Download as pdf or txt
Download as pdf or txt
You are on page 1of 37

Article

Inadequate DNA Damage Repair Promotes Mammary


Transdifferentiation, Leading to BRCA1 Breast
Cancer
Graphical Abstract Authors
Hua Wang, Dongxi Xiang, Ben Liu, ...,
Hans Clevers, Zhe Li, David M. Livingston

Correspondence
zli4@rics.bwh.harvard.edu (Z.L.),
david_livingston@dfci.harvard.
edu (D.M.L.)

In Brief
The tumor suppressor BRCA1 and certain
of its partners prevent the aberrant
transdifferentiation of mammary
epithelial cells to a mesenchymal state by
participating in interstrand crosslink DNA
repair.

Highlights
d Two BRCA1 partners, NUMB and HES1, participate in
crosslink DNA repair

d NUMB or HES1 loss or cisplatin triggers a luminal to basal/


mesenchymal transition

d BRCA1 loss leads to growing DNA damage in the mammary


epithelium and tumorigenesis

d BRCA1 tumorigenesis encompasses dynamic luminal/basal/


mesenchymal fate transitions

Wang et al., 2019, Cell 178, 135–151


June 27, 2019 ª 2019 Elsevier Inc.
https://doi.org/10.1016/j.cell.2019.06.002
Article

Inadequate DNA Damage Repair Promotes


Mammary Transdifferentiation, Leading
to BRCA1 Breast Cancer
Hua Wang,1,2,8 Dongxi Xiang,3,8 Ben Liu,1,2 Aina He,3 Helena J. Randle,1,2 Kelvin Xi Zhang,4 Anushka Dongre,5
Norman Sachs,6 Allison P. Clark,1,2 Luwei Tao,3 Qing Chen,3 Vladimir V. Botchkarev, Jr.,1,2 Ying Xie,3 Ning Dai,7
Hans Clevers,6 Zhe Li,3,* and David M. Livingston1,2,9,*
1Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
2Departments of Genetics and Medicine, Harvard Medical School, Boston, MA 02115, USA
3Division of Genetics, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
4Informatics IT, Merck & Co., Boston, MA 02115, USA
5Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
6Hubrecht Institute, Uppsalalaan 8, 3584 CT Utrecht, the Netherlands
7Department of Medicine, Robert Wood Johnson Medical School, Rutgers, New Brunswick, NJ 08901, USA
8These authors contributed equally
9Lead Contact

*Correspondence: zli4@rics.bwh.harvard.edu (Z.L.), david_livingston@dfci.harvard.edu (D.M.L.)


https://doi.org/10.1016/j.cell.2019.06.002

SUMMARY Mammary gland (MG) development is a unidirectional, hierar-


chical process wherein mammary stem cells (MaSCs) differen-
Loss of BRCA1 p220 function often results in basal- tiate into progenitors that further differentiate into mature
like breast cancer (BLBC), but the underlying disease MECs (Visvader and Stingl, 2014). Dedifferentiation of mature
mechanism is largely opaque. In mammary epithelial cells into a more primitive state rarely occurs in normal tissues.
cells (MECs), BRCA1 interacts with multiple proteins, However, when it does, it can trigger a neoplastic process (Fried-
including NUMB and HES1, to form complexes that mann-Morvinski and Verma, 2014; Van Keymeulen et al., 2015;
Visvader, 2011). Although BRCA1 can prevent transdifferentia-
participate in interstrand crosslink (ICL) DNA repair
tion of cultured, human MECs, whether this translates into BC
and MEC differentiation control. Unrepaired ICL
suppression is unclear (Wang et al., 2016).
damage results in aberrant transdifferentiation to BRCA1 p220 is necessary for maintaining a normal MEC dif-
a mesenchymal state of cultured, human basal- ferentiation state, a function that likely contributes to its BC
like MECs and to a basal/mesenchymal state in pri- suppression activity (Liu et al., 2008; Wang et al., 2016; Xu
mary mouse luminal MECs. Loss of BRCA1, et al., 1999). In humans, p220 depletion in basal-like MECs led
NUMB, or HES1 or chemically induced ICL damage to disorderly differentiation (Wang et al., 2016), and aberrant
in primary murine luminal MECs results in persistent luminal progenitors (LPs) were detected in germline BRCA1-
DNA damage that triggers luminal to basal/mesen- mutated mammary tissue (Lim et al., 2009; Nakshatri et al.,
chymal transdifferentiation. In vivo single-cell anal- 2015; Park et al., 2010; Proia et al., 2011).
ysis revealed a time-dependent evolution from Defective p220 function promotes aberrant differentiation of
otherwise normal, cultured, basal-like human MECs to a more
normal luminal MECs to luminal progenitor-like tu-
primitive, mesenchymal state that arises, at least in part, from
mor cells with basal/mesenchymal transdifferentia-
inadequate repair of DNA interstrand crosslinks (ICLs) (Wang
tion during murine BRCA1 BLBC development. et al., 2016). Specifically, p220, and its bound partners, BRG1
Growing DNA damage accompanied this malignant and FANCD2, and DNp63, with which it also communicates,
transformation. suppress ICL accumulation and MEC transdifferentiation. This
study explores these events both in cultured MECs and during
INTRODUCTION BRCA1 mouse mammary tumor development.

