Main

The liver is organized in lobules, hexagonal functional units with a portal region in each corner and a central vein as the mid-point4. At the cellular level, the liver is composed of two epithelial cell types: hepatocytes and cholangiocytes, also known as ductal cells, and several stromal, endothelial and immune resident subpopulations5,6. Hepatocytes are uniquely polarized cells, with one or more apical poles that contribute to bile canaliculi, tubular lumina formed by adjacent hepatocytes. Bile canaliculi form a continuous network of tubes 1.5 to 2.5 µm in diameter, which drain bile into cholangiocyte-lined bile ductules and bile ducts7. Dysregulation of bile flow or the connection between bile canaliculi and bile duct results in cholestatic liver diseases, which can progress to fibrosis, cirrhosis and ultimately liver failure2,6.

In cholestatic disease, portal mesenchyme and, specifically, portal fibroblasts and cholangiocytes become activated and expand, in both animal models (bile duct ligation and Mdr2−/− mice (Mdr2 is also known as Abcb4))4,8 and human disease1,9. During cholestatic disease, the interactions between the different cell populations are disrupted, which suggests that restoring them could help the repair response2,10. Spatial transcriptomic analysis revealed many of the molecules involved in the cell–cell communication in the periportal region in healthy11,12 and cholestatic13 liver tissue. However, how these contribute to homeostasis, regeneration or fibrosis remains largely unknown. This is because these interactions are highly dynamic, occur over short timescales, and require imaging with cellular resolution, which makes them extremely challenging to study in vivo, in living animals.

None of the in vitro systems that have been developed to date have modelled the complex homeostatic or fibrotic periportal cellular interactions and the periportal specialized functions and mesoscale architecture of the liver3. Tissue-derived adult mouse ductal (cholangiocyte)14 or hepatocyte15,16 organoids are made up solely of epithelial cells. Epithelial co-cultures of cholangiocyte and hepatocyte progenitors in 2D have been reported17, but similar to hepatocytes cultured in 2D sandwich culture18, they do not recapitulate the 3D network properties of the bile canaliculi network or contain stromal cells to recapitulate the periportal epithelial–stromal interactions17. Similarly, 3D liver organoids derived from pluripotent stem cells remain immature and do not replicate adult architecture19. Previous studies showed that cholangiocyte–mesenchyme organoid co-cultures retain aspects of 3D tissue morphology and the binary cell interactions20. However, this model lacks the other cell types in the periportal region.

Here, we developed hepatocyte organoids (HepOrgs) with a physiological 3D bile canaliculi network and combined them with cholangiocytes and portal mesenchyme to generate periportal liver organoids (periportal assembloids) that recapitulate the in vivo bile canaliculi–bile duct cell–cell interactions and architectural organization. Using this system, we model many aspects of biliary liver fibrosis, including hepatocyte death, matrix deposition and ductal cell expansion, but not the inflammatory reaction. Additionally, we provide proof of concept that liver assembloids can be used to investigate molecular and cell-autonomous mechanisms in liver disease.

HepOrgs form 3D bile canaliculi network

To ensure proper bile flow, hepatocytes must form a functional and physiological bile canaliculi network to connect to the bile duct lined by cholangiocytes surrounded by portal fibroblasts (Fig. 1a). Hepatocytes cultured in 2D sandwich form bile canaliculi18, but do not generate the in vivo 3D network (Extended Data Fig. 1a,b). HepOrgs express bile canaliculi markers—however, the presence of a network has not been studied15,16. By staining for canaliculi and quantifying bile canaliculi length and branching, we found that the reported models15,16 lack sufficient bile canaliculi to generate a network, which would ensure a reliable bile canaliculi–bile duct connection (Fig. 1d,e, compare Hu et al.15 and Peng et al.16 with tissue and Extended Data Fig. 1b–f). Thus, building upon the existing models, we first optimized HepOrgs to generate a physiologically relevant bile canaliculi network, while maintaining sufficient organoid expansion potential.

Fig. 1: HepOrgs retain marker expression and cell polarity of tissue.
figure 1

a, Schematic (left) and representative immunofluorescence image (right; n = 5) of the liver periportal region. CD34 marks portal fibroblasts (PFs), osteopontin (OPN) marks ductal cells, phalloidin marks membranes and DAPI labels nuclei. Chol, cholangiocyte; Hep, hepatocyte; PV, portal vein. Scale bar, 10 µm. b, Representative bright-field images of HepOrgs at passage 1 (P1) cultured under the conditions described in Hu et al.15, Peng et al.16 or in HM-FBS, HM-Wnt or HM-WntS (n = 5 experiments). Scale bars: 500 µm (top row), 10 µm (bottom row). FBS, foetal bovine serum. c, Representative BF images of HepOrgs cultured in HM-Wnt at the indicated time points. Scale bars: 50 µm (day 9), 100 µm (day 49), 200 µm (day 203 and day 355). d, Immunofluorescence staining and 3D reconstruction of bile canaliculi (marked by CD13) in HepOrgs cultured in indicated conditions and mouse tissue. HepOrgs cultured in HM-Wnt have longer bile canaliculi compared with previous studies15,16. Cell borders are indicated by filamentous actin (F-actin) staining with phalloidin (Phall). Top, maximum-intensity projections of confocal images. HepOrgs are outlined with dashed lines. Bottom, bile canaliculi segmentation and 3D reconstruction. Scale bars, 50 µm. e, Number of triple junctions in the largest bile canaliculi network in tissue and organoids cultured in indicated conditions. Dots show the total number of triple junctions per structure and the line represents the mean. Kruskal–Wallis test with Dunn’s multiple comparisons post hoc test. NS, not significant. f, Organoid formation efficiency. Dots represent biologically independent samples (n = 5 biological replicates with at least 2 technical replicates) and the line represents the mean. Kruskal–Wallis test with Dunn’s multiple comparisons post hoc test. g, Immunofluorescence staining of tissue (left) and hepatocyte and cholangiocyte organoids (HepOrgs and CholOrg, respectively) grown in HM-Wnt (right), for the hepatocyte marker HNF4α and cholangiocyte markers KRT19 and OPN (n = 2 experiments). Scale bars: 50 µm (main images), 10 µm (smaller images). Phalloidin marks membranes and DAPI labels nuclei. h, Left, schematic showing hepatocyte polarity. CD13 (apical) and ECAD (basolateral) staining in tissue (middle) and organoids (right) shows similar distributions of markers. Yellow arrowheads indicate binucleated cells. Scale bars: 20 µm (left most images), 10 µm (expanded views). All data were obtained from n = 3–5 independent experiments.

Source Data

Wnt signalling is essential to increase hepatocyte21,22,23 and cholangiocyte14 proliferation during mouse liver regeneration and in liver cancer24. Consequently, we supplemented our previously published medium25 with WNT3a ligand26 (HM-Wnt) or Wnt surrogate27 (HM-WntS). Organoids increased in size and cell numbers, presented significantly higher organoid formation efficiency (twofold to threefold increase) compared with the previous method, and could be maintained long term (Fig. 1b–f and Extended Data Fig. 1g–j). This size increase was not due to organoid merging, as cells seeded in sparse culture conditions also expanded over time (Extended Data Fig. 1i).

We observed heterogeneity in the shape of the generated structures, with some being spherical with smooth surfaces (referred to here as ball) and others exhibiting a folded shape with coarse surfaces (bubbly/grape-like) (Extended Data Fig. 2a). Using topological data analysis and the shape descriptor algorithm DETECT28 (detecting temporal shape changes with the Euler characteristic transform; Methods and Supplementary Methods, section 2), we confirmed that the ball and bubbly/grape-like structures represent quantitatively different shapes that cluster independently of each other (Extended Data Fig. 2b–d). Live imaging revealed that the majority of the ball-shaped structures started from single hepatocytes, whereas hepatocyte doublets or clusters of more than two cells generated bubbly/grape-like structures (23% versus 87% efficiency) (Extended Data Fig. 2e,f). Both types of structures presented similar gene expression and function (Extended Data Fig. 2g–j), however they differed in their bile canaliculi architecture. Bubbly/grape-like structures presented thinner, longer and more interconnected bile canaliculi and consistently secreted the bile acid analogues cholyl-l-Lys-fluorescein (CLF) and metabolized 5-carboxyfluorescein diacetate, acetoxymethyl ester (5-CFDA). Ball-like structures did not secrete CLF or metabolize 5-CFDA, but accumulated the bile acid analogues inside of hepatocytes, confirming aberrant or absent bile canaliculi (Extended Data Fig. 2k,l and Supplementary Videos 1 and 2). This observation correlated with an increase in cleaved-caspase 3 staining (Extended Data Fig. 2k), as expected from intracellular bile acid accumulation.

These results indicate that marker gene expression is not sufficient to guarantee a functional and physiological bile canaliculi network or metabolite transport. Hereafter we used HepOrg cultures that were enriched for bubbly/grape-like organoid shape.

The optimized HepOrgs retained hepatocyte identity, expressed hepatocyte markers (such as HNF4α and albumin) and the bile canaliculi transporters MDR2 (encoded by Abcb4) and MRP2 (encoded by Abcc2), but not ductal cell (for example, SOX9 and KRT19) or non-epithelial markers, thereby resembling freshly isolated hepatocytes (Fig. 1g, Extended Data Fig. 3a–f and Supplementary Table 2). HepOrgs secreted similar amounts of albumin as previously published 3D organoid models and more albumin than 2D hepatocyte sandwich cultures (Extended Data Fig. 3g,h). The optimized HepOrgs also recapitulated the complex hepatocyte cell polarity of the in vivo tissue, with the polarity markers ZO-1 and CD13 localized to the apical surface of adjacent hepatocytes and E-cadherin (ECAD) localized to the basolateral side (Fig. 1h and Extended Data Fig. 3c,e). Image analysis revealed that the optimized HepOrgs formed a significantly longer and more branched bile canaliculi network within each organoid and were closer to the tissue values compared with the previous methods (Fig.1e and Extended Data Fig. 1e,f).

Occasionally, we found cholangiocyte organoids growing in close proximity to HepOrgs, forming independent structures that were neither embedded nor formed physiological bile canaliculi–bile duct connections with hepatocytes (Extended Data Fig. 4a–e). We therefore further tested additional culture conditions with the aim of generating structures that would retain the complex hepatocyte polarity and support the culture of the three cell types while preserving their physiological ratios. In all conditions tested, HepOrgs were viable and retained the expression of hepatocyte markers (Extended Data Fig. 4f–i). Our previously published minimal medium (MM) for co-culture of ductal and portal mesenchyme cells20 was superior to the other media that we tested, and supported the maintenance of cholangiocytes and mesenchyme without their overgrowth (Extended Data Fig. 4j). Hepatocyte function was similar but hepatocytes from HepOrgs cultured in MM were more multinucleated, which is considered a hallmark of mature hepatocytes (Extended Data Fig. 4k–m). RNA-sequencing (RNA-seq) analysis revealed that in both culture conditions, HepOrgs maintain the expression of genes encoding hepatocyte markers such as Alb, Hpx and Cyp3a1, and bile canaliculi transporters (Abcb11 (also known as Bsep), Abcc2 (also known as Mrp2) and Abcb4 (also known as Mdr2)), in similar amounts as freshly isolated hepatocytes (Extended Data Fig. 5a and Supplementary Table 2). However, HepOrgs cultured in MM expressed higher levels of periportally zonated genes (Fbp1, Arg1 and Aldob), whereas pericentral genes such as Glul (encoding glutamine synthetase) and Axin2 were expressed in both conditions (Extended Data Fig. 5a). These observations suggested that the cultures exhibited some degree of zonation. To assess this, we performed single-cell RNA-seq (scRNA-seq) analysis of HepOrgs cultured in MM and our optimized expansion medium HM-Wnt (Methods). To underscore potential zonated gene expression, we generated a consensus pericentral or periportal score using publicly available datasets13,29,30 (Methods). HepOrgs cultured in HM-Wnt scored more pericentrally, whereas MM HepOrgs scored more periportal, in agreement with the expression of selected zonated markers (Extended Data Fig. 5b–d). RNAscope and immunofluorescence analysis for periportal (Gls2 RNA and albumin and ECAD protein) and pericentral (Cyp1a1 RNA and CYP2E1 and glutamine synthetase protein) markers confirmed the heterogeneous distribution of these markers, with pericentral genes being more highly expressed on the periphery of an organoid, where cells are more exposed to the WNT ligand from the medium (Fig. 2a and Extended Data Fig. 5e,f). Together, the results suggested that HepOrg cultured in MM acquired a more periportal gene expression signature.

