Abstract
The human liver is an essential multifunctional organ, and liver diseases are rising with limited treatment options. However, the cellular composition of the liver remains poorly understood. Here, we performed single-cell RNA-sequencing of ~10,000 cells from normal liver tissue of 9 human donors to construct a human liver cell atlas. Our analysis revealed previously unknown sub-types among endothelial cells, Kupffer cells, and hepatocytes with transcriptome-wide zonation of some of these populations. We reveal heterogeneity of the EPCAM+ population, which comprises hepatocyte-biased and cholangiocyte populations as well as a TROP2int progenitor population with strong potential to form bipotent liver organoids. As proof-of-principle, we utilized our atlas to unravel phenotypic changes in hepatocellular carcinoma cells and in human hepatocytes and liver endothelial cells engrafted into a mouse liver. Our human liver cell atlas provides a powerful resource enabling the discovery of previously unknown cell types in the normal and diseased liver.
The liver is a key organ in the human body. It serves as a central metabolic coordinator with a wide array of essential functions, including regulation of glucose and lipid metabolism, protein synthesis, and bile synthesis. Furthermore, the liver is a visceral organ that is capable of remarkable natural regeneration after tissue loss(1). However, prevalence and mortality of liver disease have risen dramatically within the last decades(2). The liver cellular landscape has barely been explored at single-cell resolution, which limits our molecular understanding of liver function and disease biology. The recent emergence of sensitive single-cell RNA-sequencing (scRNA-seq) methods(3) enables the investigation of cell types in health and disease.
To characterize the human liver at single-cell resolution, we developed a robust pipeline for scRNA-seq of cryopreserved and freshly isolated patient-derived human liver samples and assembled an atlas consisting of 10,372 cells from nine donors. We performed in-depth analysis of all liver cell types with a major focus on epithelial liver cell progenitors.
Single-Cell RNA-seq of the human liver
We applied mCEL-Seq2 (4) for scRNA-seq of non-diseased liver tissue from six patients who underwent liver resections for colorectal cancer metastasis or cholangiocarcinoma without history of chronic liver disease (Fig. 1a, Methods). We sorted and sequenced viable cells in an unbiased fashion as well as specific cell populations on the basis of cell surface markers (Extended Data Fig. 1, Methods). Since fresh liver tissue material is scarce and difficult to preserve, and biobanks with cryopreserved liver samples represent rich resources, we generated scRNA-seq data from cryopreserved cells in addition to single-cell suspensions from freshly prepared liver resection specimens (Methods). We then used RaceID3 for the identification of cell types (Methods) (4),(5).
Cells from different patients, isolated from freshly prepared or cryopreserved single-cell suspensions co-clustered (Extended Data Fig. 1). Furthermore, fresh and cryopreserved cells from the same patient did not reveal pronounced differential gene signatures (Extended Data Fig. 1e-h). However, we observed compositional differences both between fresh and cryopreserved samples derived from the same patient as well as across different fresh (or cryopreserved) samples. We attribute these differences to variability in cell viability and cell type composition across samples.
Since scRNA-seq of randomly sampled populations yielded almost exclusively hepatocytes and immune cells (Extended Data Fig. 1i), we applied additional sorting strategies to enrich for endothelial cells (Extended Data Fig. 1a-c) and EPCAM+ cells (see below).
Based on the expression of marker genes, our atlas comprises all the main liver cell types, including hepatocytes, EPCAM+ bile duct cells (cholangiocytes), CLEC4G+ liver sinusoidal endothelial cells (LSECs), CD34+PECAMhigh macro-vascular endothelial cells (MaVECs), hepatic stellate cells and myofibroblasts, Kupffer cells, and immune cells (Fig. 1b-d and Supplementary Data Table 1). To facilitate interactive exploration of our human liver cell atlas, we created a web interface: http://human-liver-cell-atlas.ie-freiburg.mpg.de/.
Zonation of human liver cell types
Hepatocytes are spatially heterogeneous and zonated along the portal-central axis of the liver lobule(6–8). Based on their metabolic sub-specialization, the liver lobule has been divided into the periportal zone surrounding the portal triad (portal vein, hepatic artery and bile duct), the central zone nearest to the central vein, and the remaining mid zone(6–8). While previous observations suggested sub-specialization of non-parenchymal cells like LSECs and Kupffer cells6, heterogeneity of these cell types has remained elusive, and most studies were carried out in rodents.
We were able to directly compare the signature of MaVECs and LSECs and identified several previously unknown sub-populations (see Extended Data Fig. 2 and Supplementary Note 1).
ScRNA-seq is highly informative on hepatocyte zonation in mouse(9) and a first single-cell analysis of human hepatocyte and endothelial cell zonation at limited resolution was done recently(10). To infer continuous transcriptome-wide zonation, we reasoned that the major axis of variability for a cell type could reflect gene expression changes associated with zonation. Hence, we ordered LSECs and hepatocytes by diffusion pseudo-time (dpt)(11), here interpreted as pseudo-space, along this axis and applied self-organizing maps (SOMs) to infer co-expression modules (Fig. 2, Methods).
We first validated our strategy by recovering previously characterized zonation of mouse hepatocytes(9) (Extended Data Fig. 3a-d). For our human hepatocytes, this approach recovered zonated expression patterns of landmark genes, e.g. ALB and PCK1 (periportal module 1), CYP1A2 and CYP2E1 (central/midzonal module 34 and 24, respectively), and GLUL (central module 33)(7,9) (Fig. 2a, Extended Data Fig. 3e-g, Supplementary Data Table 2,3). In total, 1,384 out of 3,395 expressed genes (41%) included in the hepatocyte analysis exhibited significant zonation (Benjamini-Hochberg corrected ANOVA P<0.01). Pathway enrichment analysis revealed that periportal hepatocyte modules are enriched in genes involved in biological oxidations, consistent with an oxygen gradient peaking in the periportal zone(6–8), and in the glycogen synthesis pathway. (Extended Data Fig. 3h). In accordance with its zonation in mouse hepatocytes, the urea cycle enzyme CPS1 peaks in periportal hepatocytes (Extended Data Fig. 3g). Midzonal hepatocyte modules are enriched in, e.g., metabolism of xenobiotics by Cytochromes P450. Immunostainings for selected genes validate the predicted zonation on the protein level (Fig. 2a).
