DIFFERENTIATION
Unbiased transcription factor CRISPR screen
identifies ZNF800 as master repressor of
enteroendocrine differentiation
Lin Lin
1,2,3
*, Jeff DeMartino
2,3
, Daisong Wang
1,2
, Gijs J. F. van Son
2,3
, Reinier van der Linden
1,2
,
Harry Begthel
1,2
, Jeroen Korving
1,2
, Amanda Andersson-Rolf
1,2
, Stieneke van den Brink
1,2
,
Carmen Lopez-Iglesias
4
, Willine J. van de Wetering
4
, Aleksandra Balwierz
3
, Thanasis Margaritis
3
,
Marc van de Wetering
2,3
, Peter J. Peters
4
, Jarno Drost
2,3
, Johan H. van Es
1,2
, Hans Clevers
1,2,3
*
Enteroendocrine cells (EECs) are hormone-producing cells residing in the epithelium of stomach,
small intestine (SI), and colon. EECs regulate aspects of metabolic activity, including insulin levels,
satiety, gastrointestinal secretion, and motility. The generation of different EEC lineages is not
completely understood. In this work, we report a CRISPR knockout screen of the entire repertoire
of transcription factors (TFs) in adult human SI organoids to identify dominant TFs controlling
EEC differentiation. We discovered ZNF800 as a master repressor for endocrine lineage commitment,
which particularly restricts enterochromaffin cell differentiation by directly controlling an endocrine
TF network centered on PAX4. Thus, organoid models allow unbiased functional CRISPR screens
for genes that program cell fate.
E
nteroendocrine cells (EECs) are special-
ized epithelial cells of the intestinal tract
that, like all other epithelial cell lineages,
derive from regionally specified Lgr5
+
in-
testinal stem cells (ISCs) (15). Balanced
differentiation of EEC lineages from ISCs is
governed by a network of transcription fac-
tors (TFs) (612). RNA sequencing and organ-
oidtechnologyhaveilluminatedthetemporal
hierarchy of gene expression profiles during
EEC development (13, 14). By using a time-
resolved reporter allele of Neurog3 (master
regulator of endocrine development) (15, 16),
a real-time and lineage-specific map of mouse
EEC differentiation in vivo was constructed at
single celllevel resolution (12). Overexpres-
sion of NEUROG3 can generate endocrine
cells for functional characterization in human
pancreatic duct cells (17). Leveraging human
gut organoids, this strategy enabled profiling
of EEC subtypes along the proximal-distal
gastrointestinal axis (18). TFs known to specify
EEC subtypes in mice were further examined in
human organoids, revealing a predominantly
conserved regulatory mechanism downstream
of NEUROG3 activatio n. However , discrepanc ies
between mouse and human data were also ob-
served. The regulatory mechanisms upstream
of NEUROG3 and the endogenous repressive
factors that control NEU R O G 3 expression at
the adult stage remain largely unknown. In this
work, we established an organoid-based plat-
form in combination with high-throughput ge-
netic screening for the unbiased discovery of TFs
that govern human EEC lineage commitment.
Results
TFome-wide CRISPR knockout screen in
human SI organoids
Optimized human SI organoids exhibit the
spontaneous gener ation of all major cell line-
ages, including EEC subtypes, without TF over-
expression (19). For the current study, we
aimed to establish a CRISPR screening platform
spanningtheentirerepertoireoftranscription
factors (TFome) in human intestinal organoids
to allow for unbiased and systematic discov-
ery of TFs that govern cell lineage commitment
from adult LGR5
+
intestinal stem cells. We
used a CRISPR knockout library comprising
7210 single-guide RNAs (sgRNAs) targeting
1800 human TFs, alongside 100 negative con-
trols (2022). In the human ileum organoid
lineused,EECsweremarkedbyCHGA-IRES-
iRFP670, goblet cells by MUC2-mNeonGreen,
and Paneth cells by DEFA5-IRES-DsRed (fig.
S1A). We performed lentiviral-mediated library
integration in expanding SI organoids (Fig. 1A).
Library coverage was assessed by sequencing
the integrated sgRNAs, comparing the organ-
oids after transduction with the plasmid pool.
Fate separation of CHGA
+
EECs and MUC2
+
cells occurs early in secretory progenitors,
whereas DEFA5
+
Paneth cells emerge later from
MUC2
+
progenitors (19), illustrating sequential
fate decisions of ISCs (23, 24). We aimed to
track the early CHGA
+
/MUC2
+
binary fate
decision. After two-step organoid differentia-
tion (19), we analyzed bulk populations as well
as sorted EECs (CHGA
+
), goblet cells (MUC2
+
)
and the CHGA
/MUC2
/DEFA5
population
(fig. S1B). 0 to ~0.7% sgRNAs yielded zero
reads in bulk populations, indicating robust
coverage (fig. S2A). Normalized read-count
distribution and Pearson correlations demon-
strated high concordance between biological
replicates (fig. S2, B and C). sgRNAs that target
essential ISC genes [KLF5 (25) and TCF7L2/
TCF4 (2629)] were depleted, along with
generic cell fi tness g enes (POLR2L, MYC,
and RAD51) at both expansion and differen-
tiation culturing stages (b score < 1; FDR <
0.05 versus plasmid library) (fig. S2D and table
S1). Furthermore, sgRNAs targeting IRF2,an
interferon pathway TF, were highly enriched
upon differentiation, consistent with existing
literature (30, 31).
sgRNAs were then assessed in CHGA
+
EECs
and MUC2
+
goblet cells versus the triple
reporter negative cell population (Fig. 1B
and table S2). As expected, NEUROG3, SOX4,
and INSM1 sgRNAs were depleted in EECs
(8, 15, 16, 32). Additionally, NFIC, TEF, and
ZHX2 appeared essential for EEC commitment.
For each of these, clonal knockout organoids
revealed a mild yet significant reduction of
CHGA
+
EECs (Fig. 1, B and C, and fig. S3, A to
C). Conversely, GFI1 sgRNAs were enriched in
EECs (Fig. 1, B and C). Mouse Gfi1 is crucial
for goblet cell differentiation by suppression
of Neurog3-driven EEC cell fate (23, 33).
Indeed, GFI1 knockout in human organoids
abrogated goblet cell formation while increas-
ingEECnumbers(fig.S3,AandC,andfig.S14,
CandD).
ZNF800 represses EEC differentiation
The strongest repressor of EEC differentiation
in the screen, ZNF800,isaC
2
H
2
zinc-finger TF
of unknown function (Fig. 1, B and C) (34).
Human ZNF800 is broadly expressed, includ-
ing in the SI and colon epithelium (fig. S4, A
and B). ZNF800
/
organoids contained in-
creased EECs and strongly reduced goblet and
Paneth cell numbers (Fig. 1, D and E, and fig.
S4, C and G). Transmission electron micros-
copy confirmed the absence of goblet and
Paneth cells with the characteristic apical
secretory vesicles and the increase of EECs
with the basolateral secretory granules (Fig. 1F).
Similarly, ZNF800 knockout in colon organoid
lines from two different donors also resulted
in a significant increase in EECs (fig. S4, D to G).
