single-cell signalling analysis of heterocellular organoids · single-cell signalling analysis of...

11
Single-Cell Signalling Analysis of Heterocellular Organoids Xiao Qin 1 , Jahangir Sufi 1* , Petra Vlckova 1* , Pelagia Kyriakidou 1* , Sophie E. Acton 2 , Vivian S. W. Li 3 , Mark Nitz 4 , and Christopher J. Tape 1, 1 Cell Communication Lab, Department of Oncology, University College London Cancer Institute, London, UK. 2 Stromal Immunology Lab, MRC Laboratory for Molecular Cell Biology, University College London, London, UK. 3 Stem Cell and Cancer Biology Lab, The Francis Crick Institute, London, UK. 4 Department of Chemistry, University of Toronto, Toronto, Canada. * These authors contributed equally to this work. Organoids are powerful biomimetic tissue models. Despite their widespread adoption, methods to analyse cell-type spe- cific post-translational modification (PTM) signalling networks in organoids are absent. Here we report multivariate single-cell analysis of cell-type specific signalling networks in organoids and organoid co-cultures. Simultaneous measurement of 28 PTMs in >1 million single small intestinal organoid cells by mass cytometry reveals cell-type and cell-state specific sig- nalling networks in stem, Paneth, enteroendocrine, tuft, gob- let cells, and enterocytes. Integrating single-cell PTM analysis with Thiol-reactive Organoid Barcoding in situ (TOBis) enables high-throughput comparison of signalling networks between organoid cultures. Multivariate cell-type specific PTM analy- sis of colorectal cancer tumour microenvironment organoids re- veals that shApc, Kras G12D , and Trp53 R172H cell-autonomously mimic signalling states normally induced by stromal fibroblasts and macrophages. These results demonstrate how standard mass cytometry workflows can be modified to perform high- throughput multivariate cell-type specific signalling analysis of healthy and cancerous organoids. Correspondence: [email protected] INTRODUCTION Organoids are self-organising 3D tissue models comprising stem and differentiated cells (1). Organoid monocultures typ- ically contain one major cell class (e.g. epithelial) and can be co-cultured with heterotypic cell-types (e.g. mesenchy- mal (2) or immune cells (3)) to model cell-cell interactions in vitro. When compared with traditional 2D cell culture, organoids more accurately represent their parental tissue and are emerging as powerful models for studying multicellular diseases such as cancer (4). Post-translational modification (PTM) signalling networks underpin fundamental biological phenotypes and are fre- quently dysregulated in disease (5). As different cell-types have different signalling networks (6, 7), organoids likely contain several cell-type specific PTM networks that are es- sential to their biology. In order to fully utilise biomimetic models of healthy and diseased tissue, we must be able to study PTM signalling networks within organoids. Un- fortunately, no technology currently exists to analyse cell- type specific PTM networks in organoids and organoid co- cultures. Organoids present several technical challenges over tradi- tional 2D cultures for PTM analysis. Firstly, organoids are embedded in a protein-rich extracellular matrix (ECM) that confounds the application of phosphoproteomic anal- ysis by liquid chromatography tandem mass spectrometry (LC-MS/MS). Organoids can be removed from ECM prior to LC-MS/MS, but as dissociation of live cells alters cell signalling (8), PTM measurements from dissociated live organoids do not truly represent in situ cellular states. Ideally, organoids should be fixed in situ to preserve PTM signalling, but LC-MS/MS analysis of heavily cross-linked phospho- proteomes is extremely challenging. Secondly, as organoids comprise multiple cell-types (e.g. stem and differentiated) and cell-states (e.g. proliferating, quiescent, and apoptotic), bulk phosphoproteomics cannot capture the biological het- erogeneity present in organoids and organoid co-cultures (9). Although single-cell RNA-sequencing (scRNA-seq) can de- scribe organoid cell-types (10), it cannot measure intracellu- lar PTM signalling at the protein level. Finally, as signalling networks comprise multiple PTM nodes, low-dimensional methods (e.g. fluorescent imaging) cannot capture the com- plexity of PTM signalling networks (9). Collectively, to study PTM networks in organoids, we require signalling data that is: 1) cell-type specific, 2) derived from cells fixed in situ, and 3) measures multiple PTMs simultaneously. Mass cytometry (MC, also known as cytometry time-of-flight (CyTOF)) uses heavy metal-conjugated antibodies to mea- sure >35 proteins in single cells (11). Although MC is tradi- tionally used for high-dimensional immunophenotyping, MC can also measure PTMs in heterocellular systems (e.g. pe- ripheral blood mononuclear cells (PBMCs) (12) and tissue (8)). Given MC’s capacity to measure PTMs in mixtures of fixed cells, we theorised that MC workflows typically applied to immunophenotyping could be modified to study cell-type specific signalling networks in organoids. Here we report the development of a custom multivariate- barcoded MC method to measure single-cell signalling in ep- ithelial organoids and organoids co-cultured with stromal and immune cells. This method reveals that intestinal organoids display cell-type specific signalling networks that are inti- mately linked with cell-state. When applied to colorectal cancer (CRC) tumour microenvironment (TME) organoid co-cultures, we discovered that epithelial oncogenic muta- tions mimic signalling networks normally induced by stromal Qin et al. | bioRiv | November 11, 2019 | 1–11 All rights reserved. No reuse allowed without permission. The copyright holder for this preprint (which was not peer-reviewed) is the author/funder. . https://doi.org/10.1101/659896 doi: bioRxiv preprint

Upload: others

Post on 12-Mar-2020

2 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Single-Cell Signalling Analysis of Heterocellular Organoids · Single-Cell Signalling Analysis of Heterocellular Organoids Xiao Qin1, Jahangir Sufi1*, Petra Vlckova1*, Pelagia Kyriakidou1*,

Single-Cell Signalling Analysis ofHeterocellular Organoids

Xiao Qin1, Jahangir Sufi1*, Petra Vlckova1*, Pelagia Kyriakidou1*, Sophie E. Acton2, Vivian S. W. Li3, Mark Nitz4, andChristopher J. Tape1,

1Cell Communication Lab, Department of Oncology, University College London Cancer Institute, London, UK.2Stromal Immunology Lab, MRC Laboratory for Molecular Cell Biology, University College London, London, UK.

3Stem Cell and Cancer Biology Lab, The Francis Crick Institute, London, UK.4Department of Chemistry, University of Toronto, Toronto, Canada.

*These authors contributed equally to this work.

Organoids are powerful biomimetic tissue models. Despitetheir widespread adoption, methods to analyse cell-type spe-cific post-translational modification (PTM) signalling networksin organoids are absent. Here we report multivariate single-cellanalysis of cell-type specific signalling networks in organoidsand organoid co-cultures. Simultaneous measurement of 28PTMs in >1 million single small intestinal organoid cells bymass cytometry reveals cell-type and cell-state specific sig-nalling networks in stem, Paneth, enteroendocrine, tuft, gob-let cells, and enterocytes. Integrating single-cell PTM analysiswith Thiol-reactive Organoid Barcoding in situ (TOBis) enableshigh-throughput comparison of signalling networks betweenorganoid cultures. Multivariate cell-type specific PTM analy-sis of colorectal cancer tumour microenvironment organoids re-veals that shApc, Kras

G12D, and Trp53R172H cell-autonomously

mimic signalling states normally induced by stromal fibroblastsand macrophages. These results demonstrate how standardmass cytometry workflows can be modified to perform high-throughput multivariate cell-type specific signalling analysis ofhealthy and cancerous organoids.

Correspondence: [email protected]

INTRODUCTIONOrganoids are self-organising 3D tissue models comprisingstem and differentiated cells (1). Organoid monocultures typ-ically contain one major cell class (e.g. epithelial) and canbe co-cultured with heterotypic cell-types (e.g. mesenchy-mal (2) or immune cells (3)) to model cell-cell interactionsin vitro. When compared with traditional 2D cell culture,organoids more accurately represent their parental tissue andare emerging as powerful models for studying multicellulardiseases such as cancer (4).Post-translational modification (PTM) signalling networksunderpin fundamental biological phenotypes and are fre-quently dysregulated in disease (5). As different cell-typeshave different signalling networks (6, 7), organoids likelycontain several cell-type specific PTM networks that are es-sential to their biology. In order to fully utilise biomimeticmodels of healthy and diseased tissue, we must be ableto study PTM signalling networks within organoids. Un-fortunately, no technology currently exists to analyse cell-type specific PTM networks in organoids and organoid co-cultures.Organoids present several technical challenges over tradi-

tional 2D cultures for PTM analysis. Firstly, organoidsare embedded in a protein-rich extracellular matrix (ECM)that confounds the application of phosphoproteomic anal-ysis by liquid chromatography tandem mass spectrometry(LC-MS/MS). Organoids can be removed from ECM priorto LC-MS/MS, but as dissociation of live cells alters cellsignalling (8), PTM measurements from dissociated liveorganoids do not truly represent in situ cellular states. Ideally,organoids should be fixed in situ to preserve PTM signalling,but LC-MS/MS analysis of heavily cross-linked phospho-proteomes is extremely challenging. Secondly, as organoidscomprise multiple cell-types (e.g. stem and differentiated)and cell-states (e.g. proliferating, quiescent, and apoptotic),bulk phosphoproteomics cannot capture the biological het-erogeneity present in organoids and organoid co-cultures (9).Although single-cell RNA-sequencing (scRNA-seq) can de-scribe organoid cell-types (10), it cannot measure intracellu-lar PTM signalling at the protein level. Finally, as signallingnetworks comprise multiple PTM nodes, low-dimensionalmethods (e.g. fluorescent imaging) cannot capture the com-plexity of PTM signalling networks (9). Collectively, to studyPTM networks in organoids, we require signalling data thatis: 1) cell-type specific, 2) derived from cells fixed in situ,and 3) measures multiple PTMs simultaneously.

Mass cytometry (MC, also known as cytometry time-of-flight(CyTOF)) uses heavy metal-conjugated antibodies to mea-sure >35 proteins in single cells (11). Although MC is tradi-tionally used for high-dimensional immunophenotyping, MCcan also measure PTMs in heterocellular systems (e.g. pe-ripheral blood mononuclear cells (PBMCs) (12) and tissue(8)). Given MC’s capacity to measure PTMs in mixtures offixed cells, we theorised that MC workflows typically appliedto immunophenotyping could be modified to study cell-typespecific signalling networks in organoids.

Here we report the development of a custom multivariate-barcoded MC method to measure single-cell signalling in ep-ithelial organoids and organoids co-cultured with stromal andimmune cells. This method reveals that intestinal organoidsdisplay cell-type specific signalling networks that are inti-mately linked with cell-state. When applied to colorectalcancer (CRC) tumour microenvironment (TME) organoidco-cultures, we discovered that epithelial oncogenic muta-tions mimic signalling networks normally induced by stromal

Qin et al. | bioR‰iv | November 11, 2019 | 1–11

All rights reserved. No reuse allowed without permission. The copyright holder for this preprint (which was not peer-reviewed) is the author/funder.. https://doi.org/10.1101/659896doi: bioRxiv preprint

Page 2: Single-Cell Signalling Analysis of Heterocellular Organoids · Single-Cell Signalling Analysis of Heterocellular Organoids Xiao Qin1, Jahangir Sufi1*, Petra Vlckova1*, Pelagia Kyriakidou1*,

cells. These results demonstrate how a modified MC methodcan enable powerful multivariate single-cell analysis of cell-type specific signalling in heterocellular organoids.