LYVE1 and CD14 were previously identified as markers distinguishing midzonal and central LSECs periportal populations(12). Analyzing LSEC zonation, we found that 806 out of 1,198 expressed genes (67%) exhibited significant zonation (Benjamini-Hochberg corrected ANOVA P<0.01) (Fig. 2b, Extended Data Fig. 3i, Supplementary Data Table 4, 5). Central and midzonal endothelial cells (modules 1 and 3) exhibited peaked expression of LYVE1 and FCN3, a ficolin protein which can activate the lectin pathway of complement activation. Interestingly, pathway enrichment analysis of the central and midzonal endothelial modules recovered pathways, like binding and uptake of ligands by scavenger receptors, that are shared with midzonal hepatocytes (Extended Data Fig. 3j). Together with a more detailed gene expression analysis (see Supplementary Note 2) this observation suggests the concept of co-zonated genes and functions across hepatocytes and endothelial cells.
Finally, a comparison between mouse(9,13) and human revealed only limited evolutionary conservation of gene expression zonation (see Supplementary Note 3 and (Supplementary Data Tables 6) and (7), reflecting widespread evolutionary changes.
Human liver immune cell populations
A detailed analysis of the CD163+VSIG4+ Kupffer cell compartment revealed sub-populations with distinct gene expression signatures (see Supplementary Note 4 and Extended Data Fig. 4), in agreement with a recent study10. Moreover, we detected shared gene expression and pathways between Kupffer cell subsets and endothelial cells (see Supplementary Note 4 and Extended Data Fig. 4), providing further evidence for functional co-operation of different cell types.
We identified an MS4A1+CD37+ B cell subset, which corresponds to circulating B cells upregulating MHC class II components, and a liver resident MZB1+ B cell subset expressing DERL3, SSR4, and IGHG4 (Extended Data Fig. 5).
Finally, we recovered a population of CD56+ NK cells (cluster 5) and CD56- (cluster 3) as well as CD56+ (cluster 1) CD8A+ NKT cells, expressing different combinations of chemokine ligands, granzymes, and killer cell lectin-like receptor genes (Extended Data Fig. 6). Clusters 12 and 18 up-regulate a number of heat shock genes. These observations demonstrate an unexpected variety of immune cell subtypes in the human liver.
Putative bipotent epithelial progenitors
Liver regeneration after tissue damage involves the replication of several liver cell types including hepatocytes and cholangiocytes. Furthermore, different types of liver damage lead to specific mechanisms of liver regeneration(14,15). However, the existence of a naïve adult stem cell population in the human liver and its contribution to turnover and regeneration remains controversial. Rare EPCAM+ cells have been termed hepatic stem cells(16), which can form dense round colonies when cultured and are bipotent progenitors of hepatoblasts, which differentiate into cholangiocytes or hepatocytes both in vitro and in vivo(16,17).
In search for genuine liver progenitor cells, we sorted and sequenced single EPCAM+ cells from adult human livers. We discovered novel biliary and potential liver progenitor cell surface markers correlating with EPCAM or TROP1 expression; these include TACSTD2 (TROP2), FGFR2, TM4SF4, and CLDN1 and immunohistochemistry confirmed predicted novel markers such as ANXA4 and the transcriptional co-activator WWTR1 (Extended Data Fig. 7a).
A focused analysis revealed that the EPCAM+ compartment is transcriptionally heterogeneous and consists of an ASGR1+ hepatocyte-biased population, KRT19highCFTRhighALBlow cholangiocyte populations, and a remaining population of putative naïve progenitor cells (Fig. 3a, Extended Data Fig. 7b,c). The EPCAM+ population exhibits only stochastic expression of proliferation markers MKI67 and PCNA and is negative for the hepatoblast marker AFP (Extended Data Fig. 7d). Hence, the transcriptional heterogeneity of this population is unlikely to arise as a result of proliferation, and the observed sub-types reside in the normal human liver.
To explore the relatedness of these sub-populations, we reanalyzed the EPCAM+ population with RaceID3 and employed StemID2 for lineage reconstruction(4,18) (Fig. 3b, Methods). This analysis revealed that the population in the center of the t-SNE map (clusters 1,2,5,6,7) bifurcates into hepatocyte progenitors and cholangiocytes. To provide further evidence for continuous differentiation trajectories connecting naïve EPCAM+ progenitors to cholangiocytes and mature hepatocytes, we performed StemID2 and diffusion map analyses on the combined population of mature hepatocytes and EPCAM+ cells (Extended Data Fig. 8a-c).
To better understand the emergence of fate bias towards the two lineages, we applied FateID for the inference of lineage probabilities in each cell(4). Consistently, FateID infers similar probabilities of the central population to differentiate towards hepatocytes and cholangiocytes (Fig. 3c). The fate bias predictions are supported by a differential gene expression analysis revealing up-regulation of common genes comprising several signaling pathway components (HES1, SFRP5, FGFR2, FGFR3) in the central population (Fig. 3d), and gradual up-regulation of distinct gene sets towards the hepatocyte-biased and cholangiocyte populations, respectively (Extended Data Fig. 8e). The expression of TACTSD2 (TROP2) anti-correlated with hepatocyte fate bias, exhibiting a gradient that ranges from high expression in mature cholangiocytes to very low expression in the hepatocyte-biased population (Fig. 3e and Extended Data Fig. 7c). Immunostaining of TROP2 in normal human liver tissue showed specific expression in cells of the bile ducts and bile ductules (Fig. 3f). Interestingly, TROP2 expression was reported in amplifying oval cells of injured mouse livers(19).
The central TROP2int population is in itself heterogeneous and contains a MUC6high population (cluster 7). MUC6 is highly expressed by pancreatic progenitors and multi-potent bile duct tree stem cells(20), which have been proposed to be the origin of the EPCAM+ hepatic stem cells. The TROP2high cholangiocyte clusters comprise a CXCL8+ population (cluster 8) and an MMP7+ population (clusters 4 and 13) (Extended Data Fig. 7c and Extended Data Fig. 8e,f), while TROP2low clusters up-regulate hepatocyte markers such as ALB, HP, HNF4A and ASGR1 (Extended Data Fig. 7c and 8e,f).