We next performed single-cell RNA sequenc-
ing (scRNA-seq) to study the phenotype of
ZNF800
/
organoids. For equal representa-
tion, CHGA
+
cells were sorted from wild type
(WT) and ZNF800
/
organoids and pooled
with CHGA
cells from the same lines in a 1:4
ratio (Fig. 2A and fig. S5A). Major intestinal
celltypeswereidentifiedbygraph-basedclus-
tering analysis (Fig. 2B and fig. S5B). We
observed the expected reduction of goblet cells
RESEARCH
1
Hubrecht Institute, Royal Netherlands Academy of Arts and
Sciences (KNAW) and UMC Utrecht, Utrecht, Netherlands.
2
Oncode Institute, Utrecht, Netherlands.
3
Princess Maxima
Center for Pediatric Oncology, Utrecht, Netherlands.
4
The
Maastricht Multimodal Molecular Imaging Institute,
Maastricht University, Maastricht, Netherlands.
*Corresponding author. Email: [email protected] (L.L.);
Current address: Pharma Research and Early Development of
F. HoffmannLa Roche Ltd., Basel, Switzerland.
Lin et al., Science 382, 451458 (2023) 27 October 2023 1of8
Downloaded from https://www.science.org on December 15, 2023
D
B
EEC
(CHGA
+
)
Goblet Cell
(MUC2
+
)
Paneth Cell
(DEFA5
+
)
0
5
10
15
Wild type
ZNF800
-/-
MUC2
DEFA5
CHGA
Human ileum organoid
Cell Percentage (%)
WT
ZNF800
-/-
***
****
**
Library integration
Expansion
Patterning
Maturation
Differentiated stage
Reporter
negative
Sequencing
gRNA
Enrichment
Log
2
FoldChange(CHGA
+
or CHGA
)
Density
C
E
GFI1
INSM1
NEUROG3
NFIC
SOX4
TEF
ZHX2
ZNF800
−1
0
1
−1 0 1
MUC2
+
Goblet cells (β score)
CHGA
+
Enteroendocrine cells (β score)
Gene knockout enriched in EECs
Gene knockout depleted in EECs
0.0
0.2
0.4
0.6
TEF
ZHX2
NFIC
INSM1
NEUROG3
SOX4
GFI1
ZNF800
NO-TARGET
−4 −2 0 2
sgRNA enrichment
Sorting
F
Wild type ZNF800
-/-
*
*
*
*
*
**
*
*
A
Basolateral
Apical
Basolateral
Apical
Fig. 1. TFome-wide CRISPR screen for endocrine differentiation in human
SI organoids. (A) Schematic of TFome CRISPR screen. (B) Scatter plot of
enrichment b score of each TF gene in CHGA
+
EECs and MUC2
+
goblet cells
versus the triple reporternegative cell population. (C) Individual sgRNA
enrichment for genes of interest presented by log
2
-fold changes in CHGA
+
EECs
versus triple reporternegative cell population. Density plot (top) represents the
distribution of nontargeting sgRNAs. (D) Representative confocal images of
WT and ZNF800
/
human SI organoids. Representative marker genes for EECs
(CHGA, magenta), goblet cells (MUC2, green), and Paneth cells (DEFA5, red) are
highlighted by fluorescent reporters. Scale bars, 100 mm. (E) Proportion of
different differentiated cell types as determined by FACS analysis of the
respective reporters. Data are shown as mean ± SEM. **P < 0.01; ***P < 0.001;
****P < 0.0001 by multiple t tests with two-stage linear step-up procedure of
Benjamini, Krieger, and Yekutieli with Q = 5% and n =3.(F) Transmission
electron microscopy images of WT and ZNF800
/
human SI organoids. Goblet
and Paneth cells are indicated with asterisks in WT organoids, and EECs are
indicated with asterisks in ZNF800
/
organoids. Scale bars, 20 mm and
10 mm (zoom in).
RESEARCH | RESEARCH ARTICLE
Lin et al., Science 382, 451458 (2023) 27 October 2023 2of8
Downloaded from https://www.science.org on December 15, 2023
80% 20%
AB
WT
ZNF800
-/-
DOX
-
DOX
+
0
5
10
15
20
Serotonin secretion (ng/mL
)
****
***
80% 20%
CHGA CHGA
+
ISCs
TA1
TA2
Early Enterocytes
Enterocytes
Secretory progenitors
Goblet cells
Early EECs
Enterochromaffin cells
other EECs
WT ZNF800
C
G
Serotonin ELISA
0
25
50
75
100
WT
(n = 328)
ZNF800
-/-
(n = 490)
Percentage of cell clusters in
CHGA
+
cell population (%)
Early EECs
Enterochromaffin cells
Other EECs
12.5%
47.6%
39.9%
33.9%
62.9%
3.3%
tight TRE
3XFLAG
ZNF800 ORF
P2A T2AhPGK
BFP
PuroR rtTA
Doxycycline
E
MUC2 TagBFP CHGA DEFA5
DOX
-
DOX
+
ZNF800
DOX
DOX
+
F
D
EC cells - CHGA
EC cells - TPH1
L cells - GCG
L cells - PYY
N cells- NTS
D cells - SST
I cells - CCK
G cells - GAST
M/X cells - MLN
M/X cells - GHRL
0.00001
0.0001
0.001
0.01
0.1
1
10
WT
ZNF800
-/-
sorted CHGA
+
cells
ns
ns
****
***
***
***
**
Relative Gene Expression
(vs GAPDH
)
n.d. n.d. n.d.
010 010
−4
0
4
UMAP_1
UMAP_2
Paneth cells
ISC
T
A1
T
A2
Ea
r
ly Enterocyte
Enterocyte
Secreto
r
y
progenitor
Gob
let cell
Early EEC
Enterochromaffin
other EECs
Percent expressed
0
25
50
75
100
Relative expression
−1
0
1
2
LGR5
NFIA
OLFM4
CCDC85B
MCM6
MKI67
F
ABP1
K
RT20
IL32
GDF15
HES1
RNF186
MUC2
REG4
PLA2G2A
DE
FA6
NEUROG3
P
AX4
CHGA
TPH1
ARX
GCG
Paneth cell
EEC
(CHGA
+
)
Goblet Cell
(MUC2
+
)
Paneth Cell
(DEFA5
+
)
0.0
0.2
0.4
0.6
0.8
1.0
5
10
15
Cell Percentage (%)
***
***
*
CHGA CHGA
+
-/-
RESEARCH | RESEARCH ARTICLE
Lin et al., Science 382, 451458 (2023) 27 October 2023 3of8
Downloaded from https://www.science.org on December 15, 2023
in ZNF800
/
organoids ( 2.3%) compared
with WT organoids (10.8%) (fig. S5C). Clonal
formation efficie ncy of WT and ZNF800
/
SI
and colon orga noids revealed no significant
differences (fig. S6, A and B). Cell-cycle pro-
gression in WT versus ZNF800
/
organoids
using a Fucci cell-cycle reporter (35) showed
no signif icant differenc es (fig. S6C). The com-
bined results indicate that ZNF800 does not
affect ISC homeostasis in organoids.
EECs were further subclustered based on
markers of early EECs (NEUROG3 and PAX4),
enterochromaffin cells (ECs, CHGA and TPH1)
and the other EEC subtypes (D cells, SST;Lcells,
GCG and PYY; M/X cells, MLN and GHRL;
I cells, CCK; G cells, GAST; N cells, NTS) (Fig.
2B and fig. S7A). Notably, early EECs and ECs
were increased in ZNF800
/
organoids, where-
as other EEC subtypes were depleted (Fig. 2,
C and D, and fig. S7A). The same phenotype
was also observed in human colon organoids
(fig. S8, A and B). Thus, ZNF800 loss induces
robust EEC differentiation at the expense of
goblet cells while also driving an EC-biased
trajectory.