RESULTSSingle-Cell Analysis of Organoids by Mass Cytometry.No technology currently exists to study cell-type specific pro-tein signalling networks in organoids. Given its capacity tomeasure multiple PTMs in single cells, we hypothesised MCcould be modified to study cell-type and cell-state specificsignalling in organoids. To test this, we first developed aMC platform to measure single-cell signalling in the classicalsmall intestinal organoid (13).In this method, we first pulse live organoids with 1275-Iodo-2’-deoxyuridine (127IdU) to identify S-phase cells (14), fixorganoids in Matrigel to preserve cell signalling, and stainorganoids with 194/8Cisplatin to label dead epithelia (15). Us-ing a custom workflow, we then physically and enzymati-cally dissociate the fixed organoids into single cells prior toextra- and intracellular heavy-metal antibody staining (Fig.1a). We next performed a comprehensive screen for in-testinal epithelial cell-type identification antibodies includ-ing stem (LGR5, LRIG1, OLFM4), Paneth (Lysozyme), gob-let (MUC2, CLCA1), enteroendocrine (CHGA, Synapto-physin), tuft cells (DCAMKL1), and enterocytes (FABP1,Na/K-ATPase) that bind fixed antigens and are compatiblewith rare-earth metal conjugation for MC. Cell-type identi-fication antibodies were validated by organoid directed dif-ferentiation (16) (Supplementary Fig. 1) and integrated intoa panel of 28 anti-PTM rare-earth metal antibodies spanningmultiple core signalling nodes (Supplementary Table 1, 45parameters (40 antibodies) / cell). When analysed by MC,this method enables the measurement of 28 signalling PTMsacross 6 cell-types in >1 million single cells from fixed in-testinal organoids (Figs. 1b, c, 2a, and Supplementary Fig.2a).Combining 194/8Cisplatin and 127IdU with canonical cell-cycle markers (e.g. pRB [S807/S811], Cyclin B1, and pHi-stone H3 [S28] (14)) allows clear identification of live /dead cells and classification of single organoid cells intocell-cycle stages including G0, G1, S, G2, and M-phase(17) (Supplementary Fig. 2b). Integrated cell-type andcell-state data from small intestinal organoids confirmed thatstem and Paneth cells are largely proliferative (pRB+, cCas-pase3 [D175]-), whereas differentiated epithelia are oftenpost-mitotic (pRB-, IdU- / pHH3-) or apoptotic (cCaspase3+)(Fig. 1c). Consistent with the finding that intestinal progen-itor cells have permissive chromatin in vivo (18), proliferat-ing intestinal organoid cells also present H3K4me2 whereaspost-mitotic cells do not (Fig. 2a). These results confirmedthat a modified MC workflow can provide cell-type and cell-state specific information from millions of single organoidcells.

Cell-Type and Cell-State Specific Signalling Networksin Intestinal Organoids. Following cell-type and cell-stateidentification, we next sought to construct cell-type specific

PTM signalling networks in small intestinal organoids. To in-vestigate whether stem, Paneth, enteroendocrine, tuft, gobletcells, and enterocytes employ different PTM signalling net-works, we combined Earth Mover’s Distance (EMD) (19, 20)and Density Resampled Estimation of Mutual Information(DREMI) (21) to build quantitative cell-type specific sig-nalling networks from single-cell organoid PTM data (Fig.2b and Supplementary Fig. 2). In these networks, EMDquantifies PTM intensity (node score) for each organoid cell-type relative to the total organoid population and DREMIquantifies PTM-PTM connectivity (edge score) within thenetwork.

EMD-DREMI analysis revealed cell-type specific PTM sig-nalling networks in small intestinal organoids. As canonicalWNT signalling is mainly driven by protein interactions, lo-calisation, and degradation (22) – not a classical PTM cas-cade – MC is not well suited to studying the WNT path-way. Despite this limitation, evidence of WNT flux via in-hibited pGSK-3— [S9] and non-phosphorylated —-Cateninis observed in all organoid cell-types (Fig. 2a). In con-trast, MAPK and PI3K pathways display unexpected cell-type specificity. For example, stem cells channel MAPK sig-nalling through pERK1/2 [T202/Y204], pP90RSK [T359],and pCREB [S133], but fail to connect with pBAD [S112](Fig. 2a, b). On the contrary, differentiated epithelia di-rect MAPK signalling away from pCREB and towards pBADwhen proliferating, and lose all MAPK activity in G0 andapoptosis (Fig. 2a). Despite their strong mitogenic signallingprofile, stem cells are unique among proliferating cells intheir failure to phosphorylate BAD. This suggests that in-testinal stem cells avoid apoptosis independent of the classi-cal BAD-BCL-BAX/BAK axis and may compensate via highMAPK and P38 flux to CREB. Consistent with the observa-tion that PI3K signalling is important for intestinal crypt cells(23), stem and Paneth cells are enriched for pSRC [Y418] anddownstream PI3K effectors such as pPDPK1 [S241], pPKC–[T497], pAKT [T308] / [S473], and p4E-BP1 [T37/T46](Fig. 2a, b). Despite the presence of the BMP inhibitor Nog-gin in organoid culture media, Paneth cells display unexpect-edly high BMP signalling (via pSMAD1/5 [S463/S465] andSMAD9 [S465/S467]) (Fig. 2a, b). This observation sug-gests that Paneth cells are either hypersensitive to BMP lig-ands or can cell-intrinsically activate SMAD1/5/9 (possiblyvia inhibition of SMAD phosphatases).

Several PTM signalling events correlate with cell-state inintestinal organoids. For example, irrespective of cell-typeor location, pP38 MAPK [T180/Y182] and pP120-Catenin[T310] are active in all proliferating cells, and both pAKT[T308] and pMKK4 [S257] are hyperactivated in M-phase(Figs. 1c and 2a). In contrast, TGF-— signalling (via pS-MAD2 [S465/S467] and SMAD3 [S423/S425]) is exclu-sively active in post-mitotic epithelia (Fig. 2a), consistentwith TGF-—’s role in epithelial growth-arrest (24). To in-vestigate the relationship between cell-type and cell-statein PTM signalling networks, we performed principal com-ponent analysis (PCA) of PTM-EMDs for each organoidcell-type, either proliferating or in G0 (pRB+/–), located in

2 | bioR‰iv Qin et al. | Single-Cell Organoid Signalling

All rights reserved. No reuse allowed without permission. The copyright holder for this preprint (which was not peer-reviewed) is the author/funder.. https://doi.org/10.1101/659896doi: bioRxiv preprint

Page 3: Single-Cell Signalling Analysis of Heterocellular Organoids · Single-Cell Signalling Analysis of Heterocellular Organoids Xiao Qin1, Jahangir Sufi1*, Petra Vlckova1*, Pelagia Kyriakidou1*,

Pan-CK

CD44

LRIG1

Lysozyme

CLCA1DCAMKL1

CHGA FABP1

pRB [S807/S811] IdU pHistone H3 [S28] cCaspase 3 [D175]

a

Organoid MassCytometry

Rare-EarthMetal Abs

Single-CellDissociation

OrganoidFixation

127IdU

×9 Cell-Type×28 PTM

PFA 194/8Cisp

Cell 1

Cell 2m/z

m/z

Cell nm/z

Y YY YY

Y

YYYY Y YY

Y

Y

Y

Single-Cell Organoid Signalling

...

b

FABP1CD44 LysozymeCel

l-Typ

e

Epithelial

Lower-Crypt Paneth Enterocyte

pRB [S807/S811] EdU cCaspase 3 [D175]pHistone H3 [S28]ApoptoticM-PhaseS-Phase

Cel

l-Sta

te

Pan-CKStem

DCAMKL1Tuft

CLCA1

EnteroendocrineCHGA

Goblet c

Single-CellProtein Data

Quadrupole TOF-MS

Proliferating

LRIG1Epithelial

Lower-Crypt Paneth Enterocyte

ApoptoticM-PhaseS-Phase

Stem

Enteroendocrine

Goblet

Proliferating

Tuft

Pro.Phos.Inhib.

115In 162Dy 173Yb 171Yb

175Lu 144Nd 174Yb 145Nd

150Nd 127I 89Y 142Nd

Relat

ive M

arke

r Int

ensit

y

UMAP1

UMAP

2

Cells

PTMs

Cell-Type Cell-State

.

.

.

12

n

Fig. 1. Cell-Type and Cell-State Identification of Single Organoid Cells by Mass Cytometry. a) Experimental workflow. Live organoids are pulsed with 127IdU to labelS-phase cells, treated with protease / phosphatase inhibitors, fixed with PFA to preserve post translational modification (PTM) signals, and stained with 194/8Cisplatin to labeldead cells. Fixed organoids are then dissociated into single cells, stained with rare-earth metal-conjugated antibodies, and analysed by single-cell mass cytometry (MC).The resulting dataset contains integrated cell-type, cell-state, and PTM signalling information. b) Confocal immunofluorescence (IF) of small intestinal organoids stainedwith rare-earth metal-conjugated MC antibodies highlighting individual cell-type and cell-state markers (red), F-Actin (white), and DAPI (blue), scale bars = 50 µm. (SeeSupplementary Fig. 1 for antibody validation via directed differentiation.) c) UMAP (Uniform Manifold Approximation and Projection) distribution of 1 million single organoidcells analysed by MC resolves six major intestinal cell-types across proliferating, S-phase, M-phase, and apoptotic cell-states. Colours represent normalised local parameterintensity. (See Supplementary Fig. 2 for cell-type and cell-state classification.)

lower-crypts or villi (CD44+/–). PCA revealed that both cell-state (PC1, 68% variance) and, to a lesser extent, cell-type(PC2, 23% variance) dictate cell-signalling in small intestinalorganoids (Fig. 2c and Supplementary Fig. 3). This analysisdemonstrates that both cell-type and cell-state are intimatelylinked with cell-signalling and warns against bulk PTM anal-ysis of organoids where cell-type and cell-state resolution islost. Collectively, these results confirmed that MC can iden-tify novel cell-type and cell-state specific signalling networksin small intestinal organoids and underscore the importanceof single-cell data when studying heterogenous systems suchas organoids.