The central TROP2int population stratified as bipotent based on the FateID-predicted bias expresses early developmental transcription factors such as HES1, which is essential for tubular bile duct formation(21), and PROX1, an early specification marker for the developing liver in the mammalian foregut endoderm which is required for hepatocyte proliferation and migration during development(22) (Fig. 3d). Furthermore, we find lower expression of hepatocytes genes such as HNF4A, HP and ALB and of cholangiocyte markers like KRT19 and CFTR in the population stratified as bipotent compared to the hepatocyte-biased and the mature cholangiocyte populations, respectively (Fig. 3d) and (Extended Data Fig. 7c) and (8f). We speculate that we enriched for the TROP2int KRT19low/- immature population during cell isolation, since mature bile duct cells require a harsher digestion for their isolation, which can negatively affect other liver cell types. Thus, the actual fraction of KRT19high cells in the tissue is presumably higher. We validated the existence of EPCAM+KRT19low/- cells in addition to EPCAM+KRT19high/+ cells in situ by immunofluorescence (Fig. 3g and Extended Data Fig. 7e).
Consistent with our scRNA-seq data, flow cytometry profiles of EPCAM and TROP2 displayed a gradient of TROP2 expression in EPCAM+ cells, and EPCAM expression correlated with TROP2 expression (Fig. 4a). Moreover, forward and side-scatter profiles of EPCAM+ cells indicate that the compartment is heterogeneous and consists of populations with different sizes and morphologies (Fig. 4a). Based on the TROP2 expression distribution, we compartmentalized EPCAM+ cells into three compartments: TROP2low/-, TROP2int, and TROP2high (Fig. 4a). To validate that the TROP2int population harbors the progenitor population, we attempted to culture bipotent organoids(23) from each compartment. In agreement with our prediction, TROP2int cells exhibited the highest organoid forming capacity, while TROP2low/- cells did not form organoids, and TROP2high cells gave rise to much smaller organoids at strongly reduced frequency compared to TROP2int cells (Fig. 4b). Single-cell culture of TROP2int cells demonstrated the organoid-forming capacity of individual cells from this gate, providing evidence for bipotency on the clonal level (Fig. 4c). ScRNA-seq of the input populations for organoid culture from each compartment expectedly showed a marked enrichment of the respective compartment in the original EPCAM+ data (Fig. 4d,e and Extended Fig. 8g,h). Strikingly, flow cytometry profiles of EPCAM and TROP2 for organoid cells grown from the TROP2int compartment recover TROP2low/-, TROP2int and TROP2high populations in the organoids (Fig. 4f).
To elucidate the cell type composition of the organoids in depth, we performed scRNA-seq. Co-analysis of the organoid cells and the EPCAM+ cells sequenced directly from the patients demonstrates marked transcriptome differences (Fig. 4e). While EPCAM and CD24 were expressed in cells from the organoids and patients, organoid cells downregulated various genes such as AQP1 or the WNT signaling modulator SFRP5 and upregulated others such as the proliferation marker MKI67+, reflected by differential enrichment of the corresponding pathways (Fig. 4g and Extended Data Fig. 8i-k). We observed several sub-populations within the organoids, including a non-dividing hepatocyte-biased SERPINA1high population and a non-dividing KRT19high cholangiocyte-biased population, consistent with the signature of the EPCAM+ cells recovered from the patients (Fig. 4e). This further supports the claim that the TROP2int compartment harbors a bipotent progenitor population, which can give rise to hepatocyte and cholangiocyte populations.
In contrast to patients’ cells, organoid cells strongly downregulate ALB but express AGR2 and other mucin family genes like MUC5AC and MUC5B, normally expressed, e.g., in intestinal cells and gastrointestinal cancers(24,25)(Fig. 4g and Extended Data Fig. 8j). These observations reflect that organoid cells express genes that are expressed in other systems, acquire a more proliferative state, and appear to upregulate stem cell-related pathways like WNT signaling.
In light of (1) these functional validation experiments, (2) the observed gene signature of the TROP2int cells, and (3) the in situ location, our data strongly suggest that the putative liver progenitor population can be defined as a subpopulation of bile duct cells.
Perturbed cell states in liver cancer
Hepatocellular carcinoma is the most common type of primary liver cancer(26). In order to demonstrate the value of our atlas as a reference for comparisons with diseased liver cells, we sequenced CD45+ and CD45- cells from HCC tissue of three different patients (Extended Data Fig. 9a,b, Methods).
We recovered several cell types from the tumors, including cancer cells, endothelial cells, Kupffer cells, NKT and NK cells (Fig. 5a and Extended Data Fig. 9c) and compared them to the normal liver cell atlas. Differential gene expression analysis and immunohistochemistry revealed that cancer cells lose the expression of cytochrome P450 genes like CYP2E1 and CYP2C8 and the periportally zonated gene CPS1 (Fig. 5b and Extended Data Fig. 9d,e) as well as the metabolic signature of normal hepatocytes (Fig. 5e). They up-regulate AKR1B10, a known biomarker of HCC with potential involvement in hepatocellular carcinogenesis(27) (Extended Data Fig. 9d). Moreover, immunohistochemistry confirmed that IL32, a pro-inflammatory TNF-alpha inducing cytokine, is highly upregulated in cancer cells (Fig. 5b). Overall, cancer cells upregulate WNT and Hedgehog signaling pathways, highlighting similarities between EPCAM+ normal liver progenitors and the observed cancer cell population (Fig. 5c).
Endothelial cells from the tumor upregulate, e.g., extracellular matrix organization genes such as COL4A2 and SPARC (Fig. 5d, Extended Data Fig. 9f). Strikingly, they do not express LSEC markers like CLEC4G but upregulate MaVEC markers like PECAM1, AQP1 and CD34 (Fig. 5e and Extended Data Fig. 9f,g). Moreover, HCC LSECs upregulate PLVAP which makes them less permeable and could potentially restrict the access of lymphocytes and soluble antigens(28) to the tumor (see Supplementary Note 5 and Extended Data Fig. 9f,g).