We reexpressed WT ZNF800 protein in
ZNF800
/
organoids by doxycycline (dox)
inducible overexpression (Fig. 2E and fig. S8C),
which effectively rescued goblet and Paneth cell
differentiation while repressing EEC lineage
commitment (Fig. 2F). Notably, the EC-biased
differentiation pattern was also reversed upon
ZNF800 rescue, which resulted in decreased
serotonin secretion, a hallmark hormone of
ECs (Fig. 2G and fig. S8D).
ZNF800 represses the endocrine T F
regulatory network
Differential gene expression (DGE) analysis in
CHGA
+
cell populations of WT and ZNF800
/
organoids highlighted distinct expression pat-
terns of EEC-specific TFs (fig. S9, A and B). To
unravel these gene regulatory mechanisms,
we performed single-cell regulatory network
inference and clustering (SCENIC) analysis
on our scRNA-seq dataset (36), which iden-
tified 249 regulons activated across different
cell clusters (table S4). Regulons controlling
EEC commitment displayed higher activity
in ZNF800
/
organoids, including SOX4,
NEUROD2,andPAX4 (Fig. 3A). Notably, TEF
and NFIC, discovered in the TFome CRISPR
screen as EEC regulators, also showed regulon
activity in EEC lineages (fig. S9C), supporting
the functional relevance of our hits.
Chromatin immunoprecipitation sequencing
(ChIP-seq) with anti-ZNF800 and anti-FLAG
antibodies revealed 11,565 consensus peaks in
WT organoids and 7085 in rescued ZNF800
/
organoids (q value < 0.01) (fig. S10A). Most
ZNF800-binding sites localized within ±5 kb of
transcription start sites (TSS) (fig. S10B). Com-
parison between ZNF800 peak-proximal genes
in WT organoids and in rescued ZNF800
/
organoids showed a clear overlap (Fig. 3B).
Of the consensus 3625 ZNF800-binding genes,
we captured 3085 (85%) in our scRNA-seq
dataset (fig . S11A). DGE analysis (adjusted
P value < 0.05) revealed that 461 (15%) of the
ZNF800-bound genes were significantly up-
regulated upon ZNF800 knockout, whereas 59
(2%) were significantly down-regulated (fig.
S11B and table S5), indicating that ZNF800
functions as a transcriptional repres sor.
Gene ontology enrichment analysis of 870
genes (corresponding to the top 1000 anti-
ZNF800 ChIP-seq peak loci) revealed enrichment
of neural and endocrine-gland development
pathways (fig. S11C and table S6). We then
constructed an enrichm ent map for gene sets
involved in gland development, endocrine sys-
tem development, and pancreas develop ment
(Fig. 3C). In particular, INSM1, NEUROG3,and
PAX4 were found to be central TFs in each node
with the highest ZNF800 binding activity. As
predicted from SCENIC (Fig . 3A), ZNF800
bound SOX4 and NEUROD2 loci (Fig. 3D and
fig. S12A). EGR2 and DLL3 were also priori-
tized as top hits by ZNF800 peak abundance;
both are implicated in neurogenesis (3741)
and specifically expressed in endocrine cell
lineages (fig. S12B). These findings were highly
correlated with their respective gene expres-
sion profiles (fig. S10C and S12B). Notably, we
also identified mild enrichment of ZNF800
binding activity on the NEUROD1 locus (fig.
S12A). Knockout of ZNF800 resulted in increased
gene expression of NEUROD1 (fig. S11B). How-
ever, NEUROD1 did not appear as a hit in our
CRISPR screen (fig. S12C). Neurod1 acts as a late-
stage EEC TF in mice (12). In human EECs,
NEUROD1 was also found to mark late pro-
genitors and mature EECs (18). We therefore
generated NEUROD1
/
organoids, which ex-
hibited no effect on EEC or goblet cell differ-
entiation (fig. S12, D to F), whereas profiling of
EEC subtypes demonstrat e d a mil d decr e as e in
ECs, G cells, and M/X cells and a mild increase
in N cells (fig. S12G). These results confirmed
that NEUROD1 is a late-stage TF regulator,
which does not affect the EEC or goblet binary
cellfate decision.
Most ZNF800-binding loci presented a bi-
valent (H3K4me3- and H3K27me3-containing)
chromatin signature in the SI and colon (Fig.
3D and fig. S13A). Such marks are associated
with stem-cell different iation, allowing timely
responsiveness to TF regulation (42, 43). The
reversibility of gene activation or suppression
by poised ZNF800-binding loci was tested in
organoids with inducible ZNF800 expression
(fig. S13B). Sequential cycles of dox induction
demonstrated that ZNF800 alone can trigger
a dynamic yet reversible equilibrium between
EECs and goblet cells.
GFI1 and ZNF800 function independently in
repressing EEC differentiation
Given that both ZNF800 and GFI1 were discovered
i n our screen as repressors of EEC differentia-
tion, we sought to understand their regulatory
interactions by generating a double knock-
out of GFI1 and ZNF800 (GFI1
/
;ZNF800
/
)
inhumanSIorganoids(fig.S14A).Furtherab-
rogation of goblet and Paneth cells was ob-
served in GFI1
/
;ZNF800
/
organoids (fig. S14,
C and D), mirrored by further induction of
EECs. The GFI1
/
single knockout did not lead
to EC bias (fig. S14E). Instead, we observed a
mild decrease of EC cells (TPH1)andasubtle
increase in L cells (PYY). Notably, ZNF800
expression was not affected by GFI1 loss (fig.
S14, B and E). Because GFI1 is expressed in
goblet and Paneth cells (33)andtheknockout
of ZNF800 significantly depleted both cell
types, we assayed GFI1 expression in goblet cells
sorted from WT (12 ± 1.0%) and ZNF800
/
(3.4 ± 0.14%) organoids to avoid biased cell
heterogeneity of the organoids, which revealed
no differences in GFI1 expression (fig. S14F).
Overall, our results indicated that ZNF800 and
GFI1 function independently as repressors of
EEC differentiation.
Fig. 2. Phenotypic characterization of ZNF800
/
organoids. (A) Uniform
manifold approximation and projection (UMAP) of WT and ZNF800
/
organoids
from scRNA-seq. Descriptive cluster labels are shown. TA1, transit-amplifying cell
stage 1; TA2, transit-amplifying cell stage 2. (B) Dot plot showing the relative
expression and the percentage of cells expressing selected markers across
scRNA-seq clusters. Two representative markers for each cluster are plotted.
(C) Stacked bar plot showing the EEC cell populations in WT and ZNF800
/
organoids. (D) Reverse transcription (RT)qPCR quantification of various EEC
markers in CHGA
+
cell populations of WT and ZNF800
/
organoids. (E) (Top)
Schematic of dox-inducible overexpression construct of ZNF800 . (Bottom left)
Representative immunohistochemical staining images of ZNF800 in ZNF800
/
organoids with or without dox-induced ZNF800 expression. (Bottom right)
Representative confocal images of organoids with fluorescent reporters. Scale
bars, 100 mm. (F) Proportion of EECs and goblet and Paneth cells as determined
by fluorescence-activated cell sorting (FACS) analysis of the respective
reporters in ZNF800
/
organoids with or without dox-induced ZNF800
expression. (G) Enzyme-linked immunosorbent assay (ELISA) quantification of
serotonin secretion of organoids in different conditions. Data in this figure are
shown as mean ± SEM. N.d. not detected; ns, not significant. *P < 0.05;
**P < 0.01; ***P < 0.001; ****P < 0.0001 by multiple t tests with two-stage
linear step-up procedure of Benjamini, Krieger, and Yekutieli with Q =5%
and n =3.