BRCA1 mutations carry a 40%–80% lifetime risk of developing


breast cancer (BC). Inadequate maintenance of mammary RESULTS
epithelial cell (MEC) genome integrity is a suspected driver of
this outcome (Chen et al., 2018; Huen et al., 2010; Silver and Liv- NUMB is a BRCA1-Interacting Protein
ingston, 2012). While p220, the primary disease-suppressing The BRCA1 C-terminal (BRCT) region is vital for the p220 DNA
BRCA1 product, exhibits multiple functions, like DNA damage repair function. It contains two adjacent BRCT repeat motifs
repair, the detailed events that lead to BRCA1 BC are largely that operate as DNA damage signal receptors (Gerloff et al.,
unknown. 2012; Wu et al., 2015). Currently, three BRCA1-associated

Cell 178, 135–151, June 27, 2019 ª 2019 Elsevier Inc. 135
Figure 1. NUMB Is a BRCA1 Partner
(A and B) Reciprocal interaction between BRCA1 and NUMB. 293T cell extracts transiently expressing HA-BRCA1 or myc-BRCA1 (A) or Flag-NUMB (long or short
isoform, B) were IP’d with anti-epitope antibody (Ab) and then immunoblotted (IB’d) with the indicated Abs.
(C and D) Endogenous BRCA1-NUMB interaction. MCF7 extracts were respectively IP’d with BRCA1 (C) or NUMB Ab (D), and the IPs were analyzed by IB.
(E and F) Effect of cisplatin or KU55933 on BRCA1-NUMB binding. Myc-tagged BRCA1-expressing (E) or naive 293T cells were or were not (UT) exposed to MMC
(100 ng/mL overnight) or KU55933 (10 mM overnight); extracts were IP’d with myc (E) or BRCA1 Ab (F) and analyzed by IB.
(G) In vitro BRCA1-BRCT/NUMB binding.
(H) NUMB mutations compromise NUMB/BRCA1 binding. Extracts of 293T cells transiently expressing FLAG-NUMB (long and short, WT or 2A) were IP’d with
FLAG Ab and the IPs analyzed by IB.
See also Figure S1.

proteins—CtIP, BACH1/BRIP1, and Abraxas—are known to NUMB also contains an SPTF and an SPTXXF motif (Fig-
bind to them. All contain pSXXF/pSPTF sequences, where the ure S1A). Immunoprecipitation (IP) of HA- or myc-tagged
serine is phosphorylated by ATM/ATR following DNA damage, BRCA1 in 293T cells showed that it coimmunoprecipitated
thereby sustaining their BRCT binding function (Kim et al., (coIP’d) with at least two endogenous human NUMB isoforms
2007; Manke et al., 2003; Wang et al., 2007; Yu and Chen, (long [L] and short [S]; Figures 1A and 1B). Moreover, endoge-
2004; Yu et al., 2003). Conceivably, these motifs participate in nous BRCA1 p220 and NUMB coIP’d (Figures 1C and 1D), an
signaling events that sustain MEC differentiation following effec- event that was stimulated by cell exposure to mitomycin C
tive DNA repair. (MMC) (Figures 1E and 1F). These results implied that BRCA1
Lacking evidence that any known BRCT-binding protein par- and NUMB co-participate in DNA damage control.
ticipates in MEC differentiation control, we initiated a search To test whether the BRCA1 BRCT motifs contribute to NUMB
for additional p220 BRCT binding proteins. Specifically, we binding, glutathione S-transferase (GST)-tagged BRCA1 frag-
screened database for heretofore undetected BRCA1-binding ments, collectively representing the entire BRCA1 sequence,
polypeptides that contain SPTF sequences (Manke et al., were assayed as candidate NUMB binding targets (Chen et al.,
2003; Yu et al., 2003) and encountered a stem cell biology-asso- 1998). GST-BRCT fragment F6 (aa 1314–1863) exhibited the
ciated protein, NUMB. Interestingly, NUMB is required for MEC strongest NUMB binding. Less impressive NUMB binding was
differentiation maintenance (Tosoni et al., 2015). detected with GST-F5 (aa 1005–1313) (Figure 1G). A clinically