We conclude that the comparison of scRNA-seq data between the cell populations of HCC and the liver cell atlas allows the inference of perturbed gene expression signatures, biomarkers and modulated functions across cell types.
A human liver chimeric mouse model
Patient derived xenograft mouse liver models are a powerful tool for studying human liver cells and diseases in vivo(29). To correctly interpret such experiments, it is crucial to understand the differences between cells directly taken from the human liver and transplanted human cells in the mouse chimeric liver.
To address this issue, we transplanted human liver cells from patient-derived hepatocyte and non-parenchymal cell fractions into an FRG-NOD (Fah-/-Rag2-/-Il2rg-/-) mouse liver model(30) (HMouse) and sorted single human cells in an unbiased fashion and on the basis of hepatocyte and endothelial cell markers post engraftment for scRNA-seq (Fig. 6a and Extended Data Fig. 10a). We then compared these engrafted cells to our reference atlas and observed that we had successfully transplanted both human hepatocytes and endothelial cells (Fig. 6b and Extended Data Fig. 10b,c), which maintained their fundamental gene signatures, such as ALB or PCK1 and CLEC4G, PECAM1 or CD34, respectively (Extended Data Fig. 10b-f). Nevertheless, many genes were differentially expressed compared to human liver cells, e.g. AKR1B10, which was also expressed by cancer cells from HCC (Fig. 6c and Extended Data Fig. 10g). Gene Set Enrichment Analysis (GSEA) of differentially expressed genes revealed that HMouse hepatocytes and endothelial cells downregulated pathways, such as hemostasis, and upregulated WNT and Hedgehog signaling as well as cell cycle genes (Fig. 6d) akin to what we observed in the HCC cancer cells and cells from the liver organoids.
Discussion
We here established a human liver cell atlas, revealing heterogeneity within major liver cell populations and the existence of an epithelial progenitor in the adult human liver.
Our atlas reveals transcriptome-wide zonation of hepatocytes and endothelial cells, and suggests that different liver cell types may cooperate in order to carry out essential functions. Although we could validate predicted zonation profiles with antibody staining, it will be essential to perform more large scale in situ gene expression analysis. The novel EPCAM+TROP2int population provides a strong candidate with potential involvement in homeostatic turnover, liver regeneration, disease pathogenesis, and tumor formation. We point out that, while our in silico analysis and the in vitro organoid culture experiments provide evidence for bipotency of this population, its lineage potential remains to be demonstrated in vivo.
As demonstrated by our HCC analysis, the atlas provides a key reference to investigate liver diseases and will contribute to advance urgently needed human liver models, including organoids as well as humanized liver chimeric mouse models.
Methods
Human liver samples
Human liver tissue samples were obtained from patients who had undergone liver resections between 2015 and 2018 at the Center for Digestive and Liver Disease (Pôle Hépato-digestif) of the Strasbourg University Hospitals University of Strasbourg, France. For the human liver cell atlas, samples were acquired from patients without chronic liver disease (defined as liver damage lasting over a period of at least six months), genetic hemochromatosis with homozygote C282Y mutation, active alcohol consumption (> 20 g/d in women and > 30 g/d in men), active infectious disease, pregnancy or any contraindication for liver resection. All patients provided a written informed consent. The protocols followed the ethical principles of the declaration of Helsinki and were approved by the local Ethics Committee of the University of Strasbourg Hospitals and by the French Ministry of Education and Research (Ministère de l'Education Nationale, de l’Enseignement Supérieur et de la Recherche; approval number DC-2016-2616). Data protection was performed according to EU legislation regarding privacy and confidentiality during personal data collection and processing (Directive 95/46/EC of the European Parliament and of the Council of the 24 October 1995). Samples (BP1) and tissue blocks were obtained from Biopredic International.
Tissue dissociation and preparation of single cell suspensions
Human liver specimens obtained from resections were perfused for 15 minutes with calcium-free 4-(2-hydroxyethyl)-1-piperazine ethanesulfonic acid buffer containing 0.5 mM ethylene glycol tetraacetic acid (Fluka) followed by perfusion with 4-(2-hydroxyethyl)-1-piperazine ethanesulfonic acid containing 0.5 mg/mL collagenase (Sigma-Aldrich) and 0.075% CaCl2 at 37°C for 15 min as previously described(32). Then the cells were washed with phosphate-buffered saline (PBS) and nonviable cells were removed by Percoll (Sigma-Aldrich) gradient centrifugation. Part of the isolated cells was further separated into primary human hepatocytes (PHH) and non-parenchymal cells (NPCs) by an additional centrifugation step. The isolated cells were frozen in liquid nitrogen using the CryoStor® CS10 solution (Sigma-Aldrich).
Transplantation of human cells into Fah-/-/Rag2-/-/Il2rg -/- mice
Fah-/-/Rag2-/-/Il2rg -/- (FRG) breeding mice were kept at the Inserm Unit 1110 SPF animal facility and maintained with 16 mg/L of 2-(2-nitro-4-trifluoro-methyl-benzoyl)-1,3 cyclohexanedione (NTBC; Swedish Orphan Biovitrum) in drinking water. Six-week old mice were intravenously injected with 1.5 x 109 pfu of an adenoviral vector encoding the secreted form of the human urokinase-like plasminogen activator (Ad-uPA)(33). Forty-eight hours later, 106 PHH and 2 x 105 NPCs from the same liver donor and isolated as previously described, were injected intra-splenically via a 27-gauge needle. For the procedure, the mice were kept under gaseous isoflurane anesthesia and received a subcutaneous injection of buprenorphine at the dose of 0.1 mg/kg. After the transplantation the NTBC was gradually decreased and completely withdrawn in 7 days. The success of the transplantation was evaluated 2 months after the procedure by dosing human albumin in mouse serum as previously described(34). All procedures are consistent with the guidelines set by the Panel on Euthanasia (AVMA) and the NIH Guide for the Care and Use of Laboratory Animals as well as the Declaration of Helsinki in its latest version, and to the Convention of the Council of Europe on Human Rights and Biomedicine. The animal research was performed within the regulations and conventions protecting the animals used for research purposes (Directive 86/609/EEC), as well as with European and national laws regarding work with genetically modified organs. The animal facility at the University of Strasbourg, Inserm U1110 has been approved by the regional government (Préfecture) and granted the authorization number N° D67-482-7, 2012/08/22.