Fig. 2: HepOrgs exhibit physiological and functional bile canaliculi size and network resembling tissue.
figure 2

a, Left, schematic of liver zonation. Right, maximum-intensity projections of staining from periportal (albumin (Alb)) and pericentral (glutamine synthetase (GS)) markers in tissue (top row) and HepOrgs cultured in MM (bottom row). Albumin and glutamine synthetase also shown as a Fire look-up table in the middle and right images (n = 3 independent experiments). Cells expressing glutamine synthetase are detected in the periphery of the organoids. Scale bars: 50 µm (top row), 20 µm (bottom row). CV, central vein. b, Histogram showing distribution of bile canaliculi diameters in tissue and HepOrgs. The curve represents the kernel density estimate, used here to estimate the probability density function of a continuous random variable, showing a smooth curve that represents the distribution of data points provided in Source Data. n = 3 independent experiments. c, Immunofluorescence staining (left), image reconstruction and analysis of bile canaliculi (BC) from HepOrgs cultured in HM-Wnt or MM, and tissue stained for CD13 (marking bile canaliculi), DAPI (nuclei) and phalloidin (F-actin). Individual interconnected networks are pseudo-coloured. The skeleton of the bile canaliculi network shows that the networks are longer and more interconnected in HepOrgs cultured in MM. Scale bar, 20 µm. n = 3 independent experiments. d, Left, length of the largest bile canaliculi network in HepOrgs cultured in HM-Wnt or MM, and tissue. Right, total number of junctions in the largest bile canaliculi network. Dots show the largest network in an individual organoid. Horizontal line indicates the median. HM-Wnt, n = 6; MM, n = 5; tissue, n = 6. Kruskal–Wallis test with Dunn’s multiple comparisons post hoc test. e, Transport of CLF and CMFDA in HepOrgs cultured in MM confirm the functionality of bile acid transporters. Compounds are shown in Royal look-up tables. Nuclei (SPY555-DNA (SPY-DNA)) and actin (SiR-actin (SiR-act)) are labelled (n = 3 independent experiments). Scale bars for each set of images are 50 µm and 10 µm (magnification). Images are stills from time-lapse imaging shown in Supplementary Videos 3 and 4.

Source Data

Of note, image analysis and reconstruction demonstrated that HepOrgs cultured in MM presented longer bile canaliculi, as well as narrower distributions of bile canaliculi diameter, which were within the physiological range31 of 1.5 to 2.5 µm, narrower than the 2 to 7 µm diameter of bile canaliculi formed by HepOrgs cultured in HM-Wnt (Fig. 2b–d and Extended Data Fig. 6a–c). Notably, the bile canaliculi network connectivity was further improved in MM compared with HM-Wnt, resembling that of the liver tissue (Fig. 2d, right). We tested the functionality of MRP2, BSEP and MDR2 bile transporters located in the canalicular membrane using fluorescently labelled bile acid analogues CLF and chloromethylfluorescein diacetate (CMFDA) (which indicate active BSEP and MRP2 transport) and the fluorescent phosphatidylcholine marker (for MDR2-mediated transport). We consistently observed uptake and transport of the fluorescent labels to the canalicular lumen (Fig. 2e, Extended Data Fig. 6d and Supplementary Videos 35), confirming the functionality of the bile canaliculi network, in agreement with the gene expression of apical transporters in our HepOrg cultures (Extended Data Fig. 6e).

Thus our optimized HepOrg system enables the growth of adult hepatocytes that preserve hepatocyte polarity, show partial hepatocyte zonation and generate a physiological and functional bile canaliculi 3D network resembling the in vivo adult liver tissue, without the emergence of cholestatic features that have been found in previous 2D hepatocyte cultures. Critically, our results highlight the importance of mimicking physiological properties at the cellular and tissue scale level, as these directly impact the overall fitness of organoid models.

Assembloids mimic periportal architecture

We next aimed to reconstruct the periportal region of the liver lobule, specifically the heterotypic cellular interactions between hepatocytes, cholangiocytes and portal mesenchyme. We adapted the concept of assembloids32 by mixing defined numbers of single cholangiocytes from ductal organoids (grown as described14) and portal mesenchymal cells (grown as described20) with a defined number of HepOrgs and cultured them in MM (Methods). We opted for a two-pronged strategy, using either a rocking platform or AggreWell plates (Fig. 3a and Methods). Both methods attained multicellular structures with high efficiency (around 70% of the total HepOrgs), which was further increased (to around 90%) when HepOrgs were pre-conditioned in the co-culture medium (Fig. 3b,c and Extended Data Fig. 7a–c). We selected the rocking platform method for subsequent experiments. The majority of assembloids were formed within the first 48 h, although assembloids containing mesenchyme could also aggregate after seeding (Extended Data Fig. 7d,e and Supplementary Videos 6 and 7). Notably, the composite structures retained the cellular proportions of the tissue (Fig. 3d) and recapitulated the tissue organization, with cholangiocytes forming bile duct structures with opened lumens (marked by KRT19, SOX9 and tdTomato, asterisk) surrounded by portal mesenchymal cells (marked by vimentin, elastin and PDGFRα–H2B–GFP) and embedded in the hepatocyte parenchyma (marked by HNF4α) (Fig. 3b,e and Extended Data Fig. 7a,f,g, comparing organoid versus tissue), thus recapitulating the mesoscale architecture of the native tissue.

Fig. 3: Assembloids recapitulate mesoscale tissue architecture and gene expression.
figure 3

a, Schematic of the experimental approach. Scale bars: 200 µm (top), 500 µm (bottom). Msc, mesenchyme. b, Representative images (n > 3 experiments) of assembloids cultured on a rocking platform (left) compared with tissue (right). PDGFRα–H2B–GFP marks mesenchyme, nuclear-tdTom and SOX9 mark cholangiocytes. Arrowheads indicate binucleated hepatocytes. Scale bars: 50 µm (main images), 20 µm (assembloids, zoom view), 10 µm (tissue, zoom view). c, Aggregation efficiency. Data are mean ± s.e.m. of n = 3 biological replicates from n = 3 independent experiments; Mann–Whitney test, two-tailed. d, Cellular composition of assembloids. Data are mean ± s.e.m. of assembloids from at least 3 independent experiments (n = 13 organoids total). Dots represent the percentage of hepatocyte, cholangiocyte or portal mesenchyme cells per structure. e, Representative confocal images of assembloids stained for the indicated markers. n > 3 experiments. Asterisks indicate bile duct lumen. Scale bars, 50 µm. f, Hepatocyte, cholangiocyte and mesenchyme marker expression in assembloids. Haemopoietic/endothelial markers (H/E) are not expressed. EGFP marks mesenchyme, tdTomato (tdTom) marks cholangiocytes. g, Uniform manifold approximation and projection (UMAP) from liver atlas datasets and assembloids (this study). h, Mesenchyme (green), hepatocytes (blue) and cholangiocytes (magenta) superimposed on UMAP data from g. i, Assembloid data superimposed on data from g. j, Immunofluorescence staining and image reconstruction of the connection between bile canaliculi from hepatocytes (ZO-1, CD13) and the lumen from bile duct (KRT19, PCK) in assembloids (top) and tissue (bottom). Right, 3D reconstruction visualizes hepatocytes (red, yellow) whose bile canaliculi (green) enter the bile duct lumen (magenta). n = 6 independent experiments, see Supplementary Videos 8 and 9. Scale bar, 10 µm. k,l, Schematic (k) and still images (l) of CLF transport (shown as Fire look-up table) from the live imaging shown in Supplementary Video 12 in assembloids indicates functional connection between bile canaliculi and bile duct lumen (mem-tdTomato). n = 3 independent experiments with n = 3 biological replicates. BA, bile acid analogue. Scale bar, 50 µm.

Source Data

scRNA-seq analysis indicated that the assembloid cells retain their identities and expression signatures, with hepatocytes, cholangiocytes and mesenchymal cells expressing known lineage markers such as Alb (hepatocytes), Krt19 (cholangiocytes) and Col1a1 (mesenchyme), respectively (Fig. 3f). Mesenchymal cells were enriched for basement membrane, metallopeptidase activity and extracellular matrix signatures, cholangiocytes were enriched for cell–cell junctions, and hepatocytes were enriched for metabolic processes and cholesterol transport, among others (Fig. 3f, Extended Data Fig. 7h–m and Supplementary Table 3). The assembloid populations mostly overlapped with in vivo hepatocyte, cholangiocyte and portal mesenchymal cells from several mouse liver cell atlases11,12,13,33,34,35,36,37,38,39 (n = 10; Fig. 3g–i, Extended Data Fig. 8a–d and Methods). Marker gene analysis and quantitative PCR with reverse transcription (RT–qPCR) on sorted cells from assembloids compared with freshly isolated cells further confirmed that the cells mostly retain the expression profile of adult mouse liver tissue (Extended Data Fig. 8f–i). As expected, the mesenchyme most closely resembled portal fibroblasts and clustered further away from other mesenchymal populations (hepatic stellate cells (HSCs) and vascular smooth muscle cells (VSMCs)) (Extended Data Fig. 8c,d), whereas the hepatocytes of the assembloids retained a certain degree of zonation, with the expression of periportally zonated markers (Extended Data Fig. 8e).

Next, we investigated the fine detail of the tissue architecture at the cellular scale—specifically, whether the bile duct was functionally connected to the hepatocyte bile canaliculi network, akin to the tissue’s canal of Hering structure4. Immunofluorescence staining revealed that the connection between bile canaliculi and bile duct in periportal assembloids recapitulated that of the native liver tissue in 100% of the cases where cholangiocytes were incorporated inside the structure (Fig. 3j, Extended Data Fig. 9a–g and Supplementary Videos 811; compare assembloid to tissue). Frequently, several bile canaliculi joined one bile duct lumen, usually surrounded by portal mesenchymal cells in close vicinity, as in the tissue (Fig. 3j, Extended Data Fig. 9a–g, bottom and Supplementary Videos 10 and 11). The bile canaliculi–bile duct connection was functional—since we readily detected CLF transported from the hepatocyte bile canaliculi network into the bile duct lumen (Fig. 3k,l, Extended Data Fig. 9h and Supplementary Videos 12 and 13)—at timescales that are close to those in the native tissue40. This fine cellular-scale detail was not observed in assembloids with aberrant ratio and non-physiological arrangements of cholangiocytes to hepatocytes or in cholangiocyte-only organoids (Extended Data Fig. 9i and Supplementary Video 14). Furthermore, we found that the bile canaliculi–bile duct connection further improved the differentiation of the cells in the structures. We observed that hepatocytes increased expression of several bile acid transporter genes (Abcb4, Abcc2 and Abcc3), whereas cholangiocytes expressed Ezr and Ano1, apical polarity and transporter markers, respectively (Extended Data Fig. 9j,k).

Collectively, these results confirm that periportal assembloids recapitulate the expression pattern, cellular function and periportal tissue architecture of the native tissue, at both the meso and cellular scale. In addition, our results further highlight that modelling a physiological bile canaliculi architecture, function and connection to the bile duct has benefits for the overall maturation of cells and the overall robustness of the assembloid model.