Single-Cell Organoid Multiplexing using Thiol-reactiveOrganoid Barcoding in situ (TOBis). We have demon-strated how a modified MC platform can be applied tocell-type and cell-state specific signalling measurement inorganoids. However, in order to study differential signaltransduction in organoid models of healthy and diseased tis-sue, we must also be able to directly compare PTM networksbetween different organoid cultures. In addition to high di-mensional single-cell PTM measurements, a major advantageof MC is its ability to perform multiplexed barcoding of ex-perimental variables (25, 26). Unfortunately, commerciallyavailable Palladium-based barcodes cannot bind organoids

in situ (Supplementary Fig. 4a) as they react with Matrigelproteins (Supplementary Fig. 4b), meaning that organoidsmust be removed from Matrigel and dissociated separatelybefore barcoding. Individually removing fixed organoidsfrom Matrigel is a very low-throughput process that limits thescalability of organoid MC multiplexing. We theorised thatif organoids could be barcoded in situ, barcoded organoidscould be pooled, dissociated, and processed as a single high-throughput MC sample. To explore this idea, we developed anew strategy to isotopically barcode organoids while still inMatrigel.

MC barcoding strategies can use amine- (26) or thiol-reactive(25) chemistries. We first used fluorescent probes to investi-gate how each of these chemistries react with ECM proteinsand organoids. Amine-reactive probes (Alexa Fluor 647 NHSester) bind ECM proteins (via lysines and N-terminal amines)and thus fail to label organoids in Matrigel. In contrast,thiol-reactive probes (Alexa Fluor 647 C2 maleimide) by-pass ECM proteins and bind exclusively to reduced-cysteineson organoids in situ (Fig. 3a, Supplementary Fig. 4c).We subsequently confirmed that thiol-reactive monoisotopicmass-tagged probes (C2 maleimide-DOTA-157Gd) also bindorganoids in situ, whereas amine-reactive probes (NHS ester-DOTA-157Gd) only react ex situ (Fig. 3b). This data con-firmed that thiol-reactive chemistries can be used to barcode

Qin et al. | Single-Cell Organoid Signalling bioR‰iv | 3

All rights reserved. No reuse allowed without permission. The copyright holder for this preprint (which was not peer-reviewed) is the author/funder.. https://doi.org/10.1101/659896doi: bioRxiv preprint

Page 4: Single-Cell Signalling Analysis of Heterocellular Organoids · Single-Cell Signalling Analysis of Heterocellular Organoids Xiao Qin1, Jahangir Sufi1*, Petra Vlckova1*, Pelagia Kyriakidou1*,

aPT

Ms

Stem100% pRB+

0% cC3+

Paneth74% pRB+

1% cC3+

Enteroendocrine72% pRB+

5% cC3+

Tuft40% pRB+

10% cC3+

Enterocyte45% S5%+

19% cC3+

Goblet43% pRB+

11% cC3+

<-1.6 -0.7 0.2 >1.2

ProteinPTM

EMD(Nodes)

Up-Regulatedin Cell-Type

Down-Regulatedin Cell-Type

PTM � PTM �

<0.1 0.3 0.6 >0.75

DREMI(Edges)

Strong Connection

Weak Connection

Total Organoid: 55% pRB+, 8% cC3+

T308 S473AKT AKT

MTORC1

APCAXINCK1

MTORC1

D175Caspase-3

S807 S811RB

S112BAD

S33 S37 T41β-Catenin

T202 Y204ERK1/2

T359p90RSK

S221MEK1/2

CDK2/4 Caspase-9S133CREB

S257MKK4

S9GSK-3β

PI3KY418SRC

S463 S465SMAD1/5/9

S529NF-κB p65

Y551BTK

S465 S467SMAD2/3

TGF-βR

MTORC2

FZD LRP LGR ZNRF

T497PKCɑ

IKKɑ/β/γ

E-cadherin

T37 T46 T37 T464E-BP1 4E-BP1

LKB1

T310p120-Catenin

S241PDPK1

SYK

S235 S236S6

S235 S236S6

BMPREGFR

T172AMPKɑ

T180 Y182p38 MAPK

T334MAPKAPK2

S189 S207MKK3/6

RAS

GRB2

MEKK

K4Histone H3

T308 S473AKT AKT

MTORC1

APCAXINCK1

MTORC1

D175Caspase-3

S807 S811RB

T180 Y182p38 MAPK

T334MAPKAPK2

S112BAD

S33 S37 T41β-Catenin

T202 Y204ERK1/2

T359p90RSK

S221MEK1/2

S189 S207MKK3/6

CDK2/4 Caspase-9S133CREB

S257MKK4

S9GSK-3β

RAS PI3K

EGFR

GRB2

Y418SRC

S463 S465SMAD1/5/9

MEKK

S529NF-κB p65

Y551BTK

S465 S467SMAD2/3

TGF-βR

MTORC2

FZD LRP LGR ZNRF

T497PKCɑ

IKKɑ/β/γ

E-cadherin

T37 T46 T37 T464E-BP1 4E-BP1

LKB1

T310p120-Catenin

S241PDPK1

SYK

S235 S236S6

S235 S236S6

T172AMPKɑ

BMPR

K4Histone H3

T308 S473AKT AKT

MTORC1

APCAXINCK1

MTORC1

D175Caspase-3

S807 S811RB

T180 Y182p38 MAPK

T334MAPKAPK2

S112BAD

S33 S37 T41β-Catenin

T202 Y204ERK1/2

T359p90RSK

S221MEK1/2

S189 S207MKK3/6

CDK2/4 Caspase-9S133CREB

S257MKK4

S9GSK-3β

RAS PI3K

EGFR

GRB2

Y418SRC

S463 S465SMAD1/5/9

BMPR

MEKK

S529NF-κB p65

Y551BTK

S465 S467SMAD2/3

TGF-βR

MTORC2

FZD LRP LGR ZNRF

T172AMPKɑ

T497PKCɑ

IKKɑ/β/γ

E-cadherin

T37 T46 T37 T464E-BP1 4E-BP1

LKB1

T310p120-Catenin

S241PDPK1

SYK

S235 S236S6

S235 S236S6

K4Histone H3

T308 S473AKT AKT

MTORC1

APCAXINCK1

MTORC1

D175Caspase-3

S807 S811RB

T180 Y182p38 MAPK

T334MAPKAPK2

S112BAD

S33 S37 T41β-Catenin

T202 Y204ERK1/2

T359p90RSK

S221MEK1/2

S189 S207MKK3/6

CDK2/4 Caspase-9S133CREB

S257MKK4

S9GSK-3β

RAS PI3K

EGFR

GRB2

Y418SRC

S463 S465SMAD1/5/9

BMPR

MEKK

S529NF-κB p65

Y551BTK

S465 S467SMAD2/3

TGF-βR

MTORC2

FZD LRP LGR ZNRF

T172AMPKɑ

T497PKCɑ

IKKɑ/β/γ

E-cadherin

T37 T46 T37 T464E-BP1 4E-BP1

LKB1

T310p120-Catenin

S241PDPK1

SYK

S235 S236S6

S235 S236S6

K4Histone H3

T308 S473AKT AKT

MTORC1

APCAXINCK1

MTORC1

D175Caspase-3

S807 S811RB

T180 Y182p38 MAPK

T334MAPKAPK2

S112BAD

S33 S37 T41β-Catenin

T202 Y204ERK1/2

T359p90RSK

S221MEK1/2

S189 S207MKK3/6

CDK2/4 Caspase-9S133CREB

S257MKK4

S9GSK-3β

RAS PI3K

EGFR

GRB2

Y418SRC

S463 S465SMAD1/5/9

BMPR

MEKK

S529NF-κB p65

Y551BTK

S465 S467SMAD2/3

TGF-βR

MTORC2

FZD LRP LGR ZNRF

T172AMPKɑ

T497PKCɑ

IKKɑ/β/γ

E-cadherin

T37 T46 T37 T464E-BP1 4E-BP1

LKB1

T310p120-Catenin

S241PDPK1

SYK

S235 S236S6

S235 S236S6

K4Histone H3

b

T308 S473AKT AKT

MTORC1

APCAXINCK1

MTORC1

S807 S811RB

T180 Y182p38 MAPK

T334MAPKAPK2

S112BAD

S33 S37 T41β-Catenin

T202 Y204ERK1/2

T359p90RSK

S221MEK1/2

S189 S207MKK3/6

CDK2/4S133CREB

S257MKK4

S9GSK-3β

RAS PI3K

EGFR

GRB2

Y418SRC

S463 S465SMAD1/5/9

BMPR

MEKK

S529NF-κB p65

Y551BTK

S465 S467SMAD2/3

TGF-βR

MTORC2

FZD LRP LGR ZNRF

T172AMPKɑ

T497PKCɑ

IKKɑ/β/γ

E-cadherin

T37 T46 T37 T464E-BP1 4E-BP1

LKB1

T310p120-Catenin

S241PDPK1

SYK

D175Caspase-3Caspase-9

S235 S236S6

S235 S236S6

K4Histone H3

Stem

Paneth

Enteroendocrine

Tuft

Enterocyte

Goblet

Lower-CryptProliferating

1 Million Small Intestinal Organoid Cells

(pRB+) (CD44+)

WNT / β-Catenin Pathway

JNK Pathway

P38 Pathway

BMP / Noggin TGF-β Perm. ChromatinAdhesions

MAPK PathwaypMEK1/2 [S221]

170Er

pERK1/2 [T202/Y4]

167Er

pP90RSK [T359]

163Dy

pCREB [S133]

153Eu

pBAD [S112]

161Dy

pGSK-3β [S9]

166Er

β-Catenin [Active]

165Ho

PI3K PathwaypSRC [Y418]

148Nd

pPDPK1 [S241]

141Pr

pPKCɑ [T497]

151Eu

pAMPKα [T172]

160Gd

pAKT [T308]

152Sm

pAKT [S473]

155Gd

pS6 [S235/S236]

172Yb

pSMAD1/5/9

154Sm

pSMAD2/3

168Er

pMKK4 [S257]

146Nd

pMKK3/6

157Gd

pP38 [T180/Y182]

158Gd

pMK2 [T334]

159Tb

pP120-Cat [T310]

164Dy

me2-HH3 [K4]

209Bi

p4E-BP1 [T37/T46]

149Sm

UMAP1

UMAP

2

Relat

ive M

arke

r Int

ensit

y

Diffe

rent

iation

Differentiation

-12 -8 -4 0 4 8 12

-6

-4

-2

0

2

4

6 StemPanethEndo.

TuftEntero.

Goblet

PC 1 (68% Variance)

PC 2

(23%

Var

ianc

e)

Cel

l-Typ

e

pRB+pRB-

CD44+

CD44-

Cell-State

Diffe

rent

iation

c

Fig. 2. Cell-Type and Cell-State Specific Signalling Analysis of Intestinal Organoids. a) UMAP distributions of PTMs across 1 million single organoid cells analysedby MC. Cell-type and cell-state UMAP guide is shown top left (see Supplementary Fig. 2 for cell-type and cell-state classification). Combining cell-type, cell-state, and PTMmeasurements enables cell-type specific analysis of intestinal organoid signalling. Colours represent normalised local parameter intensity. b) Cell-type specific PTM signallingnetworks in small intestinal organoids, with nodes coloured by PTM-EMD (Earth Mover’s Distance) scores quantifying PTM intensity (relative to all organoid cells) and edgescoloured by DREMI (Density Resampled Estimation of Mutual Information) scores quantifying PTM-PTM connectivity. Small intestinal organoids display cell-type specificsignalling networks. c) Principal Component Analysis (PCA) of PTM-EMDs for all organoid cell-types, either proliferating (pRB+) / G0 (pRB-) or in lower-crypts (CD44+) / villi(CD44-). Organoid cell signalling is dictated by cell-state (PC 1) and cell-type (PC 2). (See Supplementary Fig. 3 for complete EMD-DREMI signalling maps.)