FACS
Liver cells were sorted from mixed, hepatocyte, and non-parenchymal cell fractions on an Aria Fusion I using a 100 μm nozzle. Cells from the HCC samples were not fractionated and were sorted directly after tissue digestion. Zombie Green (Biolegend) was used as a viability dye. Cells were stained with human specific antibodies against CD45 (Biolegend), PECAM1 (Biolegend), CD34 (Biolegend), CLEC4G (R&D systems), ASGR1 (BD Biosciences), EPCAM (R&D systems), and TROP2 (Biolegend). Organoids were stained with antibodies against EPCAM and TROP2. For the humanized mouse samples, cells were stained either with antibodies against ASGR1 and PECAM1 or human HLA-ABC (BD Biosciences) and mouse H2-Kb (BD Biosciences). Viable cells were sorted in an unbiased fashion or from specific populations based on the expression of markers into the wells of 384 well plates containing lysis buffer.
Single-cell RNA amplification and library preparation
Single cell RNA sequencing was performed according to the mCEL-Seq2 protocol(4,35). Viable liver cells were sorted into 384-well plates containing 240 nL of primer mix and 1.2 μL of PCR encapsulation barrier, Vapor-Lock (QIAGEN) or mineral oil (Sigma-Aldrich). Sorted plates were centrifuged at 2200 g for a few minutes at 4°C, snap-frozen in liquid nitrogen and stored at −80°C until processed. 160nL of reverse transcription reaction mix and 2.2 μL of second strand reaction mix were used to convert RNA into cDNA. cDNA from 96 cells were pooled together before clean up and in vitro transcription, generating 4 libraries from one 384-well plate. 0.8 μL of AMPure/RNAClean XP beads (Beckman Coulter) per 1 μL of sample were used during all the purification steps including the library cleanup. Other steps were performed as described in the protocol. Libraries were sequenced on an Illumina HiSeq 2500 and 3000 sequencing system (pair-end multiplexing run, high output mode) at a depth of ~150,000-200,000 reads per cell.
Quantification of Transcript Abundance
Paired end reads were aligned to the transcriptome using bwa (version 0.6.2-r126) with default parameters(36). The transcriptome contained all gene models based on the human whole genome ENCODE V24 release. All isoforms of the same gene were merged to a single gene locus. The right mate of each read pair was mapped to the ensemble of all gene loci and to the set of 92 ERCC spike-ins in the sense direction. Reads mapping to multiple loci were discarded. The left read contains the barcode information: the first six bases corresponded to the unique molecular identifier (UMI) followed by six bases representing the cell specific barcode. The remainder of the left read contains a polyT stretch. The left read was not used for quantification. For each cell barcode, the number of UMIs per transcript was counted and aggregated across all transcripts derived from the same gene locus. Based on binomial statistics, the number of observed UMIs was converted into transcript counts(37).
Single-Cell RNA Sequencing Data Analysis
10,372 cells passed quality control threshold of >1,000 transcripts (Poisson-corrected UMIs37) for the normal human liver cell atlas. For cells from the organoids, 1052 cells passed the quality control thresholds. For cells from HCC, 1282 cells passed the quality control threshold of >1,000 transcripts. For cells from the humanized mouse, 311 cells passed the quality control threshold of >1,000 transcripts. All the datasets were analyzed using RaceID3(4). For normalization, the total transcript counts in each cell were normalized to 1 and multiplied by the minimum total transcript count across all cells passing the quality control threshold (> 1,000 transcripts per cell). Prior to normalization, cells expressing >2% of KCNQ1OT1 transcripts, a previously identified marker of low quality cells(18) were removed from the analysis. Moreover, transcripts correlating to KCNQ1OT1 with a Pearson’s correlation coefficient >0.4 were also removed. RaceID3 was run with the following parameters: mintotal=1000, minexpr=2, minnumber=10, outminc=2, cln=15 and default parameters otherwise. The t-distributed stochastic neighbor embedding (t-SNE) algorithm was used for dimensional reduction and cell cluster visualization.
Diffusion Pseudo-Time Analysis and Self-Organizing Maps
Diffusion pseudotime (dpt) analysis(11) was implemented and diffusion maps generated using the destiny R package. The number of nearest neighbors, k, was set to 100. Self-organizing maps (SOMs) were generated using the FateID package based on the ordering computed by dpt as input. Only genes with >2 counts after size normalization in at least a single cell were included for the SOM analysis. Briefly, smooth zonation profiles were derived by applying local regression on normalized transcript counts after ordering cells by dpt. Next, a one-dimensional SOM with 200 nodes was computed on these profiles after z-transformation. Neighboring nodes were merged if the Pearson’s correlation coefficient of the average profiles of these nodes exceed 0.85. The remaining aggregated nodes represent the gene modules shown in the SOM figures.
P-values for the significance of zonation were derived by binning dpt-ordered profiles into three equally-sized bins to perform ANOVA. The resulting p-values were multiple-testing corrected with the Benjamini-Hochberg method. Increasing the number of bins produced similar results.
Conservation of zonation between human and mouse
Expression data from Halpern et al.(9) (GEO accession code GSE84498) were used for analyzing evolutionary conservation of hepatocyte zonation between human and mouse. The transcript count data were analyzed with RaceID3 to determine cell types, with parameter mintotal=1,000 and cln=6. A subgroup of clusters was identified as hepatocytes based on marker gene expression and used for pseudo-time (dpt) and sub-sequent SOM analysis as was done for the human data. To obtain a similar number of genes, only genes with at least 1.5 counts after size normalization in at least a single cell were included. To identify orthologs between human and mouse for the references used in this study and by Halpern et al. as provided by the authors, we first identified pairs of orthologs based on identical gene identifiers upon capitalization of all letters. We further computed mutual blastn (run with default) best hits. The final list comprises 16,670 pairs of orthologs.