RESEARCH | RESEARCH ARTICLE
Lin et al., Science 382, 451458 (2023) 27 October 2023 4of8
Downloaded from https://www.science.org on December 15, 2023
anti-ZNF800
anti-FLAG
Small Intestine
H3K4me3
Transverse colon
H3K4me3
Small intestine
H3K27me3
Transverse colon
H3K27me3
Gene
6kb
INSM1
[0 - 300]
[0 - 24]
[0 - 24]
[0 - 34]
[0 - 30]
[0 - 84]
SOX4
[0 - 49]
[0 - 15]
[0 - 270]
[0 - 270]
[0 - 10]
[0 - 10]
12kb
PAX4
[0 - 209]
[0 - 10]
[0 - 10]
[0 - 10]
[0 - 19]
[0 - 31]
6kb
NEUROG3
[0 - 170]
[0 - 10]
[0 - 10]
[0 - 30]
[0 - 31]
[0 - 90]
8kb
anti−ZNF800
a
nti−F
L
A
G
3980
(49.8%)
391
(4.9%)
3625
(45.3%)
Genes annotated from
ChIP-seq peaks
endocrine system development
endocrine system development
endocrine system development
endocrine system development
endocrine system development
endocrine system development
endocrine system development
endocrine system development
endocrine system development
endocrine system development
endocrine system development
endocrine system development
endocrine system development
endocrine system development
endocrine system development
endocrine system developmentendocrine system development
gland development
gland development
gland development
gland development
gland development
gland development
gland development
gland development
gland development
gland development
gland development
gland development
gland development
gland development
gland development
gland developmentgland development
pancreas development
pancreas development
pancreas development
pancreas development
pancreas development
pancreas development
pancreas development
pancreas development
pancreas development
pancreas development
pancreas development
pancreas development
pancreas development
pancreas development
pancreas development
pancreas developmentpancreas development
INSM1
INSM1
INSM1
INSM1
INSM1
INSM1
INSM1
INSM1
INSM1
INSM1
INSM1
INSM1
INSM1
INSM1
INSM1
INSM1INSM1
WNT11
WNT11
WNT11
WNT11
WNT11
WNT11
WNT11
WNT11
WNT11
WNT11
WNT11
WNT11
WNT11
WNT11
WNT11
WNT11WNT11
NEUROG3
NEUROG3
NEUROG3
NEUROG3
NEUROG3
NEUROG3
NEUROG3
NEUROG3
NEUROG3
NEUROG3
NEUROG3
NEUROG3
NEUROG3
NEUROG3
NEUROG3
NEUROG3NEUROG3
TBX1
TBX1
TBX1
TBX1
TBX1
TBX1
TBX1
TBX1
TBX1
TBX1
TBX1
TBX1
TBX1
TBX1
TBX1
TBX1TBX1
NOG
NOG
NOG
NOG
NOG
NOG
NOG
NOG
NOG
NOG
NOG
NOG
NOG
NOG
NOG
NOGNOG
PAX6
PAX6
PAX6
PAX6
PAX6
PAX6
PAX6
PAX6
PAX6
PAX6
PAX6
PAX6
PAX6
PAX6
PAX6
PAX6PAX6
ONECUT1
ONECUT1
ONECUT1
ONECUT1
ONECUT1
ONECUT1
ONECUT1
ONECUT1
ONECUT1
ONECUT1
ONECUT1
ONECUT1
ONECUT1
ONECUT1
ONECUT1
ONECUT1ONECUT1
BMP2
BMP2
BMP2
BMP2
BMP2
BMP2
BMP2
BMP2
BMP2
BMP2
BMP2
BMP2
BMP2
BMP2
BMP2
BMP2BMP2
GLI2
GLI2
GLI2
GLI2
GLI2
GLI2
GLI2
GLI2
GLI2
GLI2
GLI2
GLI2
GLI2
GLI2
GLI2
GLI2
GLI2
INHBB
INHBB
INHBB
INHBB
INHBB
INHBB
INHBB
INHBB
INHBB
INHBB
INHBB
INHBB
INHBB
INHBB
INHBB
INHBBINHBB
PDGFRA
PDGFRA
PDGFRA
PDGFRA
PDGFRA
PDGFRA
PDGFRA
PDGFRA
PDGFRA
PDGFRA
PDGFRA
PDGFRA
PDGFRA
PDGFRA
PDGFRA
PDGFRAPDGFRA
WNT5A
WNT5A
WNT5A
WNT5A
WNT5A
WNT5A
WNT5A
WNT5A
WNT5A
WNT5A
WNT5A
WNT5A
WNT5A
WNT5A
WNT5A
WNT5AWNT5A
BMP6
BMP6
BMP6
BMP6
BMP6
BMP6
BMP6
BMP6
BMP6
BMP6
BMP6
BMP6
BMP6
BMP6
BMP6
BMP6BMP6
CDH2
CDH2
CDH2
CDH2
CDH2
CDH2
CDH2
CDH2
CDH2
CDH2
CDH2
CDH2
CDH2
CDH2
CDH2
CDH2CDH2
ASCL1
ASCL1
ASCL1
ASCL1
ASCL1
ASCL1
ASCL1
ASCL1
ASCL1
ASCL1
ASCL1
ASCL1
ASCL1
ASCL1
ASCL1
ASCL1ASCL1
DLL1
DLL1
DLL1
DLL1
DLL1
DLL1
DLL1
DLL1
DLL1
DLL1
DLL1
DLL1
DLL1
DLL1
DLL1
DLL1DLL1
SHH
SHH
SHH
SHH
SHH
SHH
SHH
SHH
SHH
SHH
SHH
SHH
SHH
SHH
SHH
SHHSHH
ONECUT2
ONECUT2
ONECUT2
ONECUT2
ONECUT2
ONECUT2
ONECUT2
ONECUT2
ONECUT2
ONECUT2
ONECUT2
ONECUT2
ONECUT2
ONECUT2
ONECUT2
ONECUT2ONECUT2
NKX6−2
NKX6−2
NKX6−2
NKX6−2
NKX6−2
NKX6−2
NKX6−2
NKX6−2
NKX6−2
NKX6−2
NKX6−2
NKX6−2
NKX6−2
NKX6−2