136 Cell 178, 135–151, June 27, 2019


(legend on next page)

Cell 178, 135–151, June 27, 2019 137


relevant BRCA1 point mutation (M1775R) introduced into the (Dhami et al., 2013). Moreover, the long isoform was enriched
BRCT domain of HA-tagged full-length BRCA1 abolished in CD44low MECs, while the short form was overexpressed in
BRCA1/NUMB binding as it had done for Abraxas-, CtIP-, and mesenchymal (U2OS) cells (Figure S1E). When exposed to
BACH1-BRCA1 binding (Kim et al., 2007; Manke et al., 2003; cisplatin, the cytoplasmic/nuclear distribution of NUMB in
Wang et al., 2007; Yu and Chen, 2004; Yu et al., 2003). By U2OS and MCF7 cells did not change (Figures S1G and S1H).
contrast, HA-BRCA1 bearing a RING domain mutation (C61G) These data suggest that NUMB is both nuclear and cytoplasmic.
bound NUMB normally (Figure S1B). Therefore, one or both F6 While participating in certain types of DNA repair, BRCA1 con-
BRCT motif(s) bind(s) NUMB. centrates in multiple, small, nuclear foci in S/G2 cells (Scully
In response to DNA damage, the SPTF sequence(s) that bind et al., 1997) or single, larger BRCA1-containing foci in G1/G0
the BRCA1 BRCT motifs are phosphorylated by a suitable PIKK cells (Escribano-Dı́az et al., 2013; Lukas et al., 2011). In CD44low
kinase. Since this modification is essential for SPTF/BRCT in- MECs, NUMB often colocalized with BRCA1 and 53BP1 in these
teractions (Manke et al., 2003; Yu et al., 2003), we asked if single, large G0/G1 foci (Figures 2A–2D), which are also known
BRCA1-NUMB binding is NUMB phosphorylation dependent. as ‘‘53BP1 bodies.’’ They carry DNA damaged during a prior
Specifically, we IP’d HA-tagged NUMB, exposed the IP to an S/G2 period (Lukas et al., 2011). BRCA1/NUMB colocalization
exogenous phosphatase, and NUMB binding to BRCA1 was confirmed by confocal image analysis (Figure 2B and
decreased significantly (Figure S1C). ATM inhibitor addition Video S1). By contrast, frequent colocalization of BRCA1 and
also disrupted BRCA1-NUMB binding (Figure 1E), and mutage- NUMB in single, large foci was rare in S/G2 cells (Figures
nizing NUMB serines 361 and 634 to alanines led the resulting 2A–2F). Analogous results were obtained in MCF7 cells (Figures
double mutant (NUMB 2A) to exhibit reduced BRCA1 binding S2A and S2B).
(Figure 1H). Thus, endogenous NUMB appears to interact 53BP1 sustains non-homologous end joining (NHEJ) and sup-
directly and in a phosphorylation-dependent manner with one presses homologous recombination (HR)-driven double-strand
or both BRCA1-BRCT motifs and is, therefore, an authentic DNA break (DSB) repair in both G1 and S/G2 (Bunting et al.,
BRCA1 partner. 2010; Escribano-Dı́az et al., 2013; Orthwein et al., 2015). To
determine whether NUMB and/or BRCA1 loss results in an accu-
Nuclear NUMB Is a BRCA1 Partner Protein mulation of G1 DNA damage, we searched for 53BP1 foci in cy-
NUMB was first identified in Drosophila as a membrane protein clin A-negative cells. When NUMB or BRCA1 depleted, these G1
that regulates asymmetric versus symmetric cell division and cells overly accumulated large 53BP1 foci (three or more foci per
NOTCH signaling (Guo et al., 1996; Uemura et al., 1989). At cell), suggesting the development of additional G1 DNA damage
least four NUMB isoforms exist in mammalian cells; the (Figures 2E–2F, S2C, and S2D).
longest controls cell proliferation, and the shortest affects dif- ATM/ATR signaling is essential for the formation of BRCA1 or
ferentiation (Dho et al., 1999; Verdi et al., 1999). A mammary- 53BP1 foci after DNA damage. Therefore, we asked if reduced
specific Numb knockout led to an epithelial to mesenchymal ATM/ATR signaling abolishes NUMB focus formation. Cell expo-
transition (EMT), development of stem cell-like properties, sure to the ATM inhibitor, KU55933, suppressed single 53BP1
and tumorigenesis (Tosoni et al., 2015), much like the effects and NUMB focus formation (Figures 2G–2I). 53BP1 depletion
of BRCA1 depletion. Since endogenous NUMB binds also abolished BRCA1/NUMB focal colocalization (Figures 2J
BRCA1, we asked whether NUMB function is, at least in and 2K). BRCA1 depletion also abolished NUMB focus forma-
part, nuclear, and if so, whether it and BRCA1 co-participate tion, implying its BRCA1/53BP1 co-dependence (Figures S2E
in DNA damage control. and S2F).
Specifically, human MECs were fractionated into different These observations fit with previous reports implying that
subcellular fractions that were then assayed for NUMB (Xia BRCA1 loss promotes 53BP1-dependent NHEJ repair (Bunting
et al., 2006) (Figure S1D). Both long and short NUMB isoforms et al., 2010). In addition, following the prior development of
were detected in the cytoplasmic (S100) and soluble nuclear DNA damage, significantly more cells arrested in G1/G0 after
(S400) fractions (Figures S1E–S1H). Furthermore, the relative BRCA1 or NUMB depletion, consistent with G1 checkpoint acti-
abundance of NUMB in the latter was comparable to that in vation (Figure S2D). Thus, BRCA1/NUMB complexes respond to
the former. These findings were consistently observed in multi- spontaneously arising DNA damage, at least in part, during
ple cell lines (Figures S1E–S1H), in keeping with others’ results G1/G0.