Conservation of zonation was assessed based on the Pearson’s correlation of zonated expression profiles after binning the human data into nine equally-sized bins akin to the nine zones derived in Halpern et al. Conservation of zonation of endothelial cells was evaluated based on published mouse data from Halpern et al.(13) utilizing the classification into four spatially stratified population. To calculate Pearson’s correlation coefficient between human and mouse endothelial cells, a dpt analysis was performed for all human cells mapping to endothelial cell clusters and these profiles were discretized into four equally-sized bins.
Lineage Analysis of the EPCAM+ compartment
For a separate analysis of the EPCAM+ population, all cells from clusters 4, 7, 24 and 39 were extracted and reanalyzed with RaceID3(4) using the parameters mintotal=1000 and minexpr=2, minnumber=10 outminc=2, and default parameters otherwise. StemID(24) was run on these clusters with cthr=10 and nmode=TRUE and knn=3. FateID(4) was run on the filtered and feature-selected expression matrix from RaceID3, with target clusters inferred by FateID using ASGR1 plus ALB and CXCL8 plus MMP7 as markers for hepatocyte and cholangiocyte lineage target clusters. Using the KRT19 and CFTR as mature cholangiocyte markers yields highly similar results.
Differential Gene Expression Analysis
Differential gene expression analysis between cells and clusters was performed using the diffexpnb function from the RaceID package. First, negative binomial distributions reflecting the gene expression variability within each subgroup were inferred based on the background model for the expected transcript count variability computed by RaceID3. Using these distributions, a p-value for the observed difference in transcript counts between the two subgroups was calculated and multiple testing corrected by the Benjamini-Hochberg method using the same strategy as described in Anders et al(38).
Pathway Enrichment Analysis and Gene Set Enrichment Analysis
Symbol gene IDs were first converted to Entrez gene IDs using the clusterProfiler(39) package. Pathway enrichment analysis and GSEA(40,41) were implemented using the ReactomePA(42) package. Pathway enrichment analysis was done on genes taken from the different modules in the SOMs. GSEA was done using the differentially expressed genes inferred by the diffexpnb function from the RaceID package.
Validation of protein expression using the Human Protein Atlas
Immunostaining images were collected from the Human Protein Atlas(31) (https://www.proteinatlas.org).
Immunofluorescence
Human liver tissue was fixed overnight in 3.7% formaldehyde (Figure 3j) or cryosectioned and fixed in 2.5% paraformaldehyde for 20 minutes (Extended Data Fig. 7e). The tissue was embedded in OCT and stored at -80°C. The tissue was cryosectioned into 7 micron sections. The tissue was washed twice for 5 min in 0.025% Triton 1x PBS. The tissue was then blocked in 10% FBS with 1% BSA in 1x PBS for 2 hours at room temperature. The dilution used for the anti-human KRT19 (HPA002485, Sigma) and EPCAM (SAB4200704, Sigma) was 1:100 in 100 μL of 1xPBS with 1% BSA. The antibody was incubated overnight at 4°C in the dark. The tissue was washed twice with 0.025% Triton 1x PBS and then incubated with secondary antibodies (donkey anti-rabbit IgG-AF488 (A21206, Thermo Fisher Scientific) and goat anti-mouse IgG-AF568 (A11019) (Fig. 3g) or sheep anti-mouse IgG-AF488 (515-545-062, Jackson ImmunoResearch) and donkey anti-rabbit IgG-RRX (711-295-152, Jackson ImmunoResearch) (Extended Data Fig. 7e) at a 1:200 dilution in 1x PBS with 1% BSA for 1 hour at room temperature. The tissue was then washed twice with 0.025% Triton 1x PBS. DAPI Fluoromount-G (Southern Biotech) was added to the tissue and a coverslip placed on top. Imaging was done using the Zeiss confocal microscope LSM780 (Fig. 3g) or ZEISS Axio Vert.A1. Images were taken at 63x magnification.
Organoid culturing
Organoid culturing was done as previously described(43). The cell populations from the EPCAM+ compartment were sorted on an Aria Fusion 1 using a 100 μm nozzle into tubes containing culture medium supplemented with 10 μM ROCK inhibitor (Y-27632) (Sigma-Aldrich). After sorting, cells were centrifuged in order to remove the medium and then resuspended in 25 μL of Matrigel. Droplets of the Matrigel solution containing the cells were added to the wells of a 24 well suspension plate and incubated for 5-10 min at 37°C until the Matrigel solidified. Droplets were overlaid with 250 μL of liver isolation medium and then incubated at 37°C, 5% CO2. After 3-4 days, the liver isolation medium was replaced with liver expansion medium. For the single cell culture, from each patient, single cells from the TROP2int gate were sorted into the wells of a non-tissue culture treated 96 well plate containing medium with 5% Matrigel. Organoids were passaged 14 days after isolation and then passaged multiple times 5-7 days after splitting. For FACS, single cell suspensions were prepared from the organoids by mechanical dissociation followed by TrypLE (Life Technologies) digestion as previously described(43). Organoid cells were sequenced 5 days after splitting and 17 days after initially sorting the cells for the culture.
Step-by-Step Protocol
A detailed protocol for scRNA-seq of cryopreserved human liver cells is available at Protocol Exchange(44).