4 | bioR‰iv Qin et al. | Single-Cell Organoid Signalling

All rights reserved. No reuse allowed without permission. The copyright holder for this preprint (which was not peer-reviewed) is the author/funder.. https://doi.org/10.1101/659896doi: bioRxiv preprint

Page 5: Single-Cell Signalling Analysis of Heterocellular Organoids · Single-Cell Signalling Analysis of Heterocellular Organoids Xiao Qin1, Jahangir Sufi1*, Petra Vlckova1*, Pelagia Kyriakidou1*,

1

2

20

Organoid 1

Organoid 2

Organoid 20

TOBis 1

TOBis 2

TOBis 20

MassCytometry

×40Rare-EarthMetal Abs

Pooled Dissociation

High-Throughput Organoid Processing

...

= Organoid 20

m/z

m/z

m/z

= Organoid 1

= Organoid 2

Debarcode Single Cells

...

Thiol Organoid Barcoding in situ (TOBis)

d e

TeMal

Cisplatin

ThiolReactive

20-plex TOBis(6-choose-3)

124Te126Te128Te130Te

196Pt198PtPt

NH3

NH3Cl

Cl

TeO

NHN

O

O

ex situin situ

102 103 104 1051010102 103 104 1051010

102 103 104 1051010102 103 104 1051010

99% 9%

90% 100%

a

Amine Barcodes Bind Matrigel

Thiol Barcodes Bind Organoids

NH2 NH2

NH2NH2

MalGd Mal

Gd

MalGd

HN

NH HN

NH

NHSGd

NHSGd

NHSGd

NHSGd

SH

NH2

SH

NH2

SH

NH2

cb

C2 maleimide-DOTA-157Gd

NHS ester-DOTA-157Gd

N

N

N

N

O-

O O

O-

O-

OO-N

H

N

O

O

Gd

ThiolReactive

N

N

N

N

O-

O O

O-

O-

OO-O

N

O

O

GdAmine

Reactive

Amin

e Re

activ

eTh

iol R

eact

ive

GFP

GFPNHS ester-Alexa 647

C2 maleimide-Alexa 647157Gd

157Gd

Matrigel Organoid Media

Organoids Organoids

Fig. 3. Thiol-Reactive Organoid Barcoding in situ (TOBis) for Single-Cell Organoid Multiplexing. a) Confocal IF of fixed GFP+ small intestinal organoids stained witheither amine-reactive fluorescent probe Alexa Fluor 647 NHS ester or thiol-reactive fluorescent probe Alexa Fluor 647 C2 maleimide while still in Matrigel, scale bars =50 µm. Amine-reactive probes bind diffusely to Matrigel with very poor binding to organoids, whereas thiol-reactive probes bypass Matrigel and bind directly to organoids(see Supplementary Fig. 4c for Matrigel background staining). b) Small intestinal organoids stained with either amine-reactive NHS ester-DOTA-157Gd or thiol-reactive C2

maleimide-DOTA-157Gd in situ (still in Matrigel) or ex situ (removed from Matrigel) and analysed by MC. While both probes bind organoid cells ex situ, only thiol-reactive C2

maleimide-DOTA-157Gd bind organoids in situ. c) Model of amine- and thiol-reactive barcodes in organoid culture. d) Thiol-reactive tellurium maleimide (TeMal) (124Te, 126Te,128Te, 130Te) and Cisplatin (196Pt, 198Pt) isotopologs combined to form a 20-plex (6-choose-3) doublet-filtering barcoding strategy. e) Thiol-Reactive Organoid Barcoding insitu (TOBis) workflow. When combined with cell-type, cell-state, and PTM probes, TOBis allows organoids to be barcoded while still in Matrigel and rapidly processed as asingle sample. (See Supplementary Fig. 5 for additional details.)

organoids while still in Matrigel (Fig. 3c). Using this knowl-edge, we developed a 20-plex (6-choose-3, doublet-filtering(25, 26)) thiol-reactive barcoding strategy based on monoiso-topic tellurium maleimide (TeMal) (27) (124Te, 126Te, 128Te,130Te) and Cisplatin (28) (196Pt, 198Pt) that can bypass Ma-trigel proteins and bind directly to fixed organoids in situ

(Fig. 3d and Supplementary Fig. 4d). This Thiol-reactiveOrganoid Barcoding in situ (TOBis) approach enables high-throughput multivariate single-cell organoid signalling anal-ysis in a single tube (Fig. 3e).

It is worth noting that as Te and Pt metals are not typicallyconjugated to antibodies in MC, TOBis multiplexing does notcompromise the number of antigens being measured. More-over, unlike commercially available Pd barcodes (occupy-ing 102Pd, 104Pd, 105Pd, 106Pd, 108Pd, and 110Pd channels),TOBis barcodes do not clash with the recently developedCadmium antibody labelling metals (106Cd, 110Cd, 111Cd,112Cd, 113Cd, 114Cd, and 116Cd). Furthermore, as barcod-ing is performed on fixed organoids embedded within Ma-trigel, TOBis does not require the numerous centrifugationor cell membrane permeabilisation steps used in traditionalsolution-phase barcoding. This greatly increases organoidsample-throughput (Supplementary Fig. 5a–d) and single-cell recovery (Supplementary Fig. 5e–g), thereby facilitating

high-throughput organoid MC applications.

Multivariate Cell-Type Specific Signalling Analysis ofIntestinal Organoid Development. Traditional mass-tagbarcoding allows direct comparison of solution-phase cells(e.g. immune cells) between experimental conditions (26).When combined with cell-type, cell-state, and PTM probes,TOBis multiplexing now enables PTM signalling networksto be directly compared between organoid cultures in a high-throughput manner. To demonstrate this, we applied TOBis

to study cell-type specific epithelial signalling during 7 daysof small intestinal organoid development (Fig. 4 and Supple-mentary Table 1, 50 parameters (40 antibodies) / cell).Analysis of 28 PTMs from 2 million single organoid cellsrevealed that after 1 day of culture, organoids seeded as sin-gle crypts exist in a ‘recovery’ phase where 70% cells haveentered the cell-cycle (pRB+), but <5% reach S-phase (IdU+)(compared to 20% in developed organoids) (Fig. 4a). Days2 and 3 mark a rapid ‘expansion and differentiation’ phaseof organoid development where stem, Paneth, goblet cells,and enterocytes activate MAPK, P38, and PI3K pathways –although stem cells again fail to inhibit BAD (Fig. 4c). ByDay 4, intestinal organoids reach a critical ‘divergence’ phasewhere crypt and villus signalling digress dramatically. While

Qin et al. | Single-Cell Organoid Signalling bioR‰iv | 5

All rights reserved. No reuse allowed without permission. The copyright holder for this preprint (which was not peer-reviewed) is the author/funder.. https://doi.org/10.1101/659896doi: bioRxiv preprint

Page 6: Single-Cell Signalling Analysis of Heterocellular Organoids · Single-Cell Signalling Analysis of Heterocellular Organoids Xiao Qin1, Jahangir Sufi1*, Petra Vlckova1*, Pelagia Kyriakidou1*,

a

c

EdUcCas3

TOBis 1 TOBis 2 TOBis 3 TOBis 4 TOBis 5 TOBis 6 TOBis 7

Day 1 Day 2 Day 3 Day 4 Day 5 Day 6 Day 7

RecoveryPhase: Expansion + Differentiation Stem Survival + Enterocyte Exhaustion

IdUcCas3

StemEntero.

UMAP1

UMAP

2

Divergence

Sing

le-Ce

ll Den

sitypRB

b

% C

ell-T

ype

Entero.

Stem

PanethEndo.TuftGoblet

1 2 3 6 74 5Day:

Up-Reg.in Cell-Type

Down-Reg.in Cell-Type

stem_d

ay−1 ste

m_day

−2 stem_d

ay−3 ste

m_day

−4 stem_d

ay−5 ste

m_day

−6 stem_d

ay−7

<−1.

60.

70.

2>1

.2va

lue

142N

d_cC

aspa

se 3

168E

r_pS

MAD

2_3

161D

y_pB

AD

147S

m_p

BTK1

165H

o_Be

ta−C

aten

in_A

ctive

166E

r_pG

SK3b

143N

d_C−M

YC

157G

d_pM

KK3_

6

170E

r_pM

EK1_

2

146N

d_pM

KK4_

SEK1

154S

m_p

SMAD

1_5_

9

155G

d_pA

KT S

473

156G

d_pN

F−kB

152S

m_p

AKT

T308

172Y

b_pS

6

148N

d_pS

RC

163D

y_pP

90R

SK

167E

r_pE

RK1

_2

153E

u_pC

REB

141P

r_pP

DK1

151E

u_pP

KCa

160G

d_pA

MPK

a

164D

y_pP

120−

Cat

enin

150N

d_pR

B

149S

m_p

4E−B

P1

158G

d_pP

38

159T

b_pM

APKA

PK2 0

24

6

< -1.5

> 1.5

0

EMD

1 2 3 7EnterocyteStem

6 74 5Paneth Enteroendo. Tuft Goblet

6 74 51 2 3 6 74 51 2 36 74 51 2 36 74 51 2 3 64 51 2 3

Mitotic Post-Mitotic

Divergent SignallingStable Signalling

pPDPK1 [S241]

pMKK4 [S257]pBTK [Y511]

pSRC [Y418]

p4E-BP1 [T37/T46]

pPKCɑ [T497]

pAKT [T308]

pCREB [S133]

pAKT [S473]

pNF-κB p65 [S529]

pP38 MAPK [T180/Y182]pMAPKAPK2 [T334]

pAMPKα [T172]

pBAD [S112]

pP90RSK [T359]

pP120-Catenin [T310]

β-Catenin [Non-phospho] pGSK-3β [S9]

pERK1/2 [T202/Y204]

pMEK1/2 [S221]

pS6 [S235/S236]

pSMAD1/5/9 [S463/S465]pSMAD2/3 [S465/S467]

P38

PI3K

MAPK

NF-κB

TGF-β/BMP

WNTJNK

PTM

s

MG2SG1G0ApCell-State

Day:

Fig. 4. Cell-Type Specific Signalling During Intestinal Organoid Development. a) Time-course confocal IF of intestinal organoid development illustrating S-phase (EdU+,magenta) and apoptotic (cCaspase 3 [D175]+, green) cells, scale bars = 50 µm. Each time point was barcoded by TOBis, pooled into a single sample, and analysed byMC. Cell-density UMAP distributions of 2 million single organoid cells reveal changes in cell-type and cell-state during organoid development. b) Cell-type composition ofsmall intestinal organoids during development. Stem cells accumulate at the expense of enterocytes during organoid culture. c) Cell-type specific PTMs and cell-states ofstem, Paneth, enteroendocrine, tuft, goblet cells, and enterocytes during intestinal organoid development. Cell-state analysis shows the proportions of apoptotic, G0-, G1-,S-, G2-, and M-phase cells. Irrespective of time point, stem, Paneth, and enteroendocrine cells are stably mitotic, whereas tuft, goblet cells, and enterocytes are frequentlypost-mitotic. Stem, Paneth, enteroendocrine, and tuft cells display stable signalling over time, whereas goblet cell- and enterocyte-signalling diverge from Day 4.