Assembloids model biliary fibrosis

Since portal mesenchyme contributes to mouse and human biliary fibrosis1,9, we investigated whether our periportal assembloid model could recapitulate aspects of this disease. We performed experiments in which we kept the hepatocyte and cholangiocyte numbers constant, but increased the initial number of portal mesenchymal cells (Fig. 4a). Non-physiological numbers of portal mesenchyme (10× excess) consistently resulted in structures with altered morphology compared with structures with physiological numbers (Fig. 4b). Gene expression analysis (RNA-seq) indicated that assembloids with high mesenchymal cell ratio exhibited increased expression of ductal cell markers (Krt7, Sox9 and Krt19) and several collagens (Col1a2 and Col3a1), which were persistent or even further increased in assembloids cultured longer-term (2.5 weeks; Extended Data Fig. 10a and Supplementary Table 2). Next, we compared scRNA-seq analysis of fibrotic versus homeostatic assembloids to nine publicly available single-cell liver datasets from in vivo mouse models of liver fibrosis11,12,13,33,34,35,38,39 (Fig. 4c and Extended Data Fig. 10b–h). Cholangiocytes from fibrotic-like assembloids clustered closely to cholangiocytes from various damage models, whereas hepatocytes from fibrotic-like assembloids clustered closely to cholangiocytes, potentially suggestive of a degree of transdifferentiation. Similarly, mesenchyme from fibrotic-like assembloids closely correlated to fibroblasts from several damage models (Extended Data Fig. 10h). We observed a remarkable similarity, both in strength and magnitude, between the inferred cell–cell interactions from in vivo biliary fibrosis models33 and our fibrotic assembloids. The predicted mesenchyme–mesenchyme interactions included Col1a1–Ddr2, Gas6–Axl and the metalloproteases inhibitor gene interactions Timp2–Itgb1, Timp1–Cd63, which are implicated in fibrosis progression33,41,42,43. Among the hepatocyte–mesenchyme interactions, the most notable were Fgb with Itgb1 (Fig. 4d and Supplementary Table 4). We found a significant enrichment in signalling pathways involved in liver fibrosis and known to be activated in cholestatic injury such as IL-6 signalling, collagen remodelling, deposition and degradation, matrix metalloproteases, extracellular matrix turnover and cytokine signalling (Fig. 4e and Supplementary Table 5). Hepatocytes showed upregulation of stress-related pathways such as p53 signalling and downregulation of bile acid and fatty acid metabolism (Extended Data Fig. 10e). Conversely, cholangiocytes showed a reduction in apoptosis and TGFβ signalling (Extended Data Fig. 10f), whereas mesenchymal cells presented changes in signatures and pathways known to be involved in liver fibrosis such as PI3K–AKT–mTOR44 (Extended Data Fig. 10g).

Fig. 4: Periportal assembloids mimic aspects of biliary fibrosis in vitro.
figure 4

a, Schematic of the experimental design. b, Immunofluorescence images of assembloids with homeostatic (left) and 10× excess (right) of mesenchyme. n > 3 experiments. Scale bars: 50 µm (main images), 10 µm (zoom view). c, UMAP of fibrotic-like assembloid data (from this study) integrated with datasets from liver damage models. d, Circular plots represent the inferred cell–cell interactions from fibrotic-like assembloids, bile duct ligation (BDL) and CCl4 models from Yang et al.33. Interactions reported in the literature and shared with BDL and CCl4 models are shown in red, interactions that are shared but not mentioned in the literature are in cyan and unique interactions are shown in black. Selected from the top 100 significant interactions for BDL. e, Gene set enrichment analysis (GSEA) of fibrosis-like versus homeostatic-like assembloids using MSigDB_Hallmark_2020 (black), KEGG_2019 (red) and Reactome_2022 (cyan) gene datasets. EMT, epithelial to mesenchymal transition; NES, normalized enrichment score. f, Cell composition of homeostatic-like and fibrotic-like assembloids. Dots show the percentage of cells per structure. Data are mean ± s.e.m. (3 independent experiments, n = 13 organoids); homeostatic-like data are reproduced from Fig. 3d. Mann–Whitney test, two-tailed. g, Left, immunofluorescence staining for cleaved caspase-3 (CASP3). Right, violin plot showing the median and quartiles of the percentage distribution of hepatocytes containing cleaved caspase-3 from three independent biological replicates. Each dot represents one organoid. Mann–Whitney test, two-tailed. Hom, homeostatic (n = 8); Fib, fibrotic-like (n = 7). Scale bars: 100 µm (left), 50 µm (middle), 25 µm (right). h, Left, SHG imaging reveals fibrous collagen deposition; cholangiocytes (nuc-tdTomato), mesenchyme (PDGFRα–H2B–GFP) and SiR-actin staining. Right, mean ± s.e.m. of the intensity of SHG signal from three biological replicates from n = 3 independent experiments. Mann–Whitney test, two-tailed. Homeostatic assembloids, n = 10; fibrotic-like assembloids, n = 4. Scale bars, 20 µm. i, Expression of selected genes in mesenchyme, presented as abundance, in homeostatic (Ctrl) and fibrosis-like (Damage) assembloids, or in damage or control tissue from BDL and CCl4 models from ref. 33.

Source Data

Together, these results suggested that the structures with high mesenchyme cell ratios exhibited a fibrotic-like phenotype; therefore, we called them ‘fibrotic-like’, in contrast to ‘homeostatic’ for the assembloids with physiological mesenchymal cell numbers.

Notably, in fibrotic-like assembloids, but not in homeostatic assembloids, we observed that the increase of the initial numbers of portal mesenchymal cells (SCA1+PDGFRα+) affected the other two populations: hepatocytes and ductal cells (Fig. 4f). We observed a significant reduction in hepatocyte number, which also exhibited aberrant morphologies, distorted cell membranes and weak chromatin staining (Fig. 4g and Extended Data Fig. 11a). Live imaging consistently revealed big bursts of DNA signal coming from hepatocytes (Supplementary Video 15). Cleaved caspase-3 staining indicated that hepatocyte death occurred, at least partially, through apoptosis, although gene expression analysis also indicates that other forms of cell death might be involved (Fig. 4g and Extended Data Fig. 11a–c). Hepatocyte death was also induced by freshly isolated portal mesenchymal cells in excess (fibrotic-like) numbers (Extended Data Fig. 11c). Consistently fibrotic-like assembloids lost functional bile acid (CLF) uptake and reduced bile acid drainage from the bile canaliculi into the bile duct (Extended Data Fig. 11d–f and Supplementary Video 16), although we did not detect differences in albumin secretion, total bile acid or cytochrome activity between homeostasis and fibrotic-like assembloids (Extended Data Fig. 11g). Concomitant with the hepatocyte death, we observed increased numbers of ductal cells in fibrotic-like assembloids (Fig. 4f and Extended Data Fig. 11h), which were further increased in long-term cultured assembloids (more than 40 days; Extended Data Fig. 11i,j), reminiscent of the ductular reaction observed in biliary fibrosis patients2. Notably, second-harmonic generation (SHG) microscopy confirmed a significant increase in fibrillar collagen deposition in fibrotic-like assembloids (Fig. 4h), in agreement with the gene expression data (Fig. 4i) and a mouse model of biliary fibrosis2,8. Immunostaining confirmed the identity of portal fibroblasts (Extended Data Fig. 11k). In addition, several genes encoding pro-inflammatory molecules, including Ccl11, Cxcl1 and Cxcl12 and the metalloproteases Mmp2 and Mmp3—all of which are implicated in fibrosis—were also highly upregulated in the mesenchyme of fibrotic-like assembloids (Extended Data Fig. 12a,b and Supplementary Table 4). Cytokine array analysis confirmed the secretion of CCL11, CXCL1, MMP2 and MMP3 by fibrotic-like assembloids, but not from matching control homeostatic assembloids (Extended Data Fig. 12c).

Together, the described features—namely: (1) fibrotic gene expression similar to in vivo models of biliary fibrosis; (2) hepatocyte death; (3) bile flow obstruction; (4) ductal cell expansion; and (5) collagen deposition—indicate that fibrotic assembloids recapitulate in vitro many aspects of the in vivo biliary fibrosis, except for the inflammatory reaction, as expected given that our system lacks the immune compartment.

Assembloids as a tool to study fibrosis

We next investigated whether assembloids could be used as a tool to study liver fibrosis. We first examined mesenchyme-derived inflammatory cytokines, as recent human studies on biliary fibrosis45 suggest that epithelium and mesenchyme express many cytokines, although their function remains unknown. Addition of CXCL12, CCL11 or CXCL1 had no effect on mesenchymal cells (Extended Data Fig. 12d). Similarly, blocking antibodies against selected cytokines did not rescue the fibrotic-like phenotype (Extended Data Fig. 12e). These results suggested that the paracrine signalling from mesenchymal cells was secondary to the fibrotic-like phenotype, at least for the cytokines tested, and notably, in the absence of the immune compartment.

Changes in cell adhesion and cell–extracellular matrix (ECM) interactions are common features of the fibrotic response. Therefore, we next tested whether assembloids could be exploited to investigate cell–cell and cell–ECM interactions in fibrosis. We first analysed our scRNA-seq results for potential cell adhesion, cell–ECM or ligand–receptor interactions that would be increased in the fibrotic assembloids compared with the homeostatic ones, focusing on the mesenchymal cell interactions that were also present in in vivo models of fibrosis (Fig. 4d and Extended Data Fig. 12a,b). We found that expression of the cell adhesion gene Cdh11 was increased in fibrotic-like mesenchymal cells, and the ligand–receptor interactions involving molecules including the ECM integrin subunit ITGβ1 and the ECM modulators TIMP1 and TIMP2 (Extended Data Fig. 12a,b and Supplementary Table 4) significantly changed in intensity and magnitude. We selected some of these genes to perform a small knockdown screen in assembloids cultured in fibrotic-like conditions (tenfold excess mesenchymal cells). As a first screening readout, we discriminated shape changes over time, as fibrotic-like assembloids are more compact compared to structures with physiological mesenchymal cell numbers. We again applied the shape descriptor algorithm DETECT28 and developed the DETECT metric to quantitatively and statistically compare spatiotemporal changes in morphology of organoids under different conditions. We found that inhibiting mesenchymal cell adhesion by CDH11 knockdown as well disrupting mesenchyme–ECM interactions by knocking out ITGβ1 specifically in mesenchyme cells consistently prevented the fibrotic-like phenotype in assembloids. Specifically, assembloids did not change shape over time compared with controls, which became significantly more compact over time (Fig. 5a–d and Extended Data Figs. 12f–j and 13a–i). Additionally, ITGβ1 knockout or CDH11 knockdown in mesenchymal cells also resulted in a significant reduction in collagen deposition (Fig. 5d and Extended Data Figs. 12j and 13h,i), which could be related to the disruption of the mesenchyme–mesenchyme interactions as monocultures grown at similarly high density deposited large amounts of fibrillar collagen (Extended Data Fig. 13j). None of the other molecules tested resulted in similar rescue (Extended Data Fig. 12e–h).

Fig. 5: Periportal assembloids as tools to investigate disease mechanisms.
figure 5

ad, Short interfering RNA (siRNA)-mediated knockdown experiments in assembloids. Mesenchyme cells (green nuclei and magenta membrane) were transfected with targeting or non-targeting siRNAs before assembly. a, Experimental design, including schematic of the DETECT distance ratio metric (d1/d0). b, Live imaging analysis of assembloids formed with mesenchyme cells transfected with non-targeting (left) or Cdh11 (right) siRNA. Cell boundaries are indicated by SiR-actin. The white dotted line indicates segmentation of the organoid border. Scale bars, 50 µm. c, Segmented assembloids from b were used as input for the DETECT algorithm and to calculate the DETECT distance metric. Violin plots show median and quartiles of the DETECT distance ratio for the non-targeting and Cdh11-knockdown (KD) group. Mann–Whitney test, two-tailed. Dots represent individual assembloids (n = 16 from 2 independent experiments). d, SHG images of assembloids showing fibrillar collagen deposition in non-targeting control and Cdh11-knockdown groups. PDGFRα–H2B–GFP marks portal fibroblasts. n = 2 independent experiments. Scale bars, 25 µm. e, Left, schematic of Mdr2+/+ and Mdr2−/− HepOrgs. Right, immunofluorescence images of HepOrgs derived from wild-type (top) or Mdr2−/− (bottom) livers show dilated bile canaliculi on Mdr2−/− organoids. CD13 (green) marks bile canaliculi. Phalloidin marks cell borders. n = 3 independent experiments with n = 3 biological replicates. Scale bars: 20 µm (main images), 10 µm (zoom views). fh, Chimeric assembloids were formed by Mdr2−/− HepOrgs (bright field, grey), wild-type cholangiocytes (nTom-Chol) and wild-type portal mesenchyme (PDGFRa–H2B–GFP). f, Schematic of experimental design. g, Still images from live imaging experiments (n = 2 independent experiments) of assembloids formed with Mdr2+/+ or Mdr2−/− HepOrgs. Scale bars, 50 µm. h, Change in cholangiocyte numbers between day 0 and day 5 in assembloids formed with Mdr2+/+ or Mdr2−/− HepOrgs. Data are mean ± s.e.m. of 2 biological replicates from n = 2 independent experiments with n = 23 (Mdr2+/+) and n = 22 (Mdr2−/−) assembloids. Dots represent individual assembloids. Mann–Whitney test, two-tailed.