Extended Data
Supplementary Material
Acknowledgments
This study was supported by the Max Planck Society, the German Research Foundation (DFG) (SPP1937 GR4980/1-1, GR4980/3-1, and GRK2344 MeInBio), by the DFG under Germany’s Excellence Strategy (CIBSS – EXC-2189 – Project ID 390939984), and by the Behrens-Weise-Foundation (all to D.G.). This work was supported by ARC, Paris and Institut Hospitalo-Universitaire, Strasbourg (TheraHCC and TheraHCC2.0 IHUARC IHU201301187 and IHUARC2019 to T.F.B.), the European Union (ERC-AdG-2014-671231-HEPCIR to T.F.B., EU H2020-667273-HEPCAR to T.F.B, ERC-PoC-2016-PRELICAN to T.F.B), ANRS and the Foundation of the University of Strasbourg. This work was done under the framework of the LABEX ANR-10-LABX-0028_HEPSYS and Inserm Plan Cancer and benefits from funding from the state managed by the French National Research Agency as part of the Investments for the future. We thank Sebastian Hobitz and Konrad Schuldes from the FACS facility and Dr. Ulrike Bönisch from the Deep Sequencing facility of the Max Planck Institute of Immunobiology and Epigenetics. We acknowledge the CRB (Centre de Ressources Biologiques-Biological Resource Centre of the Strasbourg University Hospitals) for the management of regulatory requirements of patient-derived liver tissue. We thank Dr. Catherine Fauvelle and Laura Heydmann (Inserm U1110, University of Strasbourg) for their contributions to the initial single cell isolations used in the study. We thank Drs. Frank Juehling, François Duong and Catherine Schuster (Inserm U1110, University of Strasbourg) for helpful discussions. We thank the patients for providing informed consent to participate in the study and the nurses, technicians and medical doctors of the hepatobiliary surgery and pathology services of the Strasbourg University Hospitals for their support.
Footnotes
Data availability. Data generated during this study have been deposited in Gene Expression Omnibus (GEO) with the accession code GSE124395. The human liver cell atlas can be interactively explored at http://human-liver-cell-atlas.ie-freiburg.mpg.de/.
Author contributions T.F.B. and D.G. conceived the study. N.A. designed, optimized, and performed cell sorting, scRNA-seq experiments, organoid culture, immunofluorescence, provided validation using the Human Protein Atlas, and performed computational analysis and interpretation of the data. A.S. managed the supply of patient material, isolated single cells from patient tissue, performed animal experiments and immunofluorescence. S. contributed to scRNA-seq analyses and performed single-cell RNA-seq experiments. L.M. performed animal experiments. J.S.H. created the web interface. S.D. isolated single cells from patient tissues. P.P. performed liver resections and provided patient liver tissues. T.F.B. established the liver tissue supply pipeline and supervised the animal experiments. D.G. analyzed and interpreted the data and supervised experiments and analysis of N.A. and S. D.G., N.A., and T.F.B. coordinated and led the study. N.A. and D.G. wrote the manuscript with input from S., A.S., and T.F.B.
Author Information Reprints and permissions information is available at www.nature.com/reprints.
Readers are welcome to comment on the online version of the paper.
The authors declare no competing interests.
References
- 1.Michalopoulos GK, DeFrances MC. Liver regeneration. Science. 1997;276:60–66. doi: 10.1126/science.276.5309.60. [DOI] [PubMed] [Google Scholar]
- 2.Ryerson AB, et al. Annual Report to the Nation on the Status of Cancer, 1975-2012, featuring the increasing incidence of liver cancer. Cancer. 2016;122:1312–1337. doi: 10.1002/cncr.29936. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Grun D, van Oudenaarden A. Design and Analysis of Single-Cell Sequencing Experiments. Cell. 2015;163:799–810. doi: 10.1016/j.cell.2015.10.039. [DOI] [PubMed] [Google Scholar]
- 4.Herman JS, Sagar, Grun D. FateID infers cell fate bias in multipotent progenitors from single-cell RNA-seq data. Nat Methods. 2018 doi: 10.1038/nmeth.4662. [DOI] [PubMed] [Google Scholar]
- 5.Grun D, et al. Single-cell messenger RNA sequencing reveals rare intestinal cell types. Nature. 2015;525:251–255. doi: 10.1038/nature14966. [DOI] [PubMed] [Google Scholar]
- 6.Jungermann K, Kietzmann T. Zonation of parenchymal and nonparenchymal metabolism in liver. Annu Rev Nutr. 1996;16:179–203. doi: 10.1146/annurev.nu.16.070196.001143. [DOI] [PubMed] [Google Scholar]
- 7.Gebhardt R. Metabolic zonation of the liver: regulation and implications for liver function. Pharmacol Ther. 1992;53:275–354. doi: 10.1016/0163-7258(92)90055-5. [DOI] [PubMed] [Google Scholar]
- 8.Kietzmann T. Metabolic zonation of the liver: The oxygen gradient revisited. Redox Biol. 2017;11:622–630. doi: 10.1016/j.redox.2017.01.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Halpern KB, et al. Single-cell spatial reconstruction reveals global division of labour in the mammalian liver. Nature. 2017;542:352–356. doi: 10.1038/nature21065. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.MacParland SA, et al. Single cell RNA sequencing of human liver reveals distinct intrahepatic macrophage populations. Nature Communications. 2018;9 doi: 10.1038/s41467-018-06318-7. doi:ARTN 4383. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Haghverdi L, Buttner M, Wolf FA, Buettner F, Theis FJ. Diffusion pseudotime robustly reconstructs lineage branching. Nat Methods. 2016;13:845–848. doi: 10.1038/nmeth.3971. [DOI] [PubMed] [Google Scholar]