stem and Paneth cells maintain active MAPK, P38, and PI3Kpathways, enterocytes lose major PI3K (pPDPK1, p4E-BP1,pS6 [S235/S236], pAMPK– [T172], pSRC, and pPKC–) andP38 (pP38 MAPK, pMAPKAPK2 [T334], and pCREB) ac-tivity (Fig. 4c). As a result, by Days 5 to 7, enterocytes arelargely post-mitotic or apoptotic (pRB- / cCaspase3+), withhigh TGF-— signalling, whereas stem cells retain mitogenicflux and cell-cycle activity (Fig. 4c). Consequently, stem cellnumber increases while enterocytes become exhausted at theend of intestinal organoid development (Fig. 4a, b). Notably,both stem and Paneth cells continue to display high MAPK,P38, and PI3K activity even at this late stage of organoid cul-ture (Fig. 4c). This suggests that maintaining a stable sig-nalling flux is a core feature of intestinal crypt cells. In con-trast, tuft cells display high TGF-— signalling, low MAPK/ P38 / PI3K activity, and low cell-cycle activity throughoutorganoid development (Fig. 4c). This implies that irrespec-tive of organoid age, tuft cells shut down mitotic signallingpathways and terminally exit the cell cycle once differenti-ated. Such variations in organoid cell-state (Fig. 4a), cell-type (Fig. 4b), and PTM activity (Fig. 4c) suggest develop-

mental stage should be carefully considered when perform-ing organoid experiments. Collectively, this analysis revealedcell-type specific PTM signalling during intestinal organoiddevelopment and confirmed that TOBis can be used to per-form multivariate single-cell signalling analysis of heteroge-nous organoid cultures.

Single-Cell Signalling Analysis of CRC TMEOrganoids. We have demonstrated how a modifiedMC workflow enables high-throughput comparison ofcell-type specific signalling networks in epithelial organoids.Given that MC can theoretically resolve any cell-type, wenext expanded this platform to study PTM signalling inheterocellular organoid co-culture models of CRC.CRC develops through successive oncogenic mutations – fre-quently resulting in loss of APC activity, activation of KRAS,and perturbation of TP53 (29). In addition to oncogenic mu-tations, stromal fibroblasts (30, 31) and macrophages (32) inthe TME have also emerged as major drivers of CRC (33).While the underlying driver mutations of CRC have beenwell studied, how they dysregulate epithelial signalling rel-

6 | bioR‰iv Qin et al. | Single-Cell Organoid Signalling

All rights reserved. No reuse allowed without permission. The copyright holder for this preprint (which was not peer-reviewed) is the author/funder.. https://doi.org/10.1101/659896doi: bioRxiv preprint

Page 7: Single-Cell Signalling Analysis of Heterocellular Organoids · Single-Cell Signalling Analysis of Heterocellular Organoids Xiao Qin1, Jahangir Sufi1*, Petra Vlckova1*, Pelagia Kyriakidou1*,

-20 0 20 40

-20

0

20

WT WT

WT WT

A

A

A

AAK

AKP

AKPAK

AK AKPAKPAK

-5 0 5 10-5

0

5

Colonic Organoid

FibroblastMacrophage

Organoid Genotype

1

—+

—++

——+

+++

+—+

—++

——+

+++

+—+

—++

——+

+++

+—+

—++

——+

+++

++—

———

+2 3 4 5 6 7 8 9 10

Microenvironment

11 12 13 14 15 16 17 18

+—

+19

StromalControls

TOBis(6-choose-3)

a b Organoid(Pan-CK)

Fibroblast(RFP)

Macrophage(CD45)

TOBis 4

ApcWT

KrasWT

Trp53WT

shApcKrasWT

Trp53WT

shApc KrasG12D/+

Trp53WT

shApc KrasG12D/+

Trp53R172H/-

cOrganoid FibroblastMacrophage

TOBis 4

UMAP1

UMAP

2

Mass Cytometry (28 PTMs / Cell)

pRB+

cC3+

d Organoid PTM Nodes (EMD)

PC 1 (52% Variance)

PC 2

(26%

Var

ianc

e)

WT

WT

WT

WT

AA

A

A

AK

AK

AKAK

AKP

AKP

AKP

AKP

Genotype

+Fibroblast

+Macrophage

e

Organoid PTM Connectivity (DREMI)

PC 1 (44% Variance)

PC 2

(24%

Var

ianc

e)

Genotype

Genotype

Genotype

f

Up-Reg.in Organoid

Down-Reg.in Organoid

stem_d

ay−1 ste

m_day

−2 stem_d

ay−3 ste

m_day

−4 stem_d

ay−5 ste

m_day

−6 stem_d

ay−7

<−1.

60.

70.

2>1

.2va

lue

142N

d_cC

aspa

se 3

168E

r_pS

MAD

2_3

161D

y_pB

AD

147S

m_p

BTK1

165H

o_Be

ta−C

aten

in_A

ctive

166E

r_pG

SK3b

143N

d_C−M

YC

157G

d_pM

KK3_

6

170E

r_pM

EK1_

2

146N

d_pM

KK4_

SEK1

154S

m_p

SMAD

1_5_

9

155G

d_pA

KT S

473

156G

d_pN

F−kB

152S

m_p

AKT

T308

172Y

b_pS

6

148N

d_pS

RC

163D

y_pP

90R

SK

167E

r_pE

RK1

_2

153E

u_pC

REB

141P

r_pP

DK1

151E

u_pP

KCa

160G

d_pA

MPK

a

164D

y_pP

120−

Cat

enin

150N

d_pR

B

149S

m_p

4E−B

P1

158G

d_pP

38

159T

b_pM

APKA

PK2 0

24

6

< -1.5

> 1.5

0

EMD(Nodes)

pPDPK1 [S241]

pMKK4 [S257]

pBTK [Y511]

pSRC [Y418]

p4E-BP1 [T37/T46]

pPKCɑ [T497]

pAKT [T308]

pCREB [S133]

pAKT [S473]

pNF-κB p65 [S529]

pP38 MAPK [T180/Y182]pMAPKAPK2 [T334]

pAMPKα [T172]

pBAD [S112]

pP90RSK [T359]

pP120-Catenin [T310]

β-Catenin [Non-phospho] pGSK-3β [S9]

pERK1/2 [T202/Y204]pMEK1/2 [S221]

pS6 [S235/S236]

pSMAD1/5/9 [S463/S465]pSMAD2/3 [S465/S467]

P38

PI3K

MAPK

NF-κB

TGF-β/BMPWNT

JNK

Cell-State

PTMs

pMKK3/6 [S189/S207]

pSTAT3 [Y705]

Organoid Genotype:Microenvironment:

WT A AK AKP

MG2SG1G0Ap

LRIG1 [Stem]CD44 [Crypt]Cell-Type

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16TOBis

Fig. 5. Single-Cell Signalling Analysis of Colorectal Cancer (CRC) Tumour Microenvironment Organoids. a) Experimental design. CRC organoid genotypes (wild-type(WT), shApc (A), shApc and KrasG12D/+ (AK), shApc, KrasG12D/+, and Trp53R172H/- (AKP)) were cultured in the presence or absence of colonic fibroblasts and/or macrophages(without exogenous growth factors). Each condition was TOBis-barcoded, pooled into a single sample, and analysed by MC (28 PTMs / cell). b) Confocal IF of a WTcolonic organoid (Pan-CK, green) co-cultured with colonic fibroblasts (RFP, red), and macrophages (CD45, grey) (TOBis 4), scale bar = 50 µm. c) UMAP distribution of thecolonic microenvironment model resolves single epithelial cells (green), fibroblasts (red), and macrophages (grey) (TOBis 4). d) PTMs, progenitor cell-types, and cell-statesof colonic epithelial organoids across all genotype / microenvironment combinations. The grey and red shades in the microenvironmental conditions represent macrophagesand fibroblasts respectively. (See Supplementary Figs. 7 and 8 for complete EMD-DREMI signalling maps of organoids, macrophages, and colonic fibroblasts.) e) PCA of 28PTM-EMDs for colonic epithelial organoids across all genotype / microenvironment combinations. CRC organoids with AK / AKP mutations mimic the signalling flux driven bycolonic fibroblasts. (See Supplementary Fig. 8c, d for PTM-EMD PCAs for macrophages and colonic fibroblasts.) f) PCA of 756 PTM-DREMIs for colonic epithelial organoidsacross all genotype / microenvironment combinations. Epithelial signalling connectivity is regulated by genotype rather than microenvironment. (See Supplementary Fig. 8e,f for PTM-DREMI PCAs for macrophages and colonic fibroblasts.)

ative to microenvironmental cues from stromal and immunecells is unclear.To investigate this, we cultured wild-type (WT), shApc (A),shApc and Kras

G12D/+ (AK), or shApc, KrasG12D/+, and

Trp53R172H/- (AKP) (34, 35) colonic epithelial organoids

either alone, with colonic fibroblasts, and/or macrophages(Fig. 5a, b, and Supplementary Fig. 6). Each CRCgenotype-microenvironment organoid culture was fixed,TOBis-barcoded, and single-cell signalling analysis was per-formed in one multivariate MC run (Fig. 5a and Sup-plementary Table 2, 50 parameters (40 antibodies) / cell).Addition of myeloid (CD68 and F4/80) and mesenchymal(Podoplanin) heavy-metal antibodies enabled clear resolu-tion of epithelial organoids, macrophages, and fibroblastsfrom each barcoded condition (Fig. 5c). This experimen-tal design allowed us to directly compare mutation- andmicroenvironment-driven cell-type specific signalling net-works in CRC organoid mono- and co-cultures.As expected, oncogenic mutations have a large cell-autonomous effect on epithelial signalling. Although APC

mutations are well known to upregulate WNT signalling(22), we found that the loss of APC also activates the P38pathway (pP38 MAPK and pMAPKAPK2), downregulatesTGF-— / BMP signalling (pSMAD2/3 and pSMAD1/5/9),and activates p120-Catenin in colonic organoids (Fig. 5d).Subsequent oncogenic Kras

G12D/+ and Trp53R172H/- muta-

tions further cell-autonomously upregulate not only the clas-sical MAPK pathway, but also major PI3K nodes (pPDPK1,pAKT, pS6, and p4E-BP1) (Fig. 5d). As a result, AKand AKP organoids display increased stem / progenitor cell-type markers LRIG1 and CD44, decreased apoptosis, and in-creased mitogenic cell-state relative to WT and A organoids(Fig. 5d).Both oncogenes and stromal cells can dysregulate cancercell signalling (7). However, to what extent this is drivenby oncogenic mutations (cell-intrinsic) or the TME (cell-extrinsic) is less clear. To investigate this, we directly com-pared mutation- and microenvironment-driven signalling inCRC organoids. To our surprise, we found that microenvi-ronmental cues have a comparable impact on PTM regula-