NKX6−2
NKX6−2NKX6−2
ALDH1A3
ALDH1A3
ALDH1A3
ALDH1A3
ALDH1A3
ALDH1A3
ALDH1A3
ALDH1A3
ALDH1A3
ALDH1A3
ALDH1A3
ALDH1A3
ALDH1A3
ALDH1A3
ALDH1A3
ALDH1A3ALDH1A3
NTN1
NTN1
NTN1
NTN1
NTN1
NTN1
NTN1
NTN1
NTN1
NTN1
NTN1
NTN1
NTN1
NTN1
NTN1
NTN1NTN1
WNT7B
WNT7B
WNT7B
WNT7B
WNT7B
WNT7B
WNT7B
WNT7B
WNT7B
WNT7B
WNT7B
WNT7B
WNT7B
WNT7B
WNT7B
WNT7BWNT7B
LEF1
LEF1
LEF1
LEF1
LEF1
LEF1
LEF1
LEF1
LEF1
LEF1
LEF1
LEF1
LEF1
LEF1
LEF1
LEF1LEF1
SIX4
SIX4
SIX4
SIX4
SIX4
SIX4
SIX4
SIX4
SIX4
SIX4
SIX4
SIX4
SIX4
SIX4
SIX4
SIX4SIX4
NKX2−3
NKX2−3
NKX2−3
NKX2−3
NKX2−3
NKX2−3
NKX2−3
NKX2−3
NKX2−3
NKX2−3
NKX2−3
NKX2−3
NKX2−3
NKX2−3
NKX2−3
NKX2−3NKX2−3
MAFB
MAFB
MAFB
MAFB
MAFB
MAFB
MAFB
MAFB
MAFB
MAFB
MAFB
MAFB
MAFB
MAFB
MAFB
MAFBMAFB
OXTR
OXTR
OXTR
OXTR
OXTR
OXTR
OXTR
OXTR
OXTR
OXTR
OXTR
OXTR
OXTR
OXTR
OXTR
OXTROXTR
NKX2−8
NKX2−8
NKX2−8
NKX2−8
NKX2−8
NKX2−8
NKX2−8
NKX2−8
NKX2−8
NKX2−8
NKX2−8
NKX2−8
NKX2−8
NKX2−8
NKX2−8
NKX2−8NKX2−8
NRG1
NRG1
NRG1
NRG1
NRG1
NRG1
NRG1
NRG1
NRG1
NRG1
NRG1
NRG1
NRG1
NRG1
NRG1
NRG1NRG1
BMP7
BMP7
BMP7
BMP7
BMP7
BMP7
BMP7
BMP7
BMP7
BMP7
BMP7
BMP7
BMP7
BMP7
BMP7
BMP7BMP7
PKD2
PKD2
PKD2
PKD2
PKD2
PKD2
PKD2
PKD2
PKD2
PKD2
PKD2
PKD2
PKD2
PKD2
PKD2
PKD2PKD2
PLXND1
PLXND1
PLXND1
PLXND1
PLXND1
PLXND1
PLXND1
PLXND1
PLXND1
PLXND1
PLXND1
PLXND1
PLXND1
PLXND1
PLXND1
PLXND1PLXND1
SERPINE2
SERPINE2
SERPINE2
SERPINE2
SERPINE2
SERPINE2
SERPINE2
SERPINE2
SERPINE2
SERPINE2
SERPINE2
SERPINE2
SERPINE2
SERPINE2
SERPINE2
SERPINE2SERPINE2
FOXF1
FOXF1
FOXF1
FOXF1
FOXF1
FOXF1
FOXF1
FOXF1
FOXF1
FOXF1
FOXF1
FOXF1
FOXF1
FOXF1
FOXF1
FOXF1FOXF1
BCL2
BCL2
BCL2
BCL2
BCL2
BCL2
BCL2
BCL2
BCL2
BCL2
BCL2
BCL2
BCL2
BCL2
BCL2
BCL2BCL2
WNT3A
WNT3A
WNT3A
WNT3A
WNT3A
WNT3A
WNT3A
WNT3A
WNT3A
WNT3A
WNT3A
WNT3A
WNT3A
WNT3A
WNT3A
WNT3AWNT3A
ID4
ID4
ID4
ID4
ID4
ID4
ID4
ID4
ID4
ID4
ID4
ID4
ID4
ID4
ID4
ID4ID4
CSF1
CSF1
CSF1
CSF1
CSF1
CSF1
CSF1
CSF1
CSF1
CSF1
CSF1
CSF1
CSF1
CSF1
CSF1
CSF1CSF1
GLI3
GLI3
GLI3
GLI3
GLI3
GLI3
GLI3
GLI3
GLI3
GLI3
GLI3
GLI3
GLI3
GLI3
GLI3
GLI3GLI3
MSX2
MSX2
MSX2
MSX2
MSX2
MSX2
MSX2
MSX2
MSX2
MSX2
MSX2
MSX2
MSX2
MSX2
MSX2
MSX2MSX2
LRP5L
LRP5L
LRP5L
LRP5L
LRP5L
LRP5L
LRP5L
LRP5L
LRP5L
LRP5L
LRP5L
LRP5L
LRP5L
LRP5L
LRP5L
LRP5LLRP5L
NEURL1
NEURL1
NEURL1
NEURL1
NEURL1
NEURL1
NEURL1
NEURL1
NEURL1
NEURL1
NEURL1
NEURL1
NEURL1
NEURL1
NEURL1
NEURL1NEURL1
WNT1
WNT1
WNT1
WNT1
WNT1
WNT1
WNT1
WNT1
WNT1
WNT1
WNT1
WNT1
WNT1
WNT1
WNT1
WNT1WNT1
PA M
PA M
PA M
PA M
PA M
PA M
PA M
PA M
PA M
PA M
PA M
PA M
PA M
PA M
PA M
PA MPA M
NRP1
NRP1
NRP1
NRP1
NRP1
NRP1
NRP1
NRP1
NRP1
NRP1
NRP1
NRP1
NRP1
NRP1
NRP1
NRP1NRP1
IGF2
IGF2
IGF2
IGF2
IGF2
IGF2
IGF2
IGF2
IGF2
IGF2
IGF2
IGF2
IGF2
IGF2
IGF2
IGF2IGF2
FOXB1
FOXB1
FOXB1
FOXB1
FOXB1
FOXB1
FOXB1
FOXB1
FOXB1
FOXB1
FOXB1
FOXB1
FOXB1
FOXB1
FOXB1
FOXB1FOXB1
TFCP2L1
TFCP2L1
TFCP2L1
TFCP2L1
TFCP2L1
TFCP2L1
TFCP2L1
TFCP2L1
TFCP2L1
TFCP2L1
TFCP2L1
TFCP2L1
TFCP2L1
TFCP2L1
TFCP2L1
TFCP2L1TFCP2L1
PTF1A
PTF1A
PTF1A
PTF1A
PTF1A
PTF1A
PTF1A
PTF1A
PTF1A
PTF1A
PTF1A
PTF1A
PTF1A
PTF1A
PTF1A
PTF1APTF1A
PA X 4
Gene set size
10
20
30
40
50
100
150
Peak enrichment
C
D
Condition
ZNF800
-/-
WT
TF activity
(z-score of AUC)
WTZNF800
-/-
-4
-2
240
A
B
ISC
TA1
TA2
Early Enterocyte
Enterocyte
Secretory progenitor
Goblet cell
Paneth cell
Early EEC
Other EECs
Enterochromaffin
SAP30
HMGB3
TCF3
HDAC2
NFYB
MYCN
HES6
KLF13
USF1
ETS1
PAX4
DMRTA2
HOXA9
NEUROD2
TEAD2
SOX4
FOXL2
SNAI1
FOXA2
UNCX
FOXA3
*
*
*
*
*
*
*
*
Early EEC
Fig. 3. ZNF800 binds to chromatin regions of TFs involved in endocrine
differentiation. (A) Heatmap with unsupervised clustering of gene regulatory
network activity in different cell clusters within WT and ZNF800
/
organoids
and visualized as row z-scores of mean area-under-the-curve (AUC) values.