- 12.Strauss O, Phillips A, Ruggiero K, Bartlett A, Dunbar PR. Immunofluorescence identifies distinct subsets of endothelial cells in the human liver. Sci Rep. 2017;7 doi: 10.1038/srep44356. 44356. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Halpern KB, et al. Paired-cell sequencing enables spatial gene expression mapping of liver endothelial cells. Nat Biotechnol. 2018;36:962–970. doi: 10.1038/nbt.4231. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Raven A, et al. Cholangiocytes act as facultative liver stem cells during impaired hepatocyte regeneration. Nature. 2017;547:350–354. doi: 10.1038/nature23015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Michalopoulos GK, Barua L, Bowen WC. Transdifferentiation of rat hepatocytes into biliary cells after bile duct ligation and toxic biliary injury. Hepatology (Baltimore, Md) 2005;41:535–544. doi: 10.1002/hep.20600. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Schmelzer E, et al. Human hepatic stem cells from fetal and postnatal donors. J Exp Med. 2007;204:1973–1987. doi: 10.1084/jem.20061603. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Turner R, et al. Human hepatic stem cell and maturational liver lineage biology. Hepatology (Baltimore, Md) 2011;53:1035–1045. doi: 10.1002/hep.24157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Grun D, et al. De Novo Prediction of Stem Cell Identity using Single-Cell Transcriptome Data. Cell Stem Cell. 2016;19:266–277. doi: 10.1016/j.stem.2016.05.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Okabe M, et al. Potential hepatic stem cells reside in EPCAM+ cells of normal and injured mouse liver. Development. 2009;136:1951–1960. doi: 10.1242/dev.031369. [DOI] [PubMed] [Google Scholar]
- 20.Cardinale V, et al. Multipotent stem/progenitor cells in human biliary tree give rise to hepatocytes, cholangiocytes, and pancreatic islets. Hepatology (Baltimore, Md) 2011;54:2159–2172. doi: 10.1002/hep.24590. [DOI] [PubMed] [Google Scholar]
- 21.Kodama Y, et al. Hes1 is required for the development of intrahepatic bile ducts. Gastroenterology. 2003;124:A123–A123. doi: 10.1016/S0016-5085(03)80604-2. [DOI] [Google Scholar]
- 22.Sosa-Pineda B, Wigle JT, Oliver G. Hepatocyte migration during liver development requires Prox1. Nat Genet. 2000;25:254–255. doi: 10.1038/76996. [DOI] [PubMed] [Google Scholar]
- 23.Huch M, et al. Long-term culture of genome-stable bipotent stem cells from adult human liver. Cell. 2015;160:299–312. doi: 10.1016/j.cell.2014.11.050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Betge J, et al. MUC1, MUC2, MUC5AC, and MUC6 in colorectal cancer: expression profiles and clinical significance. Virchows Arch. 2016;469:255–265. doi: 10.1007/s00428-016-1970-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Park S-W, et al. The protein disulfide isomerase AGR2 is essential for production of intestinal mucus. Proc Natl Acad Sci U S A. 2009;106:6950–6955. doi: 10.1073/pnas.0808722106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Forner A, Reig M, Bruix J. Hepatocellular carcinoma. Lancet. 2018;391:1301–1314. doi: 10.1016/S0140-6736(18)30010-2. [DOI] [PubMed] [Google Scholar]
- 27.Matkowskyj KA, et al. Aldoketoreductase family 1B10 (AKR1B10) as a biomarker to distinguish hepatocellular carcinoma from benign liver lesions. Hum Pathol. 2014;45:834–843. doi: 10.1016/j.humpath.2013.12.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Rantakari P, et al. The endothelial protein PLVAP in lymphatics controls the entry of lymphocytes and antigens into lymph nodes. Nat Immunol. 2015;16:386–396. doi: 10.1038/ni.3101. [DOI] [PubMed] [Google Scholar]
- 29.Grompe M, Strom S. Mice with human livers. Gastroenterology. 2013;145:1209–1214. doi: 10.1053/j.gastro.2013.09.009. [DOI] [PubMed] [Google Scholar]
- 30.Azuma H, et al. Robust expansion of human hepatocytes in Fah-/-/Rag2-/-/Il2rg-/- mice. Nat Biotechnol. 2007;25:903–910. doi: 10.1038/nbt1326. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Uhlen M, et al. Proteomics. Tissue-based map of the human proteome. Science. 2015;347 doi: 10.1126/science.1260419. 1260419. [DOI] [PubMed] [Google Scholar]
- 32.Krieger SE, et al. Inhibition of hepatitis C virus infection by anti-claudin-1 antibodies is mediated by neutralization of E2-CD81-claudin-1 associations. Hepatology. 2010;51:1144–1157. doi: 10.1002/hep.23445. [DOI] [PubMed] [Google Scholar]
- 33.Lieber A, Peeters MJ, Gown A, Perkins J, Kay MA. A modified urokinase plasminogen activator induces liver regeneration without bleeding. Hum Gene Ther. 1995;6:1029–1037. doi: 10.1089/hum.1995.6.8-1029. [DOI] [PubMed] [Google Scholar]
- 34.Mailly L, et al. Clearance of persistent hepatitis C virus infection in humanized mice using a claudin-1-targeting monoclonal antibody. Nat Biotechnol. 2015;33:549–554. doi: 10.1038/nbt.3179. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Hashimshony T, et al. CEL-Seq2: sensitive highly-multiplexed single-cell RNA-Seq. Genome Biol. 2016;17:77. doi: 10.1186/s13059-016-0938-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Li H, Durbin R. Fast and accurate long-read alignment with Burrows-Wheeler transform. Bioinformatics. 2010;26:589–595. doi: 10.1093/bioinformatics/btp698. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Grun D, Kester L, van Oudenaarden A. Validation of noise models for single-cell transcriptomics. Nat Methods. 2014;11:637–640. doi: 10.1038/nmeth.2930. [DOI] [PubMed] [Google Scholar]
- 38.Anders S, Huber W. Differential expression analysis for sequence count data. Genome Biol. 2010;11:R106. doi: 10.1186/gb-2010-11-10-r106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Yu G, Wang L-G, Han Y, He Q-Y. clusterProfiler: an R package for comparing biological themes among gene clusters. Omics. 2012;16:284–287. doi: 10.1089/omi.2011.0118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Subramanian A, et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci U S A. 2005;102:15545–15550. doi: 10.1073/pnas.0506580102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Mootha VK, et al. PGC-1alpha-responsive genes involved in oxidative phosphorylation are coordinately downregulated in human diabetes. Nat Genet. 2003;34:267–273. doi: 10.1038/ng1180. [DOI] [PubMed] [Google Scholar]
- 42.Yu G, He QY. ReactomePA: an R/Bioconductor package for reactome pathway analysis and visualization. Mol Biosyst. 2016;12:477–479. doi: 10.1039/c5mb00663e. [DOI] [PubMed] [Google Scholar]
- 43.Broutier L, et al. Culture and establishment of self-renewing human and mouse adult liver and pancreas 3D organoids and their genetic manipulation. Nat Protoc. 2016;11:1724–1743. doi: 10.1038/nprot.2016.097. [DOI] [PubMed] [Google Scholar]
- 44.Aizarani N, et al. Protocol for Single-Cell RNA-Sequencing of Cryopreserved Human Liver Cells. Protoc Exch. 2019 [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.