Source Data

Finally, we investigated whether our model would be relevant for studies of other aspects of the pathophysiology of cholestatic biliary fibrosis, such as the cell-autonomous mechanisms of liver fibrogenesis. We utilized a Mdr2−/− mouse model, a knockout mouse that lacks the phospholipid transporter MDR2, resulting in altered bile composition, and models primary sclerosing cholangitis8, a cholangiopathy that develops biliary fibrosis. HepOrgs from MDR2-knockout mice exhibited signs of cholestatic liver disease, with the presence of dilated bile canaliculi, apical bulkheads, inward blebs and accumulation of liver rosettes (Fig. 5e), recognized hallmarks of in vivo cholestasis in mouse and humans46,47,48,49. These features were also observed in Mdr2−/− liver tissue, but were absent in control tissue and HepOrg derived from control wild-type littermates (Fig. 5e and Extended Data Fig. 14a–d). To investigate the effect of Mdr2−/− hepatocytes in the mechanisms of fibrosis, we then mixed the Mdr2−/− cholestatic HepOrgs with wild-type cholangiocytes and wild-type portal mesenchyme to generate chimeric assembloids (Fig. 5f,g). As in the in vivo tissue, both Mdr2−/− and wild-type control assembloids readily connected bile canaliculi with the bile duct (Extended Data Fig. 14e). Notably, assembloids formed by Mdr2−/− hepatocytes, but not wild-type controls, exhibited ductal cell expansion (Fig. 5g,h and Extended Data Fig. 14f), resembling the ductular proliferation observed in vivo in Mdr2−/− mice8. These results suggest that alterations in hepatocyte function contribute to ductular expansion, akin to early stages of biliary fibrosis in vivo.

In summary, we obtained a periportal assembloid model that recapitulates aspects of biliary fibrosis and cholestatic liver disease and is amenable for investigation of cell-autonomous and non-autonomous mechanisms in liver disease.

Discussion

The study of the cellular mechanisms that regulate cholestatic liver injury and biliary fibrosis has been a major challenge, owing to the difficulty of modelling physiological bile canaliculi in vitro and the shortfall of cellular systems that recapitulate the architecture and cellular interactions between different cells in the periportal region.

Here, we generated a periportal assembloid model that combines portal mesenchyme, cholangiocytes and hepatocytes, and readily recapitulates the cellular and mesoscale architecture of the periportal region of the mouse liver, albeit lacking the portal endothelium and resident immune cells. By generating HepOrgs that retain apical polarity, we obtained a model that forms physiological and functional bile canaliculi 3D network. Notably, when HepOrgs are generated from the cholestatic Mdr2−/− mouse model, they faithfully recapitulate the specific histological features of cholestatic liver disease, including liver rosettes and bile canaliculi bulkheads8,46. This is in stark contrast to previous models, which were cholestatic at baseline (presented bile canaliculi bulkheads)46 and lacked the 3D network organization typical of transport networks50.

The physiological bile canaliculi architecture generated by HepOrgs enabled us to reconstruct a physiological and functional connection between the bile canaliculi and bile duct. A limitation of this study was that it did not achieve full zonation as in the liver lobule—as expected, given that the organoids are too small in size to provide a full liver axis.

Paradoxically, although recapitulating native tissue architecture is essential, too much complexity could obstruct investigations of the precise role of specific niche cell types in the physiology or pathophysiology of a tissue without introducing confounding factors. Our system represents a modular and tractable in vitro tool for investigating the dynamics of cholestatic biliary fibrosis and the contribution of different cell types. Notably, this is not possible in current epithelial-only organoid models or in organoids derived from induced pluripotent stem cells, in which the different populations co-develop from a single clone. By targeting CDH11 or ITGβ1 specifically in mesenchyme, we provide proof of the concept that assembloid cells can be used as platform for mechanistic discovery and to investigate cell-autonomous mechanisms in liver disease. How this affects the exact intracellular molecular effectors in mesenchyme–ECM or mesenchyme–mesenchyme interactions that regulate hepatocyte cell death and duct cell expansion remains unknown.

In conclusion, our periportal assembloid model recapitulates architecture and cell–cell interactions of the liver at the meso- and cellular scale and provides an in vitro liver organoid system for the study of bile canaliculi formation, bile drainage and cell-autonomous or cell-specific contributions of hepatocytes, cholangiocytes and portal mesenchyme to cholestatic liver disease.

Methods

Mouse models

Mouse experiments were performed in accordance with the German animal welfare legislation and in strict pathogen‐free conditions in the animal facility of the MPI‐CBG. Protocols were approved by the Institutional Animal Welfare Officer (Tierschutzbeauftragter), and all necessary licenses were obtained from the regional Ethical Commission for Animal Experimentation of Dresden, Germany (Tierversuchskommission, Landesdirektion Dresden, number DD24-5131/346/3; TVT08/2023; TVV42/2021; TVV49/2021). The laboratory animal housing of the MPI-CBG is exclusively barrier housing. All mice are kept in individually ventilated cages under a 12 h:12 h light:dark cycle. The animal room temperature is maintained between 20 and 24 °C and the relative humidity is 55 ± 10%. Both are subject to constant monitoring. Sterile food and water were given ad libitum. Healthy adult mice (8–25 weeks of age) of both sexes were used for experiments. For MDR2 experiments, mice were used at 8 weeks of age. Wild-type C57/Bl6 mice, Rosa26-mTmG, Rosa26-nTnG, PdgfraH2B-GFP, PdgfraH2B-GFP × Rosa26-mTmG, Prom1creERT2 × R26-LSL-ZsGreen, Itgb1fl/fl × R26-LSL-ZsGreen or Mdr2-knockout mice were used for experiments. Rosa26-mTmG [Gt(ROSA)26Sortm4(ACTB-tdTomato,-EGFP)Luo/J] and Rosa26-nTnG [B6;129S6-Gt(ROSA)26Sortm1(CAG-tdTomato*,-EGFP*)Ees/J] were obtained from the Jackson Laboratory (JAX). The PdgfraH2B-GFP ([B6.129S4-Pdgfratm11(EGFP)Sor/J] mice were described previously51 and obtained from M. Zernicka-Goetz. The PdgfraH2B-GFP × Rosa26-mTmG was generated by crossing the PdgfraH2B-GFP mice with Rosa26-mTmG were obtained and described above. The Mdr2-knockout line [FVB.129P2-Abcb4tm1Bor/J] was described before8. The R26-LSL-ZsGreen B6.Cg-Gt(ROSA)26Sortm6(CAG-ZsGreen1)Hze/J) mice were obtained from JAX. Itgb1fl/fl (B6;129-Itgb1tm1Efu/J) was described previously52, and obtained from M. Zernicka-Goetz. Itgb1fl/fl × R26-ZsGreen was generated by crossing Itgb1fl/fl mice with R26-LSL-ZsGreen mice. The Prom1creERT2 × R26-LSL-ZsGreen mice were generated by crossing Prom1creERT2 (B6N;129S-Prom1tm1(cre/ERT2)Gilb/J) mice53, obtained from R. Gilbertson, with R26-LSL-ZsGreen mice. Confetti mouse (Gt(ROSA)26Sortm1(CAG-Brainbow2.1)Cle/J) has been described54 and was recombined in vitro to obtain mCFP cholangiocyte organoids. Mice were bred onto a C57/B6 background.

HepOrg culture

Primary hepatocytes were isolated from mice following either euthanasia by cervical dislocation, or anaesthesia by intraperitoneal injection of 90 mg kg−1 bodyweight ketamine and 10 mg kg−1 Rompun (xylazine) as previously described55. When isolating hepatocytes from Mdr2−/− fibrotic livers, collagenase perfusion was longer compared to standard protocol, between 25–30 min. After isolation, hepatocytes were counted, and 5,000 viable hepatocyte cells were resuspended in 5 μl of suspension buffer and mixed with Matrigel to a total of 25 μl and seeded forming a Matrigel dome as for 25 μl Matrigel per well of a 48-well plate. Following Matrigel solidification (~15 min), 250 μl of medium per 48-well plate was overlaid. Hepatocytes were cultured in an adapted medium as described25: AdDMEM/F12 (ThermoFisher, 12634010) medium containing 1% HEPES (ThermoFisher, 15630-056), 1% penicillin/streptomycin (ThermoFisher, 15140-122), Glutamax (ThermoFisher, 35050-068), 1× B27 (Invitrogen, 12587010) and 1.25 mM N-acetylcysteine (Merck/Sigma, A9165) –referred to as basal medium, which was further supplemented with 10 nM gastrin (Merck/Sigma, G9145), 50 ng ml−1 mEGF (ThermoFisher, PMG8043), 15% RSPO1 conditioned medium (homemade), 100 ng ml−1 FGF10 (Peprotech, 100-2), 50 ng ml−1 FGF7 (Peprotech, 100-19-50), 10 mM nicotinamide (Merck/Sigma, N0636), 25 ng ml−1 HGF (Peprotech, 100-39), 3 μM CHIR9902 (Tocris, 4423), 1 μM A83-01 (Tocris, 2939) and 10 µM ROCK inhibitor (Y-27632, Merck/Sigma, Y0503), supplemented with 30% WNT3a conditioned medium (Wnt-CM) (homemade) –referred to as hepatocyte medium + 30% Wnt-CM (HM-Wnt), or Wnt surrogate (HM-WntS) (WNT Surrogate: Live Science Incubator, N001) or 3% foetal bovine serum (HM-FBS) (FBS, Merck/Sigma, F7524), or in medium described in Hu et al.15 with modified FGF10 concentration (100 ng ml−1)—or in medium described in Peng et al.16, containing TNF (Peprotech, 300-01 A) with the modification that the basal medium was Advanced DMEM/F12 and EGF and HGF concentrations were 25 ng ml−1. HepOrgs were cultured at 37 °C and 5% CO2. The medium was changed 2–3 times per week. To enrich for bubbly/grape-like HepOrg structures, the initial cell preparation was enriched with cell clusters, or alternatively, ball HepOrgs were removed by hand-picking or by centrifugation at passage 1 and 2. To enrich for ball-like structures, cells were seeded as single cells at lower density (1,000 cells) to avoid fusion and promote single cells growing out in the aberrant morphology. Sometimes unwanted cholangiocyte organoids or mesenchyme was also observed in the cultures until passage 2, and also removed by hand-picking from the hepatocyte cultures. HepOrg cultures were regularly tested for the absence of mycoplasma using MycoAlert Mycoplasma Detection Kit (Lonza LT07-118).

