Z. W., Wilson, R., et al. (2017). Massively parallel digital transcriptional profiling of single cells. Nature Communi. 8, 14049. doi: 10.1038/ncomms14049

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: chronic liver diseases, GWAS, scRNA-seq, integrated analysis, cell type

Citation: Ye X, Wei J, Yue M, Wang Y, Chen H, Zhang Y, Wang Y, Zhang M, Huang P and Yu R (2021) Leveraging Single-Cell RNA-seq Data to Uncover the Association Between Cell Type and Chronic Liver Diseases. Front. Genet. 12:637322. doi: 10.3389/fgene.2021.637322

Received: 03 December 2020; Accepted: 25 January 2021;
Published: 08 March 2021.

Edited by:

Shiquan Sun, Xi’an Jiaotong University, China

Reviewed by:

Xingjie Hao, Huazhong University of Science and Technology, China
Zhongshang Yuan, Shandong University, China

Copyright © 2021 Ye, Wei, Yue, Wang, Chen, Zhang, Wang, Zhang, Huang and Yu. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Peng Huang, huangpeng@njmu.edu.cn; Rongbin Yu, rongbinyu@njmu.edu.cn

These authors have contributed equally to this work and share first authorship

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.