Figure 2. NUMB-BRCA1 Colocalization


(A) Co-staining of NUMB and BRCA1 in WT CD44low MECs. Scale bar, 20 mm.
(B) Colocalization of BRCA1 and NUMB nuclear foci (arrows) in HME cells using confocal z stack microscopy. Scale bar, 20 mm. See also Video S1.
(C and D) Co-IF of NUMB/53BP1(C) or NUMB/Cyclin A (D) in WT CD44low MECs. Scale bar, 20 mm.
(E and F) Co-IF of 53BP1 and Cyclin A in CD44low MECs before and after NUMB depletion. >500 cells were analyzed in each experiment. See also Figures S2C
and S2D for statistics.
(G and H) Co-IF of NUMB and 53BP1 after mock treatment (DMSO, G) or KU55933 exposure (10 mM, 24 h, H). Scale bar, 10 mm.
(I) Effect of KU55933 on single-53BP1 body formation and NUMB/53BP1 colocalization.
(J) Co-IF of NUMB and BRCA1 in cells transfected with Siluc or Si53BP1. Scale bar, 10 mm.
(K) Effect of 53BP1 depletion on single-NUMB focus formation.
In (I) and (K), >200 cells were analyzed, and the data represent mean ± SD. Student’s t test was used to calculate statistical significance. **p < 0.01, ***p < 0.001
See also Figures S1 and S2 and Video S1.

138 Cell 178, 135–151, June 27, 2019


Figure 3. NUMB and MEC Differentiation
(A) Effects of overnight cisplatin exposure and NUMB depletion on CD44low MEC clonogenic survival.
(B) Western blot of NUMB in CD44low MECs before and after its short hairpin RNA (shRNA)-driven depletion.
(C) Cisplatin effect on clonogenic survival of MCF10A cells with NUMB siRNA depletion versus control upon expression of empty vector (EV), siRNA-resistant
NUMB-WT, or NUMB-2A.
(D) Western blot of NUMB in MCF10A cells after NUMB depletion or depletion and repletion with NUMB-WT or 2A.
(E) Western blot of NUMB and BRCA1 after transfecting CD44low MECs with the respective siRNAs.