For co-culture media test experiments, HepOrg grown in HM-Wnt up to passage 2 (P2) were cultured in one of the following media for 6–14 days, with a medium change every 2–3 days. Organoids were transferred to indicated medium from HM-Wnt medium, and then collected for analysis (7 days, 14 days) or immunofluorescence staining (6–7 days). Media regimes used were: (1) basal medium supplemented with Wnt-CM (30%) and 10 µM ROCK inhibitor (Y-27632, Merck/Sigma, Y0503) (MM)20; (2) and our previously published medium that supports hepatocyte differentiation from cholangiocytes with addition of small-molecule Wnt inhibitors21, differentiation medium (DM) composed of basal medium supplemented with 10 nM gastrin (Merck/Sigma, G9145), 50 ng ml−1 mEGF (ThermoFisher, PMG8043), 100 ng ml−1 FGF19 (Peprotech, 100-32-25), 25 ng ml−1 HGF (Peprotech, 100-39), 3 μM CHIR9902 (Tocris, 4423), 0.5 μM A83-01 (Tocris, 2939), 25 ng ml−1 hBMP7 (Peprotech, 120-03), 10 μM DAPT (Merck/Sigma, D5942-5MG), 3 μM IWP2 (Merck/Sigma, I0536-5mg), 25 μM iCRT3 (Merck/Sigma, SML0211-5MG) and 3 μM dexamethasone (Tocris, 4489).

For organoid formation efficiency, primary hepatocytes were isolated as described above. To prevent organoids from fusing, 1,000 viable hepatocytes (viability >80%) were plated in 25 μl Matrigel (BD Bioscience; 356231) droplet and cultured as described above. After 9 days, organoid numbers were counted and results expressed as a percentage relative to the initial seeding cell numbers.

To determine cholangiocyte contamination, Prom1creERT2 × R26-LSL-ZsGreen transgenic mouse were used to induce specific cholangiocyte labelling with ZsGreen protein in the adult mouse prior to isolation of hepatocytes. Following tamoxifen injection, cholangiocytes were labelled by fluorescent protein ZsGreen. This process has been shown to occur with close to 80% efficiency53,56. We then allowed for a tamoxifen wash-out period of 14 days, to exclude any effects relating to the injection and tamoxifen in murine system, and followed by standard hepatocyte isolation, as described in methods. For the analysis of cholangiocytes from hepatocyte isolation and HepOrg culture, primary hepatocytes and other liver cells were isolated from mice as described above. The cells were then strained with 100 μM strainer, washed one time with AdDMEM/F12 (ThermoFisher, 12634010) medium containing 1% HEPES (ThermoFisher, 15630-056), 1% penicillin/streptomycin (ThermoFisher, 15140-122), Glutamax (ThermoFisher, 35050-068), spun 5 min at 100g and stained 30 min with EpCAM antibody conjugated to APC (CD326 (EpCAM) Monoclonal Antibody (G8.8), APC, eBioscience, ThermoFisher, 17-5791-80). Before sorting, cells were washed one time with above medium, spun 5 min at 100g and resuspended in the same medium for sorting. In case of cholangiocytes from HepOrg culture, all cells from the culture were collected in above media, spun 5 min at 200g and dissociated by incubation with TrypLE Express (ThermoFisher, 12605010) for 5 min at 37 °C, before being strained and stained as outlined above.

Liver ductal and mesenchymal cell isolation and sorting

Mouse livers were collected and enzymatically digested as described14,57,58. In brief, minced livers were incubated in a solution containing 0.0125% collagenase (Merck/Sigma, C9407), 0.0125% dispase II (ThermoFisher, 17105-041) and 1% foetal bovine serum (FBS) (Merck/Sigma, F7524) in DMEM/Glutamax (ThermoFisher, 31966-021) supplemented with 1% HEPES (ThermoFisher, 15630-056) and 1% penicillin/streptomycin (ThermoFisher, 15140-122) and 0.1 mg ml−1 of DNAase (Merck/Sigma, DN25) in a shaker at 37 °C and 150 rpm for 1.5–3 h. The biliary tree fragments and associated stroma were then dissociated into single cells with TrypLE diluted to 5× (Gibco, A12177-01). Following dissociation, cells were isolated by fluorescence-activated cell sorting (FACS) using the described approach20,58. Single cells were incubated with fluorophore-conjugated antibodies for 30 min and FACS-sorted using BD FACS Aria (BD Biosciences) or SH800S (SONY) cell sorters. Cells were sequentially gated based on size and granularity (forward scatter (FSC) versus side scatter (SSC)) and singlets (FSC area versus FSC height); after which ductal cells were selected based on EPCAM positivity and negative exclusion of the haematopoietic/endothelial markers CD31, CD45 and CD11b. The mesenchyme was sorted as double positive PDGFRα–GFP+SCA1+ cells gated from the EpCAMCD31CD45CD11b fraction. Following FACS isolation, ductal cells and portal mesenchymal cells were used for ductal organoid and mesenchymal cell culture, respectively. Cholangiocyte organoid and mesenchymal cell cultures were regularly tested for the absence of mycoplasma using MycoAlert Mycoplasma Detection Kit (Lonza LT07-118).

Mesenchymal cell culture

Mesenchymal cells were cultured in the basal medium described above supplemented with Wnt-CM (30%) and 10 μM ROCK inhibitor (Y-27632, Merck/Sigma, Y0503), as described20. Cells were passaged at 1:3 and 1:2 ratios, through enzymatic digestion using TrypLE Express (ThermoFisher, 12605010) for 5 min at 37 °C. Mesenchymal cells were cultured at 37 °C and 5% CO2. For 3D culture in Matrigel, mesenchyme cells were seeded in Matrigel at density 2,500 cells per 25 μl Matrigel in a 48-well plate as the sparse condition, overlaid with medium. Alternatively, for the confluent/aggregated 3D condition, 2,500 mesenchyme cells were placed in a well of a low-attachment 24-well plate in 0.5 ml of medium supplemented with 1× methylcellulose (Sigma, M6385) and the plate was transferred to a rocker-shaker (Biosan, MR-1), incubated at 10 rpm for 18–24 h at 37 °C and 5% CO2. Mesenchyme aggregates were collected into 1.5 ml Eppendorf tube, spun down for 5 min at 200g and seeded in Matrigel as described above and overlaid with medium.

Hepatocyte 2D sandwich culture

Primary hepatocytes were isolated from mouse livers via collagenase perfusion as described above. Cells were plated onto collagen (0.9 mg ml−1) coated 24-well plates at 200,000 cells per well in Williams E medium (PAN Biotech), substituted with 10% FBS, 100 nM dexamethasone and penicillin/streptomycin and maintained at 37 °C in an atmosphere with 5% CO2. After 3–4 h of attachment, cultures were washed with phosphate buffer saline (PBS) and coated with a second layer of collagen (0.6 mg ml−1) to obtain a sandwich culture as previously described59. Medium was changed every day. For immunofluorescence analysis, cells were seeded in collagen‐coated glass cover slips and cultured as above. When required, cells were fixed in 4% paraformaldehyde at room temperature for 30 min, washed twice with PBS, permeabilized for one hour with 0.1% Triton X-100, washed and blocked in 10% horse serum for 2 h. Holes were applied to the top layer collagen using fine aspiration to ensure better antibody penetration. Cells were incubated with primary antibodies at room temperature overnight, washed for 2 h in wash buffer (300 mM NaCl, 0.1% Tween, 10 nM Tris/HCl) with frequent (8–12×) exchanges of buffer. Secondary antibodies and dyes were incubated for 5 h at 37 °C in a humidified chamber. Thereafter cells were washed extensively and mounted onto glass slides using 0.1 g ml−1 Mowiol (Calbiochem).

Itgb1-knockout induction in mesenchyme cell culture

To generate Itgb1-knockout mesenchyme, the mesenchymal cells were isolated as specified above from Itgb1-fl/fl (Itgb1tm1Ref) R26-ZsGreen transgenic mice, and sorted as positive SCA1+ cells gated from the EpCAMCD31CD45CD11b fraction. The cells were then expanded as specified above in 2D culture until passage 1, when the Itgb1fl/fl × R26-ZsGreen allele was recombined by transducing the cells using a adenovirus (Ad5-CMV-Cre; University of Iowa) at a multiplicity of infection of 10. Medium was changed up to 24 h after infection. Following culture and expansion, the cells were then FACS-sorted gated on GFP+ to enrich for the recombined cells, and plated again to passage 2. Cells were used for experiments at passage 2.

Cholangiocyte organoid culture

Cholangiocyte organoids were generated and cultured as described14. In brief, sorted EpCAM+ cholangiocytes were embedded in 50 μl Matrigel per well of a 24-well plate and cultured in EM medium (AdDMEM/F12 medium (ThermoFisher, 12634010) containing 1% HEPES, 1% penicillin/streptomycin, Glutamax, 1× B27 and 1.25 mM N-acetylcysteine supplemented with 10 nM gastrin (Merck/Sigma, G9145), 50 ng ml−1 mEGF (ThermoFisher, PMG8043), 5% RSPO1 conditioned medium (home made), 100 ng ml−1 FGF10 (Peprotech, 100-26), 10 mM nicotinamide (Merck/Sigma, N0636) and 50 ng ml−1 HGF (Peprotech, 100-39)) supplemented with 30% WNT3a conditioned medium (Wnt-CM) (homemade), 25 ng ml−1 Noggin (Peprotech, 120-10C) and 10 μM ROCK inhibitor (Y-27632, Merck/Sigma, Y0503). Then, after the first 3 days in culture, cells were switched to EM medium only (without Wnt-CM, ROCK inhibitor and Noggin). The grown cholangiocyte organoids were passaged at a 1:3 ratio once a week by mechanical dissociation, re-embedded in fresh Matrigel and cultured in EM as described in57. Cholangiocyte organoids were cultured at 37 °C and 5% CO2.

Periportal assembloid generation

For periportal assembloids generation, portal mesenchyme, cholangiocyte cells from cholangiocyte organoids and HepOrgs were aggregated in 24-well plates in rocking platform or in 24-well AggreWell plates, and mixed at different ratios according to the purpose. To define the numbers of the different cell types needed, we took advantage of our previous studies on the homeostatic proportion of hepatocytes and cholangiocytes (97% versus 3%)25, as well as of portal mesenchymal: ductal cells (3:10 ratio20). The use of cells labelled with different endogenous fluorescent proteins ensured that the structures containing three cell types had originated following the aggregation of the different cells. For healthy homeostasis ratios: 250 mesenchyme cells, 1,000 cholangiocyte cells and 10 HepOrgs were used per well. For non-physiological (fibrotic-like) ratio 2,500 mesenchyme cells, 1,000 cholangiocyte cells and 10 HepOrgs were used.

The cells were prepared as follows: cholangiocyte organoids grown in EM (passage 2–9) were dissociated to single cells by collecting them from Matrigel using cold AdDMEM/F12 (ThermoFisher, 12634010) containing 1% HEPES (ThermoFisher, 15630-056), 1% penicillin/streptomycin (ThermoFisher, 15140-122) and 1% Glutamax (ThermoFisher, 35050-068) and dissociating them into single cells using TrypLE 1× (ThermoFisher, 12605010) for 7 min at 37 °C and filtered through 40-μm cell strainers. In parallel, 80–90% confluent mesenchyme cultures (fresh from sorting or passage 0–2, as specified in legend) grown as specified above, were washed with PBS and dissociated to single cells by incubating with TrypLE 1× for 5 min at 37 °C. Both single cells suspensions were spun at 300g for 5 min and the cell concentration was determined by manual counting in haemocytometer. In parallel, HepOrgs were grown in HM-Wnt for at least 1 or 2 passages and cultured (or not) with MM for 48 h before assembling (as specified in the figure legend). Then, HepOrgs were removed from Matrigel using 2 washes with cold AdDMEM/F12 (ThermoFisher, 12634010) supplemented with 1% HEPES, 1% penicillin/streptomycin and 1% Glutamax, and incubated with cold Cell Recovery Solution (Corning, 354253) or cold PBS for 10 min on ice. Then, bubbly/grape-like shape organoids were hand-picked on a stereoscope and placed in the aggregation plate as described below. The efficiency of aggregation was increased when using HepOrgs that had been pre-conditioned for 48 h with MM (Extended Data Fig. 7c).