Zoom-in plot highlighting the top TF regulons specifically activated in early EECs.
Regulons with significant differences between WT and ZNF800
/
are indicated
with asterisks, assessed by Wilcoxon rank sum test. (B) Venn diagram of
overlapping genes annotated from ChIP-seq datasets generated by anti-ZNF800
antibody in WT organoids and anti-FLAG antibody in ZNF800
/
organoids with
dox-induced ZNF800 expression. (C) An enrichment-network plot depicting
the linkages of gene sets and three selected pathways by gene ontology analysis.
(D) ChIP-seq tracks at the INSM1, PAX4, NEUROG3, and SOX4 loci.
RESEARCH | RESEARCH ARTICLE
Lin et al., Science 382, 451458 (2023) 27 October 2023 5of8
Downloaded from https://www.science.org on December 15, 2023
PAX4 drives EC differentiation directly
downstream of ZNF800
A panel of genes was found to be up-regulated
across multiple cell types upon ZNF800 knockout
( table S7), including COL4A2, TMEM178B,and
BTB D 1 1 (fig. S15A). ZNF800 binds to these genes,
but not to the down-regulated genes such as
SULT1C2, HMGCS1,andMS4A8 (fig.S15B).De-
spite their modest up-regulation, the expression
pattern and the annotated function of these
genes did not directly relate to the EEC phe-
notype. Therefore, we focused on EEC TFs
with stronger functional implications (fig. S9,
AandB).
We focused on three direct ZNF800 target
genes, INSM1, SOX4, and PAX4 (Fig. 3D), by
performing double knockouts (fig. S16A). EEC
induction was effectively suppressed, but to
different extents (Fig. 4A). Similar to the single
depletion of INSM1 (fig. S3C), INSM1 knockout
in ZNF800
/
organoids caused an essentially
complete reversal of EEC induction, confirming
its pivotal role in the TF cascade (8, 44). Addi-
tional knockout of SOX4 or PAX4 in ZNF800
/
organoids both reduced EEC numbers.
Next, we analyzed EEC subtypes in the CHGA
+
cell population from different mutant back-
grounds (Fig. 4B and fig. S17, A and B). Ad-
ditional knockout of INSM1 or SOX4 caused
a mild reduction of EC cells (TPH1)inCHGA
+
cells while further suppressin g L cells (GCG and
PYY) and D cells (SST). Loss of PAX4 directed
a robust conversion of EC-biased EECs into all
other EEC subtypes. Basal secretion of subtype
hormones (L cell, GLP-1; M/X cell, Ghrelin),
confirmed their functionality (Fig. 4C). We also
performed a single knockout of SOX4 and
PAX4 (fig. S3A, S16B and S18A). The SOX4
/
single knockout significantly repressed L cells
(GC G), N cells (NTS), D cells (SST), and G
cells (GAST), whereas MLN-expressing M/X cells
werenotaffected,agreeingwithfindingsin
Sox4
/
mice (12)(fig.S18,BtoD).ThePAX4
/
single knockout repressed ECs while mildly up-
regulating other EEC subtypes. Overall, PAX4,
as a direct target of ZNF800, appeared respon-
sible for the EC-biased EEC differentiation
trajectory seen upon loss of ZNF800.
Mouse ECs are exclusively Pax4-dependent
(45 ), whereas Arx is essential for L, G, I, and N
cells (10, 45). We therefore probed a possible
interaction between PAX4 and ARX in our
organoids. ARX expression was down-regulated
in ZNF800
/
organoids and up-regulated in
EC cells - CHGA
EC cells - TPH1
L cells - GCG
L cells - PYY
N cells- NTS
D cells - SST
I cells - CCK
G cells - GAST
M/X cells - MLN
M/X cells - GHRL
WT
ZNF800
-/-
ZNF800
-/-
; SOX4
-/-
ZNF800
-/-
; INSM1
-/-
ZNF800
-/-
; PAX4
-/-
AB
CE
ZNF800
-/-
ZNF800
-/-
;INSM1
-/-
ZNF800
-/-
;SOX4
-/-
ZNF800
-/-
;PAX4
-/-
0
5
10
15
CHGA
+
Cell Percentage (% )
****
****
****
WT
0.0
0.5
1.0
1.5
L cell: GLP-1
Secretion (ng/mL)
n.d.
WT
ZNF800
-/-
0.001
0.01
0.1
1
10
M/X cell: Ghrelin
n.d.
ZNF800
-/-
;PAX4
-/-
ZNF800
-/-
;PAX4
-/-
ZNF800
-/-
D
Stem cell
ZNF800
Early EEC
Goblet cell
Enterochromaffin
INSM1
SOX4
PAX4
NEUROG3
PAX4
LN D
I
G
M/X
other EEC subtypes
sorted CHGA
+
cells
0.001
0.01
0.1
1
10
100
Relative fold change vs WT
(normalized by GAPDH)
n.d.
n.d.
n.d.
n.d.
n.d.
n.d.
n.d.
n.d.
n.d.
****
*
*
** ***
***
**
ns
***
ns
*
ns
*
ns
*
ns
****
ns
ns
ns
***
****
*
***
*
**
**
**
**
n.d.
ARX gene expression
in CHGA
+
sorted cells
0
2
4
6
8
Relative fold change vs WT
(normalized by GAPDH)
*
***
*
WT
ZNF800
-/-
;PAX4
-/-
ZNF800
-/-
PAX4
-/-
ARX
Fig. 4. PAX4 is responsible for EC-biased differentiation trajectory as a
direct downstream target of ZNF800. (A) Proportions of EECs as determined
by FACS analysis of organoids from different genotypes by CHGA reporter.
(B) RT-qPCR quantification of various EEC markers in CHGA
+
cell populations of
organoids from different genotypes. (C) ELISA quantification of GLP-1 and Ghrelin
secretion of organoids from different genotypes. (D) RT-qPCR quantification of
ARX expression in CHGA
+
cell populations of organoids from different conditions.
(E) Schematic of ZNF800-driven repression model during endocrine cell
differentiation. Data in this figure are shown as mean ± SEM. N.d. not detected;
ns, not significant. *P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001 by
multiple t tests with two-stage linear step-up procedure of Benjamini, Krieger,
and Yekutieli with Q = 5%, n =3.
RESEARCH | RESEARCH ARTICLE
Lin et al., Science 382, 451458 (2023) 27 October 2023 6of8
Downloaded from https://www.science.org on December 15, 2023
PAX4
/
single-knockout and ZNF800
/
;PAX4
/
double-knockout organoids (Fig . 4D and fig.
S19, A and B). Reintroduction of PAX4 in
ZNF800
/
;PAX4
/
organoids significantly
repressed ARX expression (fig. S20, A and B).
Furthermore, as expected, the EEC differentia-
tion tr a je c tor y i n Z N F800
/
;PAX4
/
organoids
was redirected to an EC-biased phenotype upon
PA X4 reexpression.
Reciprocal transcriptional repression exists
between Pax4 and Arx during mouse pancrea-
tic development (46, 47). We assessed whether
ARX also inhibits intestinal PAX4 expression
by overexpressing ARX in ZNF800
/
organoids
(fig. S21A). ARX overexpression was insuffi-
cient to reverse the EC-biased EEC cell type
specification induced by the ZNF800-PAX4
axis (fig. S21, B and C). Furthermore, PAX4
expression was not affected upon ARX over-
expression in the ZNF800
/
condition (fig. S21D).