(legend continued on next page)

Cell 178, 135–151, June 27, 2019 139


NUMB Sustains a DNA Damage-Associated decrease of the long and an increase of short isoform-encoding
Differentiation Program in MECs mRNA (Figure S3D). Using specific siRNAs, we selectively
Failed ICL repair can promote aberrant basal-mesenchymal trans- depleted each NUMB isoform and validated that the two bands
differentiation of cultured, basal-like human MECs (Wang et al., represent the two above-noted isoform species (Figure S3E).
2016). In addition, at least three proteins—BRCA1 and its part- To test further whether a suitable NUMB isoform switch and
ners, FANCD2 and BRG1—participate in ICL repair-driven MEC MEC differentiation maintenance are linked, we stably introduced
differentiation control (Wang et al., 2016). Thus, we asked if NUMB short cDNA (siRNA-resistant) or an empty vector into
NUMB is also required for ICL repair and found that depletion of CD44low MECs. Using a NUMB siRNA targeting its native 30 UTR,
NUMB in either CD44low or naive MCF10A MECs led to hypersen- the endogenous NUMB long isoform was efficiently depleted
sitivity to the ICL-inducing agents, cisplatin, and MMC (Figures while NUMB short cDNA expression was unaffected (Figure 3K).
3A–3D), much as has been detected in CD44low MECs after Depleting endogenous NUMB long or overexpressing
BRCA1, BRG1, or FANCD2 depletion (Wang et al., 2016). To NUMB short in otherwise unmanipulated cells led to only a
determine if ICL agent sensitivity was NUMB depletion depen- subtle MEC morphology effect, with most cells retaining their
dent, we expressed small interfering RNA (siRNA)-resistant wild- epithelial state (Figure S3F). By contrast, the combination of
type (WT) NUMB or NUMB-2A mutant cDNAs in MCF10A cells both triggered a clear change to mesenchymal morphology
and found that WT NUMB, but not the mutant, rescued cisplatin (Figure S3F), a marked shift from CD44low to CD44high cells
sensitivity (Figures 3C and 3D). This implied that NUMB phosphor- (Figures S3G and S3H), and reduced expression of E-Cadherin
ylation participates in sustaining ICL repair. Therefore, NUMB, and overexpression of Vimentin (Figure 3K), similar to the re-
which was not formerly known to engage in DNA repair, much sults of a loss of BRCA1, BRG1, or FANCD2 expression.
less ICL repair, is necessary for ICL repair in these cells. NUMB long depletion followed by NUMB short overexpression
We also asked whether BRCA1 and NUMB cooperate in ICL also led to a decrease in DNp63 expression (Figure 3K), which
repair. When CD44low MECs were depleted of both NUMB and is a driver of DNA damage-associated MEC dedifferentiation
BRCA1, they became more sensitive to cisplatin or MMC than af- (Wang et al., 2016). Interestingly, two human basal-like BC
ter depleting NUMB or BRCA1 alone. This suggests that BRCA1 cell lines also overexpressed the short isoform, unlike untrans-
and NUMB cooperate in ICL repair (Figures 3E–3G). In addition, formed MECs (Figure S3I), suggesting that a general isoform
NUMB depletion impaired nuclear focus formation by the key switch might accompany MEC tumorigenesis. Thus, like
ICL repair protein, FANCD2 (Figures S3A and S3B). Moreover, BRCA1 depletion, purposely altering the ambient ratio of
NUMB depletion in MECs led to failed FANCD2 monoubiquitina- NUMB long/short isoform expression in human MECs, per-
tion following MMC exposure (Figures 3H, 3I, and S3C). Thus, turbed their differentiation state. Moreover, NUMB, like
NUMB, like BRCA1 (Garcia-Higuera et al., 2001), promotes BRCA1 and its partners, BRG1 and FANCD2, is necessary
FANCD2 recruitment to ICL-damaged sites by supporting its for ICL repair, failure of which elicits an EMT.
monoubiquitination, a key event during ICL repair.
Since inadequate ICL repair threatened the normal differentia- HES1 Links BRCA1 and NUMB in ICL Repair
tion state of CD44low MECs, we asked if NUMB is required to The finding of a NUMB contribution to ICL repair and MEC differ-
maintain a normal human MEC differentiation state, much like entiation maintenance prompted the question of whether
its bound partner, BRCA1. We analyzed CD44low HME cells NOTCH signaling, a NUMB-influenced function (Flores et al.,
(epithelial) and their BRCA1-depleted, aberrantly transdifferenti- 2014), underlies the latter. Of note, the NOTCH pathway tran-
ated CD44high derivatives (Wang et al., 2016). The former ex- scription factor (TF), HES1, normally interacts with certain Fan-
pressed relatively high levels of E-Cadherin and DNp63 and a coni pathway core proteins and is required for proper ICL repair
low level of ZEB1, while CD44high cells revealed the opposite (Tremblay et al., 2008). Moreover, HES1 depletion rendered
phenotype, reflecting their respective epithelial and mesen- CD44low MECs hypersensitive to cisplatin and MMC exposure
chymal phenotypes (Figure 3J). (Figures 4A–4C).
NUMB also underwent a dramatic isoform switch as CD44low Like NUMB depletion, HES1 depletion also impaired cisplatin/
cells experienced BRCA1 depletion-driven EMT (Figure 3J). ICL-driven FANCD2 focus formation in CD44low MECs (Fig-
CD44low HME cells expressed a relatively high level of the ure S4A). By contrast, neither HES1 nor NUMB depletion altered
NUMB long and a low level of the short isoform, while BRCA1- etoposide sensitivity (data not shown). In addition, HES1 deple-
depleted, aberrantly transdifferentiated CD44high cells revealed tion led to defective MMC-driven FANCD2 monoubiquitination
the opposite pattern (Figure 3J). qPCR also revealed a significant (Figures 3H and 3I), similar to either BRCA1 or NUMB depletion.