For rocking platform aggregation, cells were mixed together in a well of a low-attachment 24-well plate, in 0.5 ml MM, which in some cases, was supplemented with 1× methylcellulose (Sigma, M6385) to facilitate aggregation. Then, the plate was transferred to a rocker-shaker (Biosan, MR-1) and incubated at 10 rpm for 14–24 h at 37 °C and 5% CO2. Following aggregation, assembloids were collected into 1.5 ml Eppendorf tube, spun down for 5 min at 200g and seeded in Matrigel as described below. For AggreWell aggregation, cells and organoids were mixed in specified ratios in 1.5 ml MM in AggreWell plates (AggreWell800, Stem Cell Technologies, 34811, pre-treated as recommended by the manufacturer), spun down 5 min at 100g and incubated for 14–24 h at 37 °C and 5% CO2. To collect the structures from AggreWell wells, the solution was disrupted by pipetting with 1 ml pipette, and all the solution was collected on a 40-μm strainer. The strainer was then placed upside down over a 6-well plate, washed with AdDMEM/F12 (ThermoFisher, 12634010) supplemented with HEPES, penicillin/streptomycin and Glutamax, and all suspension was collected and spun down for 5 min at 200g. The supernatant was removed and the structures seeded in Matrigel. Structures from both aggregation types were seeded either in 25 μl Matrigel dome in a pre-warmed 48-well plate, or in a Matrigel layer (20 μl Matrigel in a 96-well plate centrifuged in a cold centrifuge 200g for 5 min, which is then overlaid with structures in another 20 μl Matrigel on top), solidified 10 min in 37 °C incubator and overlaid with further 150–200 μl MM. The structures were grown for 7–14 days with media changes to fresh MM every 2–3 days. Unless specified in the figure legend, we opted for shaker aggregation as, in AggreWell, some assembloids tended to acquire a ball-shape structure compared to rocking platform/shaker and in addition, it did not require any specialized equipment.

Generally, for periportal assembloid generation, HepOrgs derived from wild-type mouse livers, cholangiocyte cells from cholangiocyte organoids from nuc-Tdtom mouse livers and portal mesenchymal cells from PDGFRα–H2B–GFP+SCA1+ cells were assembled either at a normal physiological ratio (10 HepOrgs: 1,000 cholagiocyte cells: 250 mesenchyme cells) or at a ratio with 10× excess mesenchyme cells, unless stated otherwise in the figure for other fluorescent colour combinations.

A step-by-step protocol for mouse periportal assembloid generation can be found on protocols.io60.

Blocking antibody treatment of assembloids

To investigate whether assembloids could be used to study cellular interactions in fibrotic-like phenotype, blocking antibodies were added to the assembloid culture at the time of assembly (rocking platform). The list of compounds and antibodies is presented in Supplementary Table 1 with relevant concentrations tested.

Assembloid experiments with siRNA-treated mesenchyme cells

Passage 2 portal fibroblasts derived from PDGFRα-H2B-GFP × Rosa26-mTmG mouse were detached using 1:1 mixture of 10X TrypLE and Accutase (StemPRO Accutase, ThermoFisher A11105015). Single cell suspensions were spun at 300g for 5 min. Cells were seeded at either 5,000 cells per well or 10,000 cells per well to be subsequently used for assembloid formation or RT–qPCR analysis to determine knockdown efficiency. Then, portal fibroblasts were transfected with 20 pmol of a pool of 4 ON-Targetplus siRNA targeting specific genes (Supplementary Table 1) for 3 h according to manufacturer instructions. After 16–24 h transfected cells were collected using pre-warmed digestion mix of Accutase and TrypLE select Enzyme (10X) at 1:1 ratio, and incubated 5 min at 37 °C. Single cells suspensions were spun at 300g and resuspended in 100 μl MM. mCFP cholangiocyte organoids were expanded in our standard cholangiocyte medium and processed to single cells as described above. To make fibrotic-like assembloids, 2,500 mesenchyme cells, 1,000 single-cell cholangiocytes and 10 HepOrgs were combined and incubated 14–24 h on a rocking platform at 37 °C as described above. Formed assembloids were hand-picked and seeded on 10 µl Matrigel pre-coated in a 96-well plate (Greiner, 655090) and incubated 15 minutes at 37 °C, following by overlay with 10 µl of Matrigel. After 30 minutes incubation at 37 °C, 200 µl MM supplemented with 0.5 μM of SiR-actin (Spirochrome, SC001) was added. Imaging started 2 h later and was performed every 24 h for 2 days on CellVoyager CV7000 spinning disc microscope with 20× objective, with z-step size 1 μm. Lasers with 405, 488, 561 and 647 nm were used to detect the mCFP, nGFP, TdTomato and SiR-actin signal, respectively, on a sCMOS camera. To determine knockdown efficiency, total RNA was extracted from cells after 16 h of siRNA incubation as described above using the Arcturus PicoPure RNA Isolation Kit (Applied Biosystems, 12204-01) according to the manufacturer’s protocol; including a 15-min digestion step with DNAse.

To quantify the number of mesenchyme cells per assembloid in the siRNA experiments, the PDGFRα–H2B–GFP signal was segmented as follows: First a max projection was used to reduce the 3D stack to a 2D image, then a log transformation was applied to compress the high dynamic range of the signal. The image was then down-sampled by a factor of four. Subsequently, the StarDist61 algorithm, using the pretrained 2D_versatile_fluo model, was used for segmentation. Although this method effectively segmented mesenchyme nuclei, it also included other structures, such as hepatocyte nuclei, which exhibited lower intensity. To ensure accurate counting, only segments with an average intensity above a manually selected threshold were included.

HepOrg formation and assembloid morphology imaging

Freshly isolated hepatocytes were seeded in a 96-well plate (Greiner, 655090) in 7 μl Matrigel (BD Bioscience; 356231) droplet in the concentrations of 40–800 cells per μl, of Matrigel, supplemented with HM-Wnt. For imaging, 2 × 2 tile z-stack images (10 μm z-step size) of each well were acquired every 24 h for 13 days on a CellVoyager CV7000 spinning disc microscope with 10× objective (Yokogawa). Membranes of HepOrgs were detected by endogenous membrane-tdTomato signal and used to determine ball or bubbly/grape-like shape of the structures. Ball structures are defined with round edges resembling a sphere, while bubbly/grape- like shape is determined by the appearance of irregular surfaces and surface invaginations. Endogenous tdTomato signal was excited with 561 nm laser. Maximum-intensity images were made using ‘Macro CV7000 z-projection’. For each well, time-lapse maximum-intensity projection videos were made using either stacklist files that contained information about every time point, z-stacks and channels (Supplementary Methods 1, appendix A, Method_section_FIJI_stack_files_KNIME). Stack lists were converted into time-lapse videos using the Fiji macro Macro_make_HyperStack_Zmax_005.ijm. For quantification, 30 randomly assigned ball or bubbly/grape-like HepOrgs were traced back to the seeding time point (day 0), and the number of cells of origin (one cell or cell cluster) was quantified.

For DETECT analysis in HepOrgs or in assembloids (details of the analysis below), maximum-intensity projections from the SiR-actin channel or bright-field channel for each time point were used to make XY coordinates of the assembloids outlines. In short, outlines of the assembloids were created in Fiji using the polygon selection tool in the clockwise orientation. xy coordinates were saved using the ‘save xy coordinates’ command as a .txt file (as described in Supplementary Methods 1, appendix A).

Organoid whole-mount staining and imaging

For in vitro staining, organoids and assembloids were first extracted from Matrigel with ice-cold cell recovery solution (Corning, 354253) or PBS, and then fixed with 4% paraformaldehyde (PFA) in PBS for 30 min on ice. Blocking and permeabilization was performed for 1 h at room temperature in PBS containing 0.2% Triton X-100 and 2% BSA. The samples were incubated with primary antibodies overnight at 4 °C in blocking solution. Following 3 washes with PBS, the samples were incubated overnight at 4 °C or for 4–8 h at room temperature with secondary antibodies, phalloidin and DAPI in PBS. The samples were washed 3 times with PBS and subsequently cleared using fructose-glycerol clearing solution (25 ml glycerol, 5.3 ml dH2O and 22.5 g fructose–60% glycerol and 2.5 M fructose)62. Alternatively, organoids and assembloids were permeabilized with 0.5–1% Triton X-100 in 1× PBS for 1 h. After, they were incubated in primary antibody in TxBuffer (0.2% gelatin 2% gelatin, 300 mM NaCl and 0.3% Triton X-100 in 1× PBS) incubated at 4 °C overnight. Following 3 washes with PBS, the samples were incubated for 2 h at room temperature with secondary antibodies in TxBuffer. The samples were washed three times with PBS and left in PBS until imaging or cleared as described above. The full list of primary and secondary antibodies used is specified in Supplementary Table 1.

Thin section staining

For thin tissue sections (8–12 μm) and staining, livers were fixed for 2 h or overnight in 10% formalin with rolling at 4 °C and tissues incubated with 15% sucrose for 1 h, and then 30% sucrose PBS for 24-48 h, embedded into cryomolds (Sakura, 4566) with OCT compound (VWR, 361603E) and snap-frozen. Tissue blocks were cryo-sectioned on ThermoScientific CryoStar NX70 cryostat. Sections were blocked in PBS with 2% DS and 1% BSA for 2 h at room temperature, incubated with primary antibodies in 1/100-diluted blocking buffer overnight at 4 °C and with secondary antibodies + DAPI for 2 h at room temperature in 0.05% BSA PBS. Sections were mounted in Vectashield. The list of used antibodies is available in Supplementary Table 1

Thick tissue section staining

For thick tissue sections, mice were perfused at 3.7 ml min−1 for 10–15 min with 4% paraformaldehyde, 0.1% Tween-20 in PBS. Livers were cut in smaller pieces and post-fixed in the same solution for 24 h on a rotator at 4 °C. After, liver pieces were washed in PBS to remove fixative. For storage, liver pieces were kept in PBS at 4 C. For sectioning, livers were mounted in 4% low-melting agarose in PBS and cut into 100 μm-thick sections on a vibratome (Leica VT1200S). For deep tissue imaging, tissue sections were permeabilized with 0.5% Triton X-100 in PBS for 1 h at room temperature. The primary antibodies were diluted in Tx buffer (0.2% gelatin, 300 mM NaCl, and 0.3% Triton X-100 in PBS) and incubated for 48 h at room temperature. After washing 5× 15 min with 0.3% Triton X-100 in PBS, the sections were incubated with secondary antibodies, DAPI (1 mg ml−1; 1:1,000) and phalloidin–Alexa Fluor 488 or 647 (Thermo Fisher Scientific; A12379 or A22287; 1:250) for another 48 h. The list of used antibodies is available in Supplementary Table 1. After washing 5× 15 min with 0.3% Triton X-100 in PBS and 3× 1 min with PBS, the optical clearing started by incubating the slices in 25% fructose for 4 h, continued in 50% fructose for 4 h, 75% fructose overnight, 100% fructose (100% wt/vol fructose, 0.5% 1-thioglycerol, and 0.1 M phosphate buffer, pH 7.5) for 6 h, and finally overnight in SeeDB solution (80.2% wt/wt fructose, 0.5% 1-thioglycerol and 0.1 M phosphate buffer)63. The samples were mounted and imaged in SeeDB.

RNAscope

For RNAscope, HepOrgs were embedded into cryomolds (Sakura, 4566) with OCT compound (VWR, 361603E) and snap-frozen after fixing in 4% PFA/PBS for 30 min on ice. Tissue blocks were cryo-sectioned on ThermoScientific CryoStar NX70 cryostat (12 μm sections). The sections were stained using RNAscope Fluorescent Multiplex and RNAscope Multiplex Fluorescent V2 (Advanced Cell Diagnostics, 323100) according to the manufacturer’s instructions. Probes for the target genes are in Supplementary Table 1.

Imaging of HepOrgs, assembloids and liver tissue

For thin tissue sections (8–12 μm), RNA scope sections (12 μm) and whole-mount imaging of assembloids, images were acquired using a single photon point-scanning confocal system (Zeiss LSM 880 Inverted or Upright), with a Quasar detector with 32 spectral detection channels in the detection channels in a gallium arsenide phosphide (GaAsP) detector with 2 photomultiplier tubes and transmitted light detector. Images were acquired using a Zeiss 20× (0.8 NA) air objective or Zeiss LD LCI Plan-Apochromat 40×/1.2 DIC Imm Corr M27 immersion corrected objective. Fluorophores were excited with 405, 458, 488, 532, 561, 594, and 633 nm lasers. Images were processed using ZEN software (Zeiss), or ImageJ/Fiji.