W e next performed ChIP-qPCR of ARX on the
PAX4 locus using FLAG-tagged ARX in human
SI organoids (fig. S21E). Unlike ZNF800, ARX
did not bind to the PAX4 upstream enhancer
region(fig.S21F).WealsoperformedChIP-qPCR
of PAX4 on the ARX locus using FLAG-tagged
PAX4 (fig. S22A), focusing on six ultraconserved
enhancers (4850) (fig . S22B). PAX4 did not
bind across these enhancers, including hs121,
which exh ibits PAX4 binding activity in the
mouse pancreas (fig. S22C) (49). Overall, our
results strongly supported that PAX4 unilater-
ally inhibits ARX, whereas the inhibitory effect
does not involve direct chromatin interaction
through the previously described regulatory
elements.
Discussion
The human SI epithelium consists of at least
14 main cell types, including six EEC lineages
(51, 52). Leveraging the near-physio logical cell
heterogeneity of human SI organoids, we per-
formed a CRISPR screen for positive and neg-
ative TF regulators of EEC lineage commitment.
Our findings defin e a ZNF800 -repressive
mechanism upstream of the classic endocrine
gene regulatory network (Fig. 4E). Among its
direct downstream targets, INSM1, SOX4,and
NEUROG3 are well described central players
that drive early EEC commitment. We also
found that PAX4, in the absence of ZNF800,
drives an EC-specific cell fate by suppressing
differentiation of all other EEC subtypes.
The PAX4 knockout rescued somatostatin-
producing D cells in ZNF800
/
organoids
(Fig. 2D and fig. S7A). Although Pax4 con-
trols both b- and somatostatin-producing d-cell
specification in the mouse pancreas (46), our
single PAX4 knockout had no effect on SST-
expressing D cells (fig. S18C). This prompts
questions into the generalizability of endo-
crine fate regulation between different diges-
tive organs. Double depletion of Pax4 and Arx
promoted d-cell specification in mice (49),
suggesting that a third TF is involved in d-cell
cell differentiation, and that Pax4 is likely to in-
duce a b-cell fate at the expense of d cells. There-
fore, the up-regulation of PAX4 by ZNF800
/
in human SI organoids might drive a further
binary cellfate decision between ECs and
D cells.
ZNF800 is ubiquitously expressed along the
crypt-villus axis in the adult human gut epi-
thelium (fig. S4A), marked by H3K4me3 en-
richment and low H3K27me3 levels (fig. S4B),
indicating an open chromatin state and active
expression. The scRNA-seq dataset of the devel-
oping human gut revealed that ZNF800 is
already expressed at the earliest assayed time
point of 6.1 postconception weeks (53). Given
the downstream TF network described in our
study, it appears likely that ZNF800 also plays
an important function in embryonic develop-
ment. A recent coexpression network study in
mouse pancreatic development (54) revealed
that expres sion of the mouse ortholog zfp800
correlates with endocrine specification from
embryonic day 8 (E8) until E15.5. Global knock-
out of zfp800 caused postnatal lethality and
disrupted early pancreatic development (in-
cluding both endocrine and acinar cells at
E18.5). Thus, the constitutive null phenotype
hindered a mechanistic study of mouse zfp800
function in the endocrine lineages. It is con-
ceivable that ZNF800 might regulate b-cell
differentiation in the human pancreas through
the TF network described in this study.
REFERENCES AND NOTES
1. N. Barker et al., Nature 449, 10031007 (2007).
2. Y. Sei, X. Lu, A. Liou, X. Zhao, S. A. Wank, Am. J. Physiol.
Gastrointest. Liver Physiol. 300, G345G356 (2011).
3. S. J. A. Buczacki et al., Nature 495,6569 (2013).
4. O. Basak et al., EMBO J. 33, 20572068 (2014).
5. O. Basak et al., Cell Stem Cell 20, 177190.e4 (2017).
6. H. Mutoh et al., Proc. Natl. Acad. Sci. U.S.A. 94, 35603564
(1997).
7. F. J. Naya et al., Genes Dev. 11, 23232334 (1997).
8. M. S. Gierl, N. Karoulias, H. Wende, M. Strehle, C. Birchmeier,
Genes Dev. 20, 24652478 (2006).
9. S. Desai et al., Dev. Biol. 313,5866 (2008).
10. A. Du et al., Dev. Biol. 365, 175188 (2012).
11. S. Gross et al., Development dev.130682 (2016).
12. H. Gehart et al., Cell 176, 11581173.e16 (2019).
13. T. Sato et al., Nature 459, 262265 (2009).
14. T. Sato et al
., Gastroenterology 141, 17621772 (2011).
15. G. Gradwohl, A. Dierich, M. LeMeur, F. Guillemot, Proc. Natl.
Acad. Sci. U.S.A. 97, 16071611 (2000).
16. M. Jenny et al., EMBO J. 21, 63386347 (2002).
17. J. Lee et al., eLife 2, e00940 (2013).
18. J. Beumer et al., Cell 181, 12911306.e19 (2020).
19. G.-W. He et al., Cell Stem Cell 29, 13331345.e6
(2022).
20. J. G. Doench et al., Nat. Biotechnol. 34, 184191 (2016).
21. S. A. Lambert et al., Cell 172 , 650665 (2018).
22. L. Lin et al., Cell Rep. 33, 108426 (2020).
23. N. F. Shroyer, D. Wallis, K. J. T. Venken, H. J. Bellen,
H. Y. Zoghbi, Genes Dev. 19, 24122417 (2005).
24. T. K. Noah, B. Donahue, N. F. Shroyer, Exp. Cell Res. 317,
27022710 (2011).
25. T. Nakaya et al., Cancer Res. 74, 28822891 (2014).
26. V. Korinek et al., Nat. Genet. 19, 379383 (1998).
27. E. Batlle et al., Cell 111
, 251263 (2002).
28. M. van de Wetering et al., Cell 111, 241250 (2002).
29. J. H. van Es et al., Mol. Cell. Biol. 32, 19181927
(2012).
30. K. Minamide et al., Sci. Rep. 10, 14639 (2020).
31. T. Sato et al., Nat. Cell Biol. 22, 919926 (2020).
32. A. D. Gracz et al., Gastroenterology 155, 15081523.e10
(2018).
33. M. Bjerknes, H. Cheng, Dev. Biol. 345,4963 (2010).
34. H. S. Najafabadi et al., Nat. Biotechnol. 33, 555562
(2015).
35. A. Sakaue-Sawano et al., Cell 132, 487498 (2008).
36. S. Aibar et al., Nat. Methods 14, 10831086 (2017).
37. M. H. Sham et al., Cell 72, 183196 (1993).
38. J. Ghislain, C. Desmarquet-Trin-Dinh, P. Gilardi-Hebenstreit,
P. Charnay, M. Frain, Development 130, 941953
(2003).
39. A. Desmazières, P. Charnay, P. Gilardi-Hebenstreit, J. Biol. Chem.
284,1083110840 (2009).
40. S. L. Dunwoodie, D. Henrique, S. M. Harrison,
R. S. P. Beddington, Development 124, 3065
3076
(1997).
41. R. M. Henke, D. M. Meredith, M. D. Borromeo, T. K. Savage,
J. E. Johnson, Dev. Biol. 328, 529540 (2009).
42. B. E. Bernstein et al., Cell 125, 315326 (2006).
43. P. Voigt, W.-W. Tee, D. Reinberg, Genes Dev. 27, 13181338
(2013).