(F and G) Clonogenic survival after depleting NUMB or BRCA1 or both in CD44low MECs exposed overnight to increasing concentrations of cisplatin (F) or
MMC (G).
(H and I) Effect of NUMB or HES1 depletion on monoubiquitination of FANCD2 after ICL damage in CD44low MECs (H) or in U2OS cells (I). Cells were transfected
with SiLuc or SiNUMB or SiHES1 for 48 h and exposed to MMC (100 ng/mL overnight). The lysates were IB’d with the indicated Abs.
(J) Western blotting for ZEB1, E-cadherin (E-CAD), DNp63, and long and short NUMB in undepleted, acutely depleted CD44low (DOX+) and chronically BRCA1-
depleted CD44high MECs.
(K) Western blotting for NUMB/E-CAD/VIMENTIN/DNp63 after depleting NUMB in CD44low MECs that overexpressed siRNA-resistant NUMB short.
The data (A, C, F, and G) represent mean ± SD. Two-way ANOVA analysis was used for statistical significance. N.S., not significant; *p < 0.05, **p < 0.01,
***p < 0.001, ****p < 0.0001.
See also Figure S3.

140 Cell 178, 135–151, June 27, 2019


Figure 4. HES1 Facilitates BRCA1/NUMB-
Dependent ICL Repair
(A and B) Effects of overnight cisplatin (A) or MMC
(B) exposure and HES1 depletion on CD44low MEC
clonogenic survival. Data represent mean ± SD;
two-way ANOVA analysis was used for statistical
analysis. ****p < 0.0001.
(C) IB of HES1 after transfecting the indicated
siRNAs into CD44low MECs.
(D) Interaction between myc-tagged BRCA1 and
HES1. Cell extracts from myc-BRCA1-expressing
293T cells were incubated with myc Ab versus
control IgG and analyzed by IB.
(E) Interaction between NUMB and BRCA1/HES1/
FANCD2/BARD1. A FLAG-tagged NUMB WT or 2A
mutant was expressed in 293T cells, and cell ly-
sates were IP’d with FLAG Ab and IB’d with the
indicated Abs.
(F) The effect of KU55933 (10 mM) or MMC
(100 ng/mL) on interaction between NUMB and
HES1.
(G) NUMB and HES1 are required for interaction
between BRCA1, CTIP, and FANCD2. 293T cells
were cotransfected with myc-BRCA1 and the
indicated SiRNAs. The corresponding cell extracts
were IP’d with myc Ab followed by IB.
See also Figure S4.