For 3D reconstruction of bile canaliculi in organoids and assembloids, images of optically cleared organoids and assembloids were acquired with an inverted multiphoton laser-scanning microscope (Zeiss LSM 780 NLO) or a single photon point-scanning confocal system (Zeiss LSM 880 Inverted) with a Quasar detector with 32 spectral detection channels in the GaAsP detector with 2 photomultiplier tubes (Zeiss LSM 880). Images were acquired using a Zeiss LD LCI Plan-Apochromat 40×/1.2 DIC Imm Corr M27 immersion corrected objective, or a Zeiss LD LCI Plan-Apochromat 63×/1.2 DIC Imm Corr M27, with a voxel size 0.3 μm. Fluorophores were excited with 405, 488, 561, 594, and 633 nm laser lines and detected with GaAsp detectors.

Optically cleared 100-μm liver sections were imaged with an upright multiphoton laser-scanning microscope (Zeiss LSM 780 NLO) equipped with two photomultiplier tubes and one 32-channel GaAsP detector for spectral detection in the scanhead, four GaAsP non-descanned detectors for multiphoton detection, three transmitted light detectors. Liver slices were imaged twice, at low (Zeiss Plan Apo 10×/0.45 NA Air) and high resolution (63×/1.3, or 40×/1.2 Zeiss LD LCI Plan-Apochromat DIC immersion corrected objective; 0.15 or 0.3 μm voxel size), respectively. Low-resolution overviews of the large surface of liver sections were created and used to find the central and portal vein regions, and image bile canaliculi of a dedicated region. Selected regions (300 μm × 300 μm × 100 μm; x, y, z) were then acquired at high resolution (0.3 μm voxel size).

For 3D visualization of bile canaliculi, bile ducts and hepatocytes, high-resolution images were processed and segmented, based on CD13, PCK and phalloidin staining, respectively, using Motion Tracking software (http://motiontracking.mpi-cbg.de) as described31.

High-resolution imaging with Airyscan technology

For imaging detail (for example, bile canaliculi, polarity) of liver tissue, HepOrg, and assembloids with high-resolution, Airyscan images were acquired on an inverted single photon point-scanning confocal system (Zeiss Celldiscoverer 7 with LSM 900 and Airyscan 2) using a Plan-APOCHROMAT 20×/0.95 Autocorr, Air (Zeiss), with a 1× Tubelens, and a voxel size 0.082 × 0.082 × 0.340 μm, with an image size of 179.87 × 179.87 μm. Fluorophores were excited with 405, 488, 561 and 640 nm (T10/R90) laser lines and detected with GaAsP-photomultiplier tube detectors.

Image analysis

For quantification of organoid and assembloid morphology, nuclei and cells, custom-made pipelines in Arivis 4D software (Zeiss) were used. For organoid morphology, segmentation was based on membrane staining of organoids. At first, a median denoising filter was applied followed by setting an intensity threshold for segmentation which was determined manually depending on membrane signal intensity. Subsequently, the morphological operations ‘inclusion filling’ and ‘close objects’ were applied. At last, segmented objects were filtered by size to match the expected morphology of the organoids of interest. In case of incomplete organoid segmentation caused by weak fluorescence signal, missing segmentation was added manually.

For cell shape, segmentation was based on membrane staining. At first, a discrete gaussian denoising filter was applied followed by two top-hat filters with specific radii. Cells were segmented using minimal intensity threshold, split sensitivity and maximal area to match the expected morphology of the cells of interest in the fluorescence images. For nuclei, segmentation was based on nuclei staining or fluorescently tagged nuclei. At first, a discrete gaussian filter was applied followed by two top-hat filters with two specific radii. Further, median and particle enhancement filters were applied.

Nuclei were segmented using diameter, probability threshold, and split sensitivity to match the expected morphology of the nuclei in the fluorescence images. In case of incomplete nuclei segmentation caused by weak fluorescence signal, missing nuclei were added manually. To determine co-localized objects, an Arivis pipeline was applied in which segmented objects of interest were imported and objects inside or intersecting with the chosen compartment by at least one voxel were obtained. This method was applied to determine the number of nuclei in cells, the number of cells in organoids and the number of co-localized nuclei. All segmentation was checked manually and corrected manually, whenever needed.

For quantification of fibrillar collagen (SHG signal) in the assembloids, max projection images of the SHG channel were used to segment the area of the fibrillar collagen by using manually selected threshold in Fiji (min value 60, max value 110). To avoid unspecific signal outside of assembloids only SHG segmented area inside of the assembloid was used for quantification. Assembloid area was segmented using the free-hand selection tool to outline the boundaries of the assembloids in the SiR-actin channel. For quantification of fibrillar collagen signal in healthy and fibrotic-like assembloids without siRNA treatment, or in Itgb1-knockout mesenchymal cells and their matched control assembloids, or in mesenchyme single 3D and 2D culture, whole frame or organoid intensity of SHG channel was measured for the sample and control images, matched between experiments, using the ‘Integrated density’ measurement function in Fiji, and displayed as RawIntDen (the sum of the values of the pixels in the image or selection) or IntDen (the product of area and mean grey value).

Quantification of organoid shape changes over time

To quantify differences between two phenotypes, we used topological data analysis. Specifically, Marsh et al.28 devised a temporal shape descriptor algorithm, DETECT, which extends the smooth Euler characteristic transform64. The DETECT algorithm processes shapes by generating a curve that captures both the geometric and topological characteristics of the shape, along with its morphological changes over time, while also being rotationally invariant. For this study, the inputs to the algorithm are the boundaries of 2D projections of each organoid at each time point, generated as described above in ‘Organoid formation and assembloid morphology’, and we calculate the corresponding DETECT curve (Supplementary Methods 2, appendix B). The collection of curves at all time points measures how the morphology of a particular organoid changes over time. The new metric DETECT metric developed here (described in Supplementary Methods 2, appendix C) enables us to quantify the difference between two DETECT measurements, and demonstrates its stability to small perturbations to the shape. In particular, organoids that maintain similar shapes over time yield closely aligned DETECT curves when evaluated with this metric (see Supplementary Methods 2, with details in Supplementary Fig. 2 and Supplementary Table 7, appendix B and C).

Bile canaliculi segmentation and network features extraction

The bile canaliculi were reconstructed from high-resolution (voxel size: 0.3 × 0.3 × 0.3 μm) fluorescent image stacks (50–80 μm depth) of optically cleared HepOrg. Segmentation was performed on CD13 (for bile canaliculi) and F-actin (cell boarders) staining with phalloidin. The analysis of bile canaliculi morphology and bile canaliculi network properties was performed using a Fiji65 script. In brief, images were imported using Bioformats66. Both channels (for CD13 and Phalloidin) were smoothened with a median filter and segmented independently, CD13 with the default ImageJ method, and Phalloidin67. The overlap between the two segmented images was kept as the basis to identify apical patches. The apical patches were then completed by a series of inflations followed by the ‘fill holes’ function and a series of deflations68. At this point a size threshold was applied on the remaining potential lumina, and the lumina touching the borders of the images were removed68. The resulting segmented image is then used for local thickness measurement69, skeletonization70 and skeleton analysis71. The features were then exported as a .csv file and plotted in GraphPad Prism software package (v10.0.2 and v.10.0.0). The custom script described in this manuscript can be found at GitHub (https://git.mpi-cbg.de/huch_lab/assembloid-paper).

Immunofluorescence images from several conditions were used in this analysis: Hu et al.15, Peng et al.16, HM-Wnt HepOrg, MM HepOrg, liver tissue; hereafter, these are referred to as ‘structure’ and individual bile canaliculi networks are referred to as ‘network’. We analysed the features of the largest network per structure (length, junction). A network is defined as an uninterrupted continuation of bile canaliculi branches, which are interconnected (have different amount of triple and quadruple junction points). Please refer to Extended Data Fig. 1c,d for a schematic explanation of networks, branches and junctions. We analysed the features (total length = sum of all branches), and number of junctions, triple or quadruple or combined (total number) of the bile canaliculi for each of the structures. We used number of junctions as a proxy of ‘connectivity’; that is, a network with more junctions is more interconnected than a network with fewer junctions. To compare between structures of different conditions, we plotted these values in dot plots, with one dot representing one structure. To plot the frequency distribution of bile canaliculi diameters of the different conditions (Fig. 2b), we took the diameters of the branches per condition and distribute them in bins of 0.25 μm and plotted them as a percentage of the total number of branches of that bin per condition (HepOrg or liver tissue).

Time-lapse and live imaging

Organoids and assembloids images were acquired with a Leica DMIL LED (bright field only) using a Leica DF C450C camera and Leica Application Suite software (Leica) microscope. Whole-well pictures were acquired with a Leica M80 microscope using a Leica MC170 HD camera and Leica Application Suite software (Leica).

For live/dead staining a Viability/Cytotoxicity Assay Kit (Biotium, 30002) was used according to the manufacturer’s protocol. In brief, 2 μM calcein and 4 μM ethidium homodimer III in PBS were added to cover the Matrigel containing organoids or mesenchyme culture, and incubated for 30 min. Then, the solution was removed and organoid or mesenchymal medium was added and cultures were imaged using a confocal microscope (Zeiss LSM 880) and processed using ImageJ/Fiji.

Time-lapse imaging of assembloid formation and cell death was carried out at 37 °C and 5% CO2 using a Viventis LS1 Live microscope with a double illumination through 10× objectives and a detection 25×1.1 NA objective. MM was supplemented with SPY620-DNA stain (Spirochrome, SC401) at 1 µM final concentration. Videos were generated with ImageJ/Fiji.

The fibrous collagen signal was imaged on an upright or inverted laser-scanning confocal microscope (LSM 780 NLO equipped with Observer Z.1 microscope stand, Zeiss). We used SHG of collagen in response to illumination with 800 nm femtosecond pulsed Chameleon Vision II Ti-Sapphire laser (Coherent). SHG signal was detected in forward propagated direction with the use of photomultiplier tube detector equipped with the bandpass filter 402/15 nm ET. For SHG of collagen fibres in live organoid cultures, W Plan-Apochromat 20×/1.0 DIC UV VIS IR water-dipping objective (Zeiss) was used. For SHG of collagen fibres in fixed organoid cultures, Plan-Apochromat 20×/0.8 objective was used. For tissue imaging, LD LCI Plan-Apochromat 40×/1.2 DIC Imm Corr M27 immersion corrected objective (Zeiss) was used. As an immersion medium for imaging samples mounted in glycerol-based mounting solution (Vectashield, Vector Laboratories) we used 80% glycerol immersion medium of refractive index n = 1,45 - Type G immersion liquid (Leica Microsystems).

Bile acid analogue transport live imaging

We used CLF72 to assess the functionality of bile canaliculi in HepOrg and assembloids (BSEP and MRP2 transporters). To assess both the detail of bile canaliculi and an overall CLF uptake, we performed live imaging using two imaging systems, a single photon point-scanning confocal system (Zeiss LSM 880 Inverted) for high-resolution, and Viventis LS1 lightsheet for imaging the whole structures.