Qin et al. | Single-Cell Organoid Signalling bioR‰iv | 7

All rights reserved. No reuse allowed without permission. The copyright holder for this preprint (which was not peer-reviewed) is the author/funder.. https://doi.org/10.1101/659896doi: bioRxiv preprint

Page 8: Single-Cell Signalling Analysis of Heterocellular Organoids · Single-Cell Signalling Analysis of Heterocellular Organoids Xiao Qin1, Jahangir Sufi1*, Petra Vlckova1*, Pelagia Kyriakidou1*,

tion to oncogenic mutations (Fig. 5e). In contrast, while mu-tations and stromal cells can both drive epithelial PTM ac-tivity, PTM-PTM connectivity is regulated largely by geno-type, not microenvironment (Fig. 5f). This observation sug-gests that oncogenic mutations fundamentally re-wire sig-nalling networks, whereas stromal cells regulate acute sig-nalling flux. We also found that stromal cells further upreg-ulate the PI3K pathway (pS6, p4E-BP1, and pAKT) in CRCorganoids that already contain Kras

G12D and Trp53R172H mu-

tations (Fig. 5d and Supplementary Fig. 7). Microenviron-mental hyper-activation of the epithelial PI3K pathway maycontribute towards the poor prognosis of CRC patients withhighly stromal tumours (30, 31).In addition to mutation- and microenvironment-driven ep-ithelial signalling, we discovered previously unreported po-larity in fibroblast and macrophage cell-cell communica-tion. For example, macrophage signalling pathways (MAPK,PI3K, and NF-ŸB) are heavily upregulated by fibroblasts(Supplementary Fig. 8a, c, e), whereas fibroblast signallingis scarcely altered by macrophages (Supplementary Fig. 8b,d, f). In contrast, epithelial cells upregulate MAPK and P38signalling in fibroblasts, which in turn, reciprocally activateMAPK and P38 signalling in epithelial cells (SupplementaryFig. 7 and 8b). These results suggest that colonic fibrob-lasts are major regulators of intercellular signalling in thecolonic microenvironment and should be further investigatedas drivers of CRC.

Oncogenic Mutations Mimic Stromal Signalling Net-works. Cell-type specific PCA of EMD-PTMs suggestedthat mutation- and microenvironment-driven signalling incolonic organoids are related (Fig. 5e). To further inves-tigate the parity between genotypic and microenvironmentalregulation of epithelial signalling, we overlaid single-cell MCdata from WT, A, AK, and AKP organoids onto a fixed-nodemicroenvironmental Scaffold map (36) constructed from WTcolonic organoids alone or co-cultured with colonic fibrob-lasts and/or macrophages (Fig. 6a and Supplementary Fig.9a). This unsupervised analysis confirmed that Apc, Kras,and Trp53 oncogenic mutations mimic the signalling profileof WT organoids in the presence of stromal cells. Invertedorganoid genotype Scaffold maps also expose a striking sim-ilarity between mutation- and microenvironment-driven sig-nalling (Supplementary Fig. 9b). Direct comparison oforganoid PTMs revealed that both PI3K / PKC (pPDPK1,pPKC–, pAKT, p4E-BP1, pS6, pSRC, pP120-Catenin, andpAMPK–) and P38 / MAPK (pP38 MAPK, pMAPKAPK2,pP90RSK, pCREB, and pBAD) nodes are analogously up-regulated by oncogenic mutations and microenvironmentalcues (Figs. 5d and 6b). Activation of these pathways byeither oncogenic mutations or stromal cells correlates withdecreased apoptosis and increased mitogenic cell-state incolonic organoids (Fig. 5d).Taken together, multivariate cell-type specific PTM analy-sis of organoid co-cultures elucidated several fundamentalprocesses in CRC: 1) oncogenic mutations re-structure sig-nalling networks in cancer cells, whereas microenvironmen-tal cues drive acute signalling flux, 2) stromal cells hyper-

activate PI3K signalling in colonic epithelial cells that al-ready carry Kras and Trp53 mutations, and 3) oncogenic mu-tations cell-autonomously mimic an epithelial signalling statenormally induced by stromal cells. These results collectivelyconfirmed that TOBis-multiplexed MC enables discoveries ofnovel cell-type specific signalling relationships between dif-ferent cell-types in organoid models of the tumour microen-vironment.

DISCUSSIONOrganoids are heterocellular systems that comprise multiplecell-types and cell-states. Cell-type specific PTM signallingnetworks regulate major biological processes and are fre-quently dysregulated in disease. As a result, understandingcell-type specific signalling networks is fundamental to theutility of organoids and organoid co-cultures. Existing bulkPTM technologies (e.g. LC-MS/MS and anti-phospho an-tibody arrays) cannot describe cell-type or cell-state specificsignalling relationships and therefore limit our understandingof organoid biology (9). While scRNA-seq can characterisecell-type specific transcription, it cannot measure protein-level signal transduction which ultimately drives biologicalphenotypes. To overcome these challenges, we demonstratedhow a modified MC workflow that combines monoisotopiccell-type, cell-state, and PTM probes can be used to studycell-type specific signalling networks in organoids. Thismethod uncovered novel cell-type specific signalling in in-testinal epithelia and revealed an intimate relationship be-tween cell-signalling and cell-state in organoids. We showedhow Thiol-reactive Organoid Barcoding in situ (TOBis) en-ables high-throughput comparison of signalling networksacross different organoid mono- and co-cultures. Applica-tion of this technology to CRC TME organoid co-culturesrevealed that oncogenic mutations mimic stromal signallingcues and demonstrated how highly mutated CRC cells can befurther dysregulated by fibroblasts and macrophages.While this study has focused on intestinal organoids, we ex-pect this method to be fully compatible with organoids de-rived from other tissues (e.g. brain, liver, pancreas, kidneyetc.). Cell-type identification probes for each tissue shouldbe carefully validated, but otherwise the TOBis multiplex-ing and PTM analysis framework we report should be com-patible with all organoid models (including those grown indefined hydrogels (37)). Moreover, our extension of MCto study colonic fibroblasts and macrophages implies thatPTM signalling can be measured in any cell-type co-culturedwith organoids (e.g. PBMCs co-cultured with organoids (3)and air-liquid interface tumour microenvironment organoids(38)).In addition to standard single-cell organoid signalling exper-iments, the new barcoding technology reported here holdssubstantial promise for organoid screening. While drugscreens of patient-derived organoid (PDO) monocultureshave shown great potential (39, 40), their reliance on bulk vi-ability measurements (e.g. CellTiter-Blue) implies that theycannot be used to evaluate drugs targeting stromal and/orimmune cells or provide any mechanistic understanding of

8 | bioR‰iv Qin et al. | Single-Cell Organoid Signalling

All rights reserved. No reuse allowed without permission. The copyright holder for this preprint (which was not peer-reviewed) is the author/funder.. https://doi.org/10.1101/659896doi: bioRxiv preprint

Page 9: Single-Cell Signalling Analysis of Heterocellular Organoids · Single-Cell Signalling Analysis of Heterocellular Organoids Xiao Qin1, Jahangir Sufi1*, Petra Vlckova1*, Pelagia Kyriakidou1*,

a

TOBis 1: WT

TOBis 5: A

TOBis 9: AK

TOBis 13: AKP

bMicroenvironment Scaffold Map

Organoid

+Macrophage

+Fibroblast

+Macrophage+Fibroblast

Mic

roen

viro

nmen

tG

enot

ype

PI3K / PKC Pathway

S235 S236S6

T497PKCɑ

S112BAD

T37 T464E-BP1

S133CREB

T180 Y182p38 MAPK

S241PDPK1

T310p120-Catenin

Y418SRC

T172AMPKɑ

T308AKT

S473AKT

T359p90RSK

T334MAPKAPK2

S189MKK3/6

S207

Genotype Microenvironment

WT A AK AKP WT WT WT WT

P38 / MAPK Pathway

Genotype Microenvironment

Organoid PTM Nodes

WT A AK AKP WT WT WT WT

-1.5

-1.0

-0.5

0.0

0.5

-1.5

-1.0

-0.5

0.0

0.5

-0.6

-0.4

-0.2

0.0

0.2

0.4

-0.6

-0.4

-0.2

0.0

0.2

0.4

-0.4

-0.2

0.0

0.2

-0.4

-0.2

0.0

0.2

-1.0

-0.5

0.0

0.5

-1.0

-0.5

0.0

0.5

-0.4

-0.2

0.0

0.2

-0.4

-0.2

0.0

0.2

-0.5

0.0

0.5

1.0

-0.5

0.0

0.5

1.0

-0.1

0.0

0.1

0.2

0.3

0.4

-0.1

0.0

0.1

0.2

0.3

0.4

-0.5

0.0

0.5

-0.5

0.0

0.5

-0.2

0.0

0.2

0.4

-0.2

0.0

0.2

0.4

-0.1

0.0

0.1

0.2

0.3

0.4

-0.1

0.0

0.1

0.2

0.3

0.4

-0.5

0.0

0.5

-0.5

0.0

0.5

-0.5

0.0

0.5

-0.5

0.0

0.5

-0.4

0.0

0.4

-0.4

0.0

0.4

-0.2

0.0

0.2

0.4

0.6

-0.2

0.0

0.2

0.4

0.6

-0.5

0.0

0.5

-0.5

0.0

0.5

EMD

EMD

EMD

EMD

EMD

EMD

EMD

EMD

EMD

EMD

EMD

EMD

EMD

EMD

EMD

Fig. 6. Oncogenic Mutations Mimic Stromal Signalling Networks. a) Scaffold maps constructed from WT organoids either alone or co-cultured with colonic fibroblastsand/or macrophages. Unsupervised distribution of A, AK, and AKP colonic organoids revealed that oncogenic mutations mimic signalling profiles driven by stromal fibroblastsand macrophages. (See Supplementary Fig. 9a for all genotype / microenvironment combinations and Supplementary Fig. 9b for mutation-driven Scaffold maps.) b)PTM-EMDs for PI3K / PKC and P38 / MAPK signalling nodes in colonic organoids following genotypic and microenvironmental regulation. Cell-type specific PTM analysisdemonstrates oncogenic mutations and microenvironmental cues upregulate analogous signalling nodes in epithelial colonic organoids.

drug performance and/or resistance. In contrast, a TOBis-multiplexed MC drug / CRISPR screen will characterise cell-type specific signalling networks, cell-cycle states, and apop-totic readouts at single-cell resolution across all cell-types inPDO and PDO co-cultures. Given its ability to resolve multi-ple cell-types, TOBis MC would be particularly powerful forevaluating biological therapies against solid tumours wherecell-type specificity is essential for resolving drug (e.g. CART-cell) and target (organoid) phenotypes. Future develop-ment of TOBis barcodes using additional TeMal (×7 possi-ble) and Cisplatin (×4 possible) isotopologs will greatly ex-pand organoid multiplexing capacity and advance this tech-nology to high-throughput organoid screening applications.In summary, this study demonstrates how a modified MCplatform can reveal cell-type specific signalling networks inorganoid monocultures and uncover novel cell-cell signallingrelationships in organoid co-cultures. Given the widespreadadoption of organoids as biomimetic models of healthy and

diseased tissue, we propose cell-type specific PTM analysisas a powerful technology for multivariate organoid phenotyp-ing.