44. A. B. Osipovich et al., Development 141, 29392949
(2014).
45. A. Beucher et al., PLOS ONE 7, e36449 (2012).
46. P. Collombat et al., Genes Dev. 17, 25912603 (2003).
47. P. Collombat et al., Cell 138, 449462 (2009).
48. G. Bejerano et al., Science 304, 13211325 (2004).
49. P. Collombat et al., Development 132, 29692980
(2005).
50. D. E. Dickel et al., Cell 172, 491499.e15 (2018).
51. R. Elmentaite et al., Nature 597, 250255 (2021).
52. J. Burclaff et al., Cell. Mol. Gastroenterol. Hepatol. 13,
15541589 (2022).
53. R. Elmentaite et al.,
Dev. Cell 55, 771783.e5
(2020).
54. A. B. Osipovich et al., Development 148, dev196964
(2021).
55. ENCODE Project Consortium, Nature 489,5774
(2012).
56. Y. Luo et al., Nucleic Acids Res. 48 (D1), D882D889
(2020).
AC KN OW LE D GM E NT S
We thank R. Sherwood (Harvard Medical School, Brigham and
Women's Hospital, Division of Genetics) for providing the TFome
sgRNA library in the CRISPR-v2-FE backbone. We thank D. Krueger,
A. de Graaff, and the Hubrecht Imaging Centre for microscopy
assistance and the Hubrecht Flow Cytometry Core Facility for flow
cytometry analysis and cell sorting. We thank the Máxima Single
Cell Genomics Facility and Utrecht Sequencing Facility for
sequencing support. We thank the Microscopy CORE Lab of
the Faculty of Health, Medicine, and Life Sciences of Maastricht
University for its help in transmission electron microscopy
imaging. We thank the ENCODE Consortium and the
ENCODE production laboratories (B. Bernstein, Broad;
J. Stamatoyannopoulos, UW; B. Ren, UCSD) for generating the
related ChIP-seq public datasets. Funding: This work was
supported by the Netherlands Organ-on-Chip Initiative, a Dutch
Research Council (NWO) Gravitation project (024.003.001) funded
by the Ministry of Education, Culture, and Science of the
government of the Netherlands (H.C.); the Oncode Institute (partly
financed by the Dutch Cancer Society) (H.C. and J.H.v.E.); and the
European Research Council under ERC advanced grant no.
101020405 (GutHormones) (H.C.). The project Organoids in time
with project no. 2019.085 of the research program NWO
Investment Large is financed by the NWO (H.C. S.v.d.B.). Author
contributions: Conceptualization: L.L. and H.C.; Methodology: L.L.,
J.De., D.W., G.J.F.v.S., R.v.d.L., H.B., J.K., S.v.d.B., A.A.-R., C.L.-I.,
W.J.v .d.W., A.B., T.M., M.v.d.W., P.J.P., J.Dr ., J.H.v.E., and H.C.; Investigation:
L.L., J.De., G.J.F.v.S., H.B., J.K., A.A.-R., C.L.-I., and W.J.v.d.W.;
Visualization: L.L., J.De., G.J.F.v.S., and C.L.-I.; Funding acquisition:
J.H.v. E. and H.C.; Project administration: J.H.v. E. and H.C.; Supervision:
J.H.v.E. and H.C.; Writing original draft: L.L. and H.C.; Writing
review and editing: all authors. Competing interests: H.C. is an
inventor on patents held by the Royal Netherlands Academy of Arts
and Sciences that cover organoid technology. He is currently
head of pharma Research and Early Development (pRED) at
RESEARCH | RESEARCH ARTICLE
Lin et al., Science 382, 451458 (2023) 27 October 2023 7of8
Downloaded from https://www.science.org on December 15, 2023
Roche, Basel, Switzerland. H.C.s full disclosure is given at https://
www.uu.nl/staff/JCClevers/. Data and materials availability:
Further information and requests for resources and reagents
should be directed to the corresponding author. Specific and stable
reagents generated in this study are available and can be
requested from the corresponding author; a completed Materials
Transfer Agreement may be required. Sharing organoid lines used
in this study requires approval by our local institutional review
board. CRISPR screen sequencing data, scRNA-seq data, and
ChIP-seq data from this study have been deposited to the Gene
Expression Omnibus (GEO) under accession no. GSE229586.
ChIP-seq data for histone marks H3K4me3 and H3K27me3 from
human small intestine and colon were obtained from the ENCODE
portal. We downloaded the ChIP-seq datasets from the ENCODE
portal (55, 56) (https://www.encodeproject.org/) with the
following identifiers: ENCFF661MCT, ENCFF988AAQ, ENCFF219ZIO,
ENCFF284XHG, ENCFF603QDZ, ENCFF825TAZ, ENCFF745IGA, and
EENCFF830NSP. License information: Copyright © 2023 the
authors, some rights reserved; exclusive licensee American
Association for the Advancement of Science. No claim to original
US government works. https://www.science.org/about/science-
licenses-journal-article-reuse
SUPPLEMENTARY MATERIALS
science.org/doi/10.1126/science.adi2246
Materials and Methods
Figs. S1 to S22
References (5776)
Tables S1 to S8
MDAR Reproducibility Checklist
Submitted 17 April 2023; resubmitted 9 August 2023
Accepted 8 September 2023
10.1126/science.adi2246
RESEARCH | RESEARCH ARTICLE
Lin et al., Science 382, 451458 (2023) 27 October 2023 8of8
Downloaded from https://www.science.org on December 15, 2023
Use of this article is subject to the Terms of service
Science (ISSN 1095-9203) is published by the American Association for the Advancement of Science. 1200 New York Avenue NW,
Washington, DC 20005. The title Science is a registered trademark of AAAS.
Copyright © 2023 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim
to original U.S. Government Works
Unbiased transcription factor CRISPR screen identifies ZNF800 as master
repressor of enteroendocrine differentiation
Lin Lin, Jeff DeMartino, Daisong Wang, Gijs J. F. van Son, Reinier van der Linden, Harry Begthel, Jeroen Korving, Amanda
Andersson-Rolf, Stieneke van den Brink, Carmen Lopez-Iglesias, Willine J. van de Wetering, Aleksandra Balwierz,
Thanasis Margaritis, Marc van de Wetering, Peter J. Peters, Jarno Drost, Johan H. van Es, and Hans Clevers
Science 382 (6669), . DOI: 10.1126/science.adi2246
Editor’s summary
Enteroendocrine cells (EECs) reside in the epithelium of the digestive tract and produce various hormones involved
in metabolism. The generation of different EEC lineages is governed by a dedicated network of transcription factors.
However, given the low efficiency of EEC specification from adult stem cells, it has been difficult to elucidate the
components of this regulatory network. Lin et al. used an optimized human small intestinal organoid culture system
to perform an unbiased, systematic screen for transcription factors that regulate EEC differentiation. The screen
implicated ZNF800 as a key factor exerting a dominant repressive role in controlling the endocrine transcription factor
network. This work showcases the use of optimized human organoids for CRISPR-based functional screens, paving
the way for the identification of additional regulators in human gut physiology and pathophysiology. —Stella M. Hurtley
View the article online
https://www.science.org/doi/10.1126/science.adi2246
Permissions
https://www.science.org/help/reprints-and-permissions
Downloaded from https://www.science.org on December 15, 2023