For the imaging of HepOrg were released from Matrigel using Cell Recovery Solution (Corning, 354253) and re-seeded in an 8-well ibidi µ-Slide 8-well Glass Bottom coverslip (Ibidi, 80827) pre-coated with Matrigel (100%). An overlay of pure Matrigel was added and allowed to form a gel for 30 min at 37 °C. HM-Wnt medium was then added to these wells and incubated for 48 h. Medium was refreshed 24 h prior to imaging. 0.5 µM of SiR-actin (Spirochrome, SC001) was added in the culture medium 60 min before imaging. Ibidi slides were mounted into the stage (Zeiss Axio Observer. Z1 inverted stand with stage-top piezo) and allowed to equilibrate for 20 min to reduce thermal drift. XY positions were marked on Zen Black for z-stack acquisition with step size of 2.5 µm and pixel size of 0.08 µm × 0.08 µm or 0.1 µm × 0.1 µm. Live acquisition was performed on the the Zeiss LSM 880 Inverted system with the LD LCI Plan-Apochromat 63×/1.2 Imm Korr DIC M27 objective. The temperature was maintained at 37 °C with 5% humidified CO2 in the sample holder chamber. All structures were imaged with 633 and 488 nm laser excitation and detection via the Fast Airyscan modality on a piezo-controlled stage at T0 prior to addition of CLF. CLF (Corning, 451041), at 10 µg ml−1 final concentration, was added in situ, and acquisition was resumed 30 min after with acquisition every 25 min for 4.5 h. Time series hyperstacks were processed online via Airyscan processing function on Zen Black using the default settings for further analysis.

For the imaging of whole HepOrgs and assembloids, organoids were seeded in Viventis lightsheet holders in Matrigel, and overlaid with MM. HepOrgs were pre-treated for 2 days with MM before the start of imaging. CLF (Corning, 451041), at 10 µg ml−1 final concentration was added 1 frame after the start of imaging. CMFDA73 (also assessing BSEP and MRP2 transporters) and fluorescent phosphatidylcholine (to detect MDR2 function74) were used for further tests. Similarly to CLF imaging, HepOrg were imaged with CMFDA (ThermoFisher, C2925) at 5 µM final concentration, 16:0-06:0 NBD PC fluorescent lipid at 4 µM final concentration (Avanti Polar lipids, 810130P-1mg), and 5-CFDA at 5 µM final concentration (ThermoFisher, C1354), and the compounds were added 1 frame after the start of imaging. Imaging was performed at the intervals specified in the figure or figure legend. For both assembloids and HepOrg, MM was additionally supplemented with SPY620-DNA stain (Spirochrome, SC401), SPY555-DNA stain (Spirochrome, SC201) or SiR-actin (Spirochrome, SC001) at 0.5–1 µM final concentration. Time-lapse imaging was carried out at 37 °C and 5% CO2 using a Viventis LS1 Live microscope with a double illumination through 10× objectives and a detection 25×1.1 NA objective. Videos were generated with ImageJ/Fiji.

RT–qPCR

Total RNA was extracted from cells using the Arcturus PicoPure RNA Isolation Kit (Applied Biosystems, 12204-01) according to the manufacturer’s protocol; including a 15 min digestion step with DNAse to remove traces of genomic DNA. The RNA (50-250 ng) was reverse-transcribed with the Moloney Murine Leukemia Virus reverse transcriptase (M-MLVRT) (Promega, M368B) or alternatively using or SuperScript III Reverse Transcriptase (Invitrogen, 18080-044) according to the manufacturer’s instructions. Finally, cDNA was amplified using FastStart Essential DNA Green Master (Roche, 06402712001) on the LightCycler 96 machine (Roche) or using PowerUp SYBR Green Master Mix (ThermoFisher, A25741) on the Thermo Fisher QuantStudio 7 Pro. The list of primers used for RT–qPCR is provided in Supplementary Table 1. Gene expression levels were normalized to the housekeeping gene as specified in the graph axis labels.

Cytokine array and functional assays

The supernatants (medium) from HepOrgs, homeostasis-like or fibrotic-like assembloid cultures were collected after 48 h of culturing. For albumin ELISA, bile acid measurements, and cytochrome assay, the organoids or assembloids were subsequently dissociated by TrypLE 1× treatment (10 min at 37 °C), and cell number per well was counted using a haemocytometer. The supernatant was then stored in −20 °C until analysis. For supernatant-based assays, the supernatant was used in the assay as per manufacturer’s instructions—Mouse Albumin AssayMax ELISA Kit (Assaypro, EMA3201-1), Total Bile Acid Assay Kit (Colorimetric) (Cell Biolabs, STA-631) and Proteome Profiler Mouse XL Cytokine Array (R&D systems Biotechne, ARY028). For cytochrome activity measurements, the organoids were removed from Matrigel using Cell Recovery Solution (Corning, 354253) for 10 min on ice and subsequently placed in suspension 24-well plate with 0.5 ml PBS or medium in each well. Luciferin substrate was added as specified by manufacturer (P450-Glo CYP1A2 Induction/Inhibition Assay, Promega V8421; P450-Glo CYP3A4 Assay and Screening System, Luciferin IPA, Promega V9001) and left incubating with the cells 6 h in the incubator at 37 °C and 5% CO2. Luciferin detection reagent was equilibrated at room temperature prior to measurement, and mixed at 1:1 ratio with supernatant from cells, incubated 20 min at room temperature and read using luminometer. CellTiter-Glo assay to measure viability was used following the manufacturer’s protocol (G7570, Promega). The data was acquired on Perkin Elmer Envision 2104 for Albumin, BA, CellTiter-Glo and cytochrome activity measurement, and membranes were scanned on an iBright instrument (FL1500) for cytokine array. The unedited cytokine array scans are available in Supplementary Fig. 1.

Bulk RNA-seq and analysis

HepOrg cultured in specified media for 1 or 2 weeks and assembloids cultured for 1 week or 2.5 weeks in their specified media were collected from Matrigel using Cell Recovery solution (354253, Corning), and then processed to RNA using PicoPure protocol, as described above. RNA was eluted with water. RNA libraries were prepared using the SMARTseq2 protocol75 using Illumina Nextera library prep. Samples were sequenced on NovaSeq instrument, and sequencing of paired-end, 2× 100-bp reads was performed at depth of 40mio reads/sample.

RNA-seq data were aligned to the mouse genome GRCm39 release 109 using STAR aligner (2.7.11b). featureCount (v2.0.6) was used to assigned reads exons, transcripts and CDS. Differential gene expression analysis was performed using the R (4.2.0) package DESeq2 (1.36.0). GSEA for bulk RNA-seq data was performed using the R package fgsea (1.22.0).

scRNA-seq

For the scRNA-seq, hepatocytes from wild-type mice, cholangiocytes from a Rosa26-nTnG mouse and portal mesenchyme from a PDGFRα-H2B-GFP mouse sorted for SCA1+ cells were used. Mesenchymes, cholangiocyte organoids and HepOrgs were expanded to passage 2 for the experiment. Cells were aggregated at both healthy homeostatic and fibrotic-like ratios as described above. Hepatocytes were pre-conditioned 48 h in MM before aggregation. The assembloids were dissociated to single cells using TrypLE 1× for 10 min at 37 °C with (repeats 2 and 3) or without pre-warming TrypLE prior to dissociation. The cells were resuspended in PBS with 0.04% BSA, 0.2 U μl−1 RNAse inhibitor (Merck, 3335399001) and 10 μM ROCK inhibitor (Y-27632, Merck/Sigma, Y0503) in BSA-coated tubes for the first replicate, additionally supplemented with DNAse (Sigma, DN25, 1:1000 from 10 mg ml−1 stock) for the second and third biological replicate. The cells were filtered with 100 μm strainer, cell suspensions (7,000–20,000 cells) were loaded into Chromium Next GEM ChipG and processed as specified in manufacturer’s instructions for 10X Chromium Next GEM Single Cell 3′ Reagent Kits v3.1 (10X Genomics). Single-cell RNA-seq libraries were prepared using Single Cell 3′ v3.1 Gel Bead kit (10X Genomics) as per manufacturer’s instructions, and library amplified with 9 cycles. For the cDNA amplification, 11 cycles were used. Libraries were sequenced on NovaSeq 6000, to a depth of 30,000 reads per cell with paired-end 100 bp reads.

scRNA-seq processing

The sequencing data was aligned to the GRCm39 (release 109) mouse genome with the count from 10X Cellranger (7.1.0). Ambient RNA was removed using Cellbender (0.3.0), the number of expected cells estimated by Cellranger and the total number of included droplets set to 25,000. Cells were retained if they had a cell probability >0.99. The sequencing data was further processed with scanpy (1.9.276). Cells with fewer than 1,000 genes expressed were excluded from further analysis. We further excluded cells with more than 25 % mitochondrial read counts, cells with more than 10,000 genes expressed, and total read count over 50,000 as potential doublets. We further eliminated doublets using Scrublet77 (https://github.com/swolock/scrublet). We further eliminated doublets using the eGFP and tdTomato marker genes in Mesenchyme and Cholangiocyte cell populations, respectively. Counts were normalized per cell and log-transformed. Variation in total gene count and percent mitochondrial gene count were regressed out before count scaling. For generating a consensus zonation score, we averaged the expression of validated pericentral or periportal markers from three different publicly available datasets13,29,30, using the scanpy function score_genes.

scRNA-seq dimensionality reduction and clustering

Cells were clustered using the Leiden algorithm implemented in scanpy based on the k-nearest neighbours (kNN) ggraph calculated using scanpy’s neighbours function using 15 neighbours and 40 principal components. We assigned cell per cluster as one of three cell types, Hepatocytes (Hnf4a, Apoa1, Apoc1, Hpx, Ttr, Alb, Mup20, Gpx1, Cyp3a11, Abcb4), Mesenchyme (Pdgfrα, Pdgfrb, Des, Vim, Col1a1, Col1a2, Col3a1, Col6a2, Ly6a) and Cholangiocytes (Krt19, Krt7, Epcam, Ezr, Sox9, Prom1) based on marker gene expression. All dotplots, UMAPs and correlation plots were produced with the corresponding scanpy functions. Violin plots were created using the plotnine python package. Pearson coefficient of all groups indicated in plots was calculated as the correlation between normalized counts.

Integration with published single cell datasets

We obtained raw read count data from several previously published liver cell datasets11,12,13,33,34,35,36,37,38,39. Datasets were aggregated before processing using scanpy and then processed together as described above. Dimensionality reduction and clustering was performed as described above with the difference that the kNN ggraph for clustering was calculated using BBKNN78. Cell types were assigned as in the original publications used. We clustered cell types across different datasets and conditions using Pearson correlation coefficient.

Differential gene expression and GSEA

Differential gene expression was calculated pairwise between conditions using a Wilcoxon rank-sum test. Genes were considered differentially expressed if they showed an absolute log-foldchange >1 and a Benjamini–Hochberg adjusted P value < 0.05. Gene set enrichment analysis was performed using the gseapy (1.0.5) package. Whole-organoid GSEA between healthy and fibrotic conditions was performed for the MSigDB hallmark (2020), WikiPathways (2019, Mouse), Reactome (2022), Elsevier Pathway Collection and GO Molecular Function (2023) gene sets. Cell type specific GSEA between conditions was performed for the MSigDB hallmark (2020) gene set. Functional identity was assessed by GSEA by comparing each cell type to all other cell types using the GO Molecular Function (2023), GO Biological Process (2023) and GO Cellular Component (2023) gene sets. For all GSEA, normalized enrichment scores (NES) and FDR-adjusted P values are reported.

Cell–cell communication analysis

We used the ligand–receptor analysis framework (LIANA, 0.1.9)79 to explore cell–cell communication via ligand–receptor expression. We used LIANAs rank_aggregation method to calculate seven different cell–cell communication metrics as well as their rank aggregation. We used the magnitude rank as interaction score. We only retained interactions with a specificity rank less than 0.05. Ligand–receptor interactions were visualized using the R (4.2.0) package ggraph (2.1.0).

Statistical analysis

Data were analysed as detailed in figure legends and as appropriate for each experiment by using two-tailed Mann–Whitney test, Welch’s t-test, unpaired t-test with Welch’s correction, paired t-test, a Student’s t-test, or Kruskal–Wallis test with Dunn’s multiple comparisons post hoc test. P < 0.05 was considered statistically significant. Calculations were performed using GraphPad Prism software package (v10.0.2 and v.10.0.0). All P values are given in the corresponding figure legends or figures. Dispersion and precision measures (such as mean, median, s.d. and s.e.m.) are specified in the figure legends. Statistics for scRNA-seq data are described in the corresponding section.

Reporting summary

Further information on research design is available in the Nature Portfolio Reporting Summary linked to this article.