METHODSMethods, including statements of data availability and anyassociated accession codes and references, are available assupplementary information on bioRxiv.

ACKNOWLEDGEMENTSWe are extremely grateful to Dr. Lukas Dow (Cornell Univer-sity) for sharing CRC organoids, Dr. Xin Lu (University ofOxford) for sharing mouse intestines, and Dr. Olga Ornatsky(Fluidigm) for providing monoisotopic Cisplatin (195Pt and196Pt). We thank the UCL CI Flow-Core for MC supportand Dr. Leland McInnes (Tutte Institute) for UMAP advise.Graphical organoid renders were designed by Dr. Jeroen

Qin et al. | Single-Cell Organoid Signalling bioR‰iv | 9

All rights reserved. No reuse allowed without permission. The copyright holder for this preprint (which was not peer-reviewed) is the author/funder.. https://doi.org/10.1101/659896doi: bioRxiv preprint

Page 10: Single-Cell Signalling Analysis of Heterocellular Organoids · Single-Cell Signalling Analysis of Heterocellular Organoids Xiao Qin1, Jahangir Sufi1*, Petra Vlckova1*, Pelagia Kyriakidou1*,

Claus (Phospho Biomedical Animation). This work was sup-ported by Cancer Research UK (C60693/A23783), UCLHBiomedical Research Centre (BRC422), The Royal Society(RSG\Rd1\180234), and Rosetrees Trust (A1989).

AUTHOR CONTRIBUTIONSX.Q. Designed the study, performed organoid and MCexperiments, analysed the data, and wrote the paper.J.S. Developed TOBis, designed rare-earth metal antibodypanels, performed MC analysis, and analysed data.P.V. Isolated and characterised colonic fibroblasts,macrophages, cultured organoids, and analysed data.P.K. Performed UMAP, EMD, DREMI, and PCA dataanalysis.M.N. Developed TeMal barcodes.S.A. Provided murine monocytes and intestines for fibroblastisolation.V.L. Provided murine small intestines for organoid isolation.C.T. Designed the study, analysed the data, and wrote thepaper.

COMPETING INTERESTSM.N. has pending intellectual property on the use of telluriumreagents for mass cytometry applications which has been li-censed to Fluidigm Corporation.

REFERENCES1. H. Clevers. Modeling development and disease with organoids. Cell, 165(7):1586–1597,

2016. ISSN 1097-4172 (Electronic) 0092-8674 (Linking). doi: 10.1016/j.cell.2016.05.082.2. A. Pastula, M. Middelhoff, A. Brandtner, M. Tobiasch, B. Hohl, A. H. Nuber, I. E. Demir,

S. Neupert, P. Kollmann, G. Mazzuoli-Weber, and M. Quante. Three-dimensional gastroin-testinal organoid culture in combination with nerves or fibroblasts: A method to characterizethe gastrointestinal stem cell niche. Stem Cells Int, 2016:3710836, 2016. ISSN 1687-966X(Print). doi: 10.1155/2016/3710836.

3. K. K. Dijkstra, C. M. Cattaneo, F. Weeber, M. Chalabi, J. van de Haar, L. F. Fanchi,M. Slagter, D. L. van der Velden, S. Kaing, S. Kelderman, N. van Rooij, M. E. van Leerdam,A. Depla, E. F. Smit, K. J. Hartemink, R. de Groot, M. C. Wolkers, N. Sachs, P. Snaebjorns-son, K. Monkhorst, J. Haanen, H. Clevers, T. N. Schumacher, and E. E. Voest. Generationof tumor-reactive t cells by co-culture of peripheral blood lymphocytes and tumor organoids.Cell, 174(6):1586–1598 e12, 2018. ISSN 1097-4172 (Electronic) 0092-8674 (Linking). doi:10.1016/j.cell.2018.07.009.

4. D. Tuveson and H. Clevers. Cancer modeling meets human organoid technology. Science,364(6444):952–955, 2019. ISSN 1095-9203 (Electronic) 0036-8075 (Linking). doi: 10.1126/science.aaw6985.

5. T. Pawson and J. D. Scott. Protein phosphorylation in signaling–50 years and counting.Trends Biochem Sci, 30(6):286–90, 2005. ISSN 0968-0004 (Print) 0968-0004 (Linking).doi: 10.1016/j.tibs.2005.04.013.

6. K. Miller-Jensen, K. A. Janes, J. S. Brugge, and D. A. Lauffenburger. Common effectorprocessing mediates cell-specific responses to stimuli. Nature, 448(7153):604–8, 2007.ISSN 1476-4687 (Electronic) 0028-0836 (Linking). doi: 10.1038/nature06001.

7. C. J. Tape, S. Ling, M. Dimitriadi, K. M. McMahon, J. D. Worboys, H. S. Leong, I. C. Nor-rie, C. J. Miller, G. Poulogiannis, D. A. Lauffenburger, and C. Jorgensen. Oncogenic krasregulates tumor cell signaling via stromal reciprocation. Cell, 165(4):910–20, 2016. ISSN1097-4172 (Electronic) 0092-8674 (Linking). doi: 10.1016/j.cell.2016.03.029.

8. A. J. Simmons, A. Banerjee, E. T. McKinley, C. R. Scurrah, C. A. Herring, L. S. Gewin,R. Masuzaki, S. J. Karp, J. L. Franklin, M. J. Gerdes, J. M. Irish, R. J. Coffey, and K. S. Lau.Cytometry-based single-cell analysis of intact epithelial signaling reveals mapk activationdivergent from tnf-alpha-induced apoptosis in vivo. Mol Syst Biol, 11(10):835, 2015. ISSN1744-4292 (Electronic) 1744-4292 (Linking). doi: 10.15252/msb.20156282.

9. C. J. Tape. Systems biology analysis of heterocellular signaling. Trends Biotechnol, 34(8):627–637, 2016. ISSN 1879-3096 (Electronic) 0167-7799 (Linking). doi: 10.1016/j.tibtech.2016.02.016.

10. A. L. Haber, M. Biton, N. Rogel, R. H. Herbst, K. Shekhar, C. Smillie, G. Burgin, T. M.Delorey, M. R. Howitt, Y. Katz, I. Tirosh, S. Beyaz, D. Dionne, M. Zhang, R. Raychowdhury,W. S. Garrett, O. Rozenblatt-Rosen, H. N. Shi, O. Yilmaz, R. J. Xavier, and A. Regev. Asingle-cell survey of the small intestinal epithelium. Nature, 551(7680):333–339, 2017. ISSN1476-4687 (Electronic) 0028-0836 (Linking). doi: 10.1038/nature24489.

11. M. H. Spitzer and G. P. Nolan. Mass cytometry: Single cells, many features. Cell, 165(4):780–91, 2016. ISSN 1097-4172 (Electronic) 0092-8674 (Linking). doi: 10.1016/j.cell.2016.04.019.

12. S. C. Bendall, E. F. Simonds, P. Qiu, A. D. Amir el, P. O. Krutzik, R. Finck, R. V. Bruggner,R. Melamed, A. Trejo, O. I. Ornatsky, R. S. Balderas, S. K. Plevritis, K. Sachs, D. Pe’er,S. D. Tanner, and G. P. Nolan. Single-cell mass cytometry of differential immune and drugresponses across a human hematopoietic continuum. Science, 332(6030):687–96, 2011.ISSN 1095-9203 (Electronic) 0036-8075 (Linking). doi: 10.1126/science.1198704.

13. T. Sato, R. G. Vries, H. J. Snippert, M. van de Wetering, N. Barker, D. E. Stange, J. H. vanEs, A. Abo, P. Kujala, P. J. Peters, and H. Clevers. Single lgr5 stem cells build crypt-villusstructures in vitro without a mesenchymal niche. Nature, 459(7244):262–5, 2009. ISSN1476-4687 (Electronic) 0028-0836 (Linking). doi: 10.1038/nature07935.

14. G. K. Behbehani, S. C. Bendall, M. R. Clutter, W. J. Fantl, and G. P. Nolan. Single-cell masscytometry adapted to measurements of the cell cycle. Cytometry A, 81(7):552–66, 2012.ISSN 1552-4930 (Electronic) 1552-4922 (Linking). doi: 10.1002/cyto.a.22075.

15. H. G. Fienberg, E. F. Simonds, W. J. Fantl, G. P. Nolan, and B. Bodenmiller. A platinum-based covalent viability reagent for single-cell mass cytometry. Cytometry A, 81(6):467–75,2012. ISSN 1552-4930 (Electronic) 1552-4922 (Linking). doi: 10.1002/cyto.a.22067.

16. X. Yin, H. F. Farin, J. H. van Es, H. Clevers, R. Langer, and J. M. Karp. Niche-independenthigh-purity cultures of lgr5+ intestinal stem cells and their progeny. Nat Methods, 11(1):106–12, 2014. ISSN 1548-7105 (Electronic) 1548-7091 (Linking). doi: 10.1038/nmeth.2737.

17. M. A. Rapsomaniki, X. K. Lun, S. Woerner, M. Laumanns, B. Bodenmiller, and M. R.Martinez. Cellcycletracer accounts for cell cycle and volume in mass cytometry data.Nat Commun, 9(1):632, 2018. ISSN 2041-1723 (Electronic) 2041-1723 (Linking). doi:10.1038/s41467-018-03005-5.

18. T. H. Kim, F. Li, I. Ferreiro-Neira, L. L. Ho, A. Luyten, K. Nalapareddy, H. Long, M. Verzi, andR. A. Shivdasani. Broadly permissive intestinal chromatin underlies lateral inhibition andcell plasticity. Nature, 506(7489):511–5, 2014. ISSN 1476-4687 (Electronic) 0028-0836(Linking). doi: 10.1038/nature12903.

19. J. H. Levine, E. F. Simonds, S. C. Bendall, K. L. Davis, A. D. Amir el, M. D. Tadmor, O. Litvin,H. G. Fienberg, A. Jager, E. R. Zunder, R. Finck, A. L. Gedman, I. Radtke, J. R. Downing,D. Pe’er, and G. P. Nolan. Data-driven phenotypic dissection of aml reveals progenitor-likecells that correlate with prognosis. Cell, 162(1):184–97, 2015. ISSN 1097-4172 (Electronic)0092-8674 (Linking). doi: 10.1016/j.cell.2015.05.047.

20. D. Y. Orlova, N. Zimmerman, S. Meehan, C. Meehan, J. Waters, E. E. Ghosn, A. Fi-latenkov, G. A. Kolyagin, Y. Gernez, S. Tsuda, W. Moore, R. B. Moss, L. A. Herzenberg,and G. Walther. Earth mover’s distance (emd): A true metric for comparing biomarker ex-pression levels in cell populations. PLoS One, 11(3):e0151859, 2016. ISSN 1932-6203(Electronic) 1932-6203 (Linking). doi: 10.1371/journal.pone.0151859.

21. S. Krishnaswamy, M. H. Spitzer, M. Mingueneau, S. C. Bendall, O. Litvin, E. Stone, D. Pe’er,and G. P. Nolan. Systems biology. conditional density-based analysis of t cell signaling insingle-cell data. Science, 346(6213):1250689, 2014. ISSN 1095-9203 (Electronic) 0036-8075 (Linking). doi: 10.1126/science.1250689.

22. R. Nusse and H. Clevers. Wnt/beta-catenin signaling, disease, and emerging therapeuticmodalities. Cell, 169(6):985–999, 2017. ISSN 1097-4172 (Electronic) 0092-8674 (Linking).doi: 10.1016/j.cell.2017.05.016.

23. H. Gehart and H. Clevers. Tales from the crypt: new insights into intestinal stem cells. NatRev Gastroenterol Hepatol, 16(1):19–34, 2019. ISSN 1759-5053 (Electronic) 1759-5045(Linking). doi: 10.1038/s41575-018-0081-y.

24. J. Massague. Tgfbeta signalling in context. Nat Rev Mol Cell Biol, 13(10):616–30, 2012.ISSN 1471-0080 (Electronic) 1471-0072 (Linking). doi: 10.1038/nrm3434.

25. B. Bodenmiller, E. R. Zunder, R. Finck, T. J. Chen, E. S. Savig, R. V. Bruggner, E. F. Si-monds, S. C. Bendall, K. Sachs, P. O. Krutzik, and G. P. Nolan. Multiplexed mass cytometryprofiling of cellular states perturbed by small-molecule regulators. Nat Biotechnol, 30(9):858–67, 2012. ISSN 1546-1696 (Electronic) 1087-0156 (Linking). doi: 10.1038/nbt.2317.

26. E. R. Zunder, R. Finck, G. K. Behbehani, A. D. Amir el, S. Krishnaswamy, V. D. Gonzalez,C. G. Lorang, Z. Bjornson, M. H. Spitzer, B. Bodenmiller, W. J. Fantl, D. Pe’er, and G. P.Nolan. Palladium-based mass tag cell barcoding with a doublet-filtering scheme and single-cell deconvolution algorithm. Nat Protoc, 10(2):316–33, 2015. ISSN 1750-2799 (Electronic)1750-2799 (Linking). doi: 10.1038/nprot.2015.020.

27. L. M. Willis, H. Park, M. W. L. Watson, D. Majonis, J. L. Watson, and M. Nitz. Tellurium-based mass cytometry barcode for live and fixed cells. Cytometry A, 2018. ISSN 1552-4930(Electronic) 1552-4922 (Linking). doi: 10.1002/cyto.a.23495.

28. R. L. McCarthy, D. H. Mak, J. K. Burks, and M. C. Barton. Rapid monoisotopic cisplatinbased barcoding for multiplexed mass cytometry. Sci Rep, 7(1):3779, 2017. ISSN 2045-2322 (Electronic) 2045-2322 (Linking). doi: 10.1038/s41598-017-03610-2.

29. Network Cancer Genome Atlas. Comprehensive molecular characterization of human colonand rectal cancer. Nature, 487(7407):330–7, 2012. ISSN 1476-4687 (Electronic) 0028-0836(Linking). doi: 10.1038/nature11252.

30. A. Calon, E. Lonardo, A. Berenguer-Llergo, E. Espinet, X. Hernando-Momblona, M. Iglesias,M. Sevillano, S. Palomo-Ponce, D. V. Tauriello, D. Byrom, C. Cortina, C. Morral, C. Barcelo,S. Tosi, A. Riera, C. S. Attolini, D. Rossell, E. Sancho, and E. Batlle. Stromal gene expres-sion defines poor-prognosis subtypes in colorectal cancer. Nat Genet, 47(4):320–9, 2015.ISSN 1546-1718 (Electronic) 1061-4036 (Linking). doi: 10.1038/ng.3225.

31. C. Isella, A. Terrasi, S. E. Bellomo, C. Petti, G. Galatola, A. Muratore, A. Mellano, R. Senetta,A. Cassenti, C. Sonetto, G. Inghirami, L. Trusolino, Z. Fekete, M. De Ridder, P. Cassoni,G. Storme, A. Bertotti, and E. Medico. Stromal contribution to the colorectal cancer tran-scriptome. Nat Genet, 47(4):312–9, 2015. ISSN 1546-1718 (Electronic) 1061-4036 (Link-ing). doi: 10.1038/ng.3224.

32. J. Lan, L. Sun, F. Xu, L. Liu, F. Hu, D. Song, Z. Hou, W. Wu, X. Luo, J. Wang, X. Yuan, J. Hu,and G. Wang. M2 macrophage-derived exosomes promote cell migration and invasion incolon cancer. Cancer Res, 2018. ISSN 1538-7445 (Electronic) 0008-5472 (Linking). doi:10.1158/0008-5472.CAN-18-0014.

33. C. J. Tape. The heterocellular emergence of colorectal cancer. Trends Cancer, 3(2):79–88,2017. ISSN 2405-8033 (Print) 2405-8025 (Linking). doi: 10.1016/j.trecan.2016.12.004.

34. L. E. Dow, K. P. O’Rourke, J. Simon, D. F. Tschaharganeh, J. H. van Es, H. Clevers, andS. W. Lowe. Apc restoration promotes cellular differentiation and reestablishes crypt home-ostasis in colorectal cancer. Cell, 161(7):1539–1552, 2015. ISSN 1097-4172 (Electronic)0092-8674 (Linking). doi: 10.1016/j.cell.2015.05.033.

10 | bioR‰iv Qin et al. | Single-Cell Organoid Signalling

All rights reserved. No reuse allowed without permission. The copyright holder for this preprint (which was not peer-reviewed) is the author/funder.. https://doi.org/10.1101/659896doi: bioRxiv preprint

Page 11: Single-Cell Signalling Analysis of Heterocellular Organoids · Single-Cell Signalling Analysis of Heterocellular Organoids Xiao Qin1, Jahangir Sufi1*, Petra Vlckova1*, Pelagia Kyriakidou1*,

35. K. P. O’Rourke, E. Loizou, G. Livshits, E. M. Schatoff, T. Baslan, E. Manchado, J. Simon,P. B. Romesser, B. Leach, T. Han, C. Pauli, H. Beltran, M. A. Rubin, L. E. Dow, and S. W.Lowe. Transplantation of engineered organoids enables rapid generation of metastaticmouse models of colorectal cancer. Nat Biotechnol, 35(6):577–582, 2017. ISSN 1546-1696 (Electronic) 1087-0156 (Linking). doi: 10.1038/nbt.3837.

36. M. H. Spitzer, P. F. Gherardini, G. K. Fragiadakis, N. Bhattacharya, R. T. Yuan, A. N. Hotson,R. Finck, Y. Carmi, E. R. Zunder, W. J. Fantl, S. C. Bendall, E. G. Engleman, and G. P. Nolan.Immunology. an interactive reference framework for modeling a dynamic immune system.Science, 349(6244):1259425, 2015. ISSN 1095-9203 (Electronic) 0036-8075 (Linking). doi:10.1126/science.1259425.

37. R. Cruz-Acuna, M. Quiros, A. E. Farkas, P. H. Dedhia, S. Huang, D. Siuda, V. Garcia-Hernandez, A. J. Miller, J. R. Spence, A. Nusrat, and A. J. Garcia. Synthetic hydrogelsfor human intestinal organoid generation and colonic wound repair. Nat Cell Biol, 19(11):1326–1335, 2017. ISSN 1476-4679 (Electronic) 1465-7392 (Linking). doi: 10.1038/ncb3632.

38. J. T. Neal, X. Li, J. Zhu, V. Giangarra, C. L. Grzeskowiak, J. Ju, I. H. Liu, S. H. Chiou,A. A. Salahudeen, A. R. Smith, B. C. Deutsch, L. Liao, A. J. Zemek, F. Zhao, K. Karlsson,L. M. Schultz, T. J. Metzner, L. D. Nadauld, Y. Y. Tseng, S. Alkhairy, C. Oh, P. Keskula,D. Mendoza-Villanueva, F. M. De La Vega, P. L. Kunz, J. C. Liao, J. T. Leppert, J. B. Sunwoo,C. Sabatti, J. S. Boehm, W. C. Hahn, G. X. Y. Zheng, M. M. Davis, and C. J. Kuo. Organoidmodeling of the tumor immune microenvironment. Cell, 175(7):1972–1988 e16, 2018. ISSN1097-4172 (Electronic) 0092-8674 (Linking). doi: 10.1016/j.cell.2018.11.021.

39. M. van de Wetering, H. E. Francies, J. M. Francis, G. Bounova, F. Iorio, A. Pronk, W. vanHoudt, J. van Gorp, A. Taylor-Weiner, L. Kester, A. McLaren-Douglas, J. Blokker, S. Jaksani,S. Bartfeld, R. Volckman, P. van Sluis, V. S. Li, S. Seepo, C. Sekhar Pedamallu, K. Cibulskis,S. L. Carter, A. McKenna, M. S. Lawrence, L. Lichtenstein, C. Stewart, J. Koster, R. Ver-steeg, A. van Oudenaarden, J. Saez-Rodriguez, R. G. Vries, G. Getz, L. Wessels, M. R.Stratton, U. McDermott, M. Meyerson, M. J. Garnett, and H. Clevers. Prospective derivationof a living organoid biobank of colorectal cancer patients. Cell, 161(4):933–45, 2015. ISSN1097-4172 (Electronic) 0092-8674 (Linking). doi: 10.1016/j.cell.2015.03.053.

40. G. Vlachogiannis, S. Hedayat, A. Vatsiou, Y. Jamin, J. Fernandez-Mateos, K. Khan,A. Lampis, K. Eason, I. Huntingford, R. Burke, M. Rata, D. M. Koh, N. Tunariu, D. Collins,S. Hulkki-Wilson, C. Ragulan, I. Spiteri, S. Y. Moorcraft, I. Chau, S. Rao, D. Watkins, N. Fo-tiadis, M. Bali, M. Darvish-Damavandi, H. Lote, Z. Eltahir, E. C. Smyth, R. Begum, P. A.Clarke, J. C. Hahne, M. Dowsett, J. de Bono, P. Workman, A. Sadanandam, M. Fassan,O. J. Sansom, S. Eccles, N. Starling, C. Braconi, A. Sottoriva, S. P. Robinson, D. Cun-ningham, and N. Valeri. Patient-derived organoids model treatment response of metastaticgastrointestinal cancers. Science, 359(6378):920–926, 2018. ISSN 1095-9203 (Electronic)0036-8075 (Linking). doi: 10.1126/science.aao2774.

Qin et al. | Single-Cell Organoid Signalling bioR‰iv | 11

All rights reserved. No reuse allowed without permission. The copyright holder for this preprint (which was not peer-reviewed) is the author/funder.. https://doi.org/10.1101/659896doi: bioRxiv preprint