plex(es) that contribute(s) to ICL repair


and (2) the absence of any of them com-
promises BRCA1-NUMB-HES1 complex
formation.
BRCA1 also tethers both CtIP and
FANCD2 to damaged chromatin (Garcia-
Higuera et al., 2001; Yu et al., 2006), and
the latter proteins are both required for
crosslink lesion resolution (Murina et al.,
2014; Unno et al., 2014). Therefore, since
HES1-NUMB-BRCA1 complex formation
HES1 expression was also lower in BRCA1-depleted CD44high depends upon all three proteins being simultaneously available,
than in mock-treated CD44low MECs (Figures S4B and S4C). we asked if HES1 or NUMB is also required for the interaction of
These observations fit well with the finding that BRCA1-depleted FANCD2 and CtIP with BRCA1. Indeed, when either NUMB or
CD44high MECs also manifest a defect in ICL repair (Wang HES1 was depleted, BRCA1/CtIP/FANCD2 complex formation
et al., 2016). and the capacity for ICL repair (Figures 3 and 4) also decreased.
To probe the context in which HES1 functions in ICL repair, we Therefore, BRCA1/FANCD2/CtIP/NUMB/HES1 interactivity may
asked if BRCA1, like other FANC proteins, interacts with HES1. be necessary for proper ICL repair and, thus, for MEC differenti-
Indeed, BRCA1 coIP’d with endogenous HES1, FANCD2, and ation maintenance.
NUMB (Figure 4D). NUMB-WT also coIP’d with HES1 and To assess HES1 participation in MEC differentiation mainte-
FANCD2 (Figure 4E), and the ATM inhibitor KU55933 disrupted nance, we stably depleted it in CD44low MECs and succeeded
BRCA1-NUMB (Figure 1E) and NUMB-HES1 binding (Figure 4F). in isolating multiple, derivative CD44high clones. The latter ex-
Importantly, mutations of NUMB phosphorylation sites (in the 2A hibited mesenchymal morphology while CD44low cells remained
mutant) reduced NUMB binding to HES1 and to other DNA dam- largely epithelial. Western blotting also showed that HES1-
age repair proteins—i.e., BRCA1, FANCD2, and BARD1 (Fig- depleted CD44high cells exhibited high ZEB1 and Vimentin and
ure 4E)—suggesting that NUMB phosphorylation contributes low E-Cadherin expression, consistent with EMT development
to BRCA1-NUMB-HES1 complex integrity. Indeed, BRCA1 (Figures S4E and S4F). These HES1-depleted CD44high cells
bound HES1 weakly after siNUMB expression (Figure 4G). also exhibited spontaneously arising DNA damage in the form
BRCA1/NUMB coIP was also perturbed when HES1 was of anaphase bridges (Figure S4G). Therefore, similar to BRCA1,
depleted (Figure 4G). These results imply that (1) BRCA1, FANCD2, BRG1, and NUMB, HES1 also sustains an ICL DNA
NUMB, and HES1 co-populate a common biochemical com- damage repair-associated differentiation program in MECs.

Cell 178, 135–151, June 27, 2019 141


Figure 5. BRCA1 Loss in Luminal MECs Promotes Development of Basal-like Mammary Tumors
(A) Schematic diagrams depicting Cre-mediated excision of either a floxed transcriptional Stop cassette (STOP) in the R26Y reporter, floxed exons 2–10 in the
Trp53L allele, and floxed exons 22–24 in the Brca1L allele, leading to YFP expression and disruption of Trp53 and Brca1.
(B) FACS analysis of MECs from virgin females with the indicated genotypes 3 days or 3 weeks after intraductal injection of Ad-K8-Cre. The YFP-marked/lineage
negative MECs were analyzed with CD24 and CD29 Abs.
(C) Statistical analysis of aberrant YFP+ MECs in the basal gate 3 weeks post injection (in B). ***p % 0.005.
(legend continued on next page)

142 Cell 178, 135–151, June 27, 2019


To investigate the hypothesis that a CD44low to CD44high MEC expression profiles to expression signatures of human and
transition represents a step during mammary tumorigenesis, we murine BC subtypes (Pfefferle et al., 2013). The mouse Brca1-
compared the soft agar colony formation ability of CD44low and deleted tumors most closely resembled the human basal-like
CD44high cells that arose after BRCA1 depletion. CD44high cells breast cancer (BLBC) subtype and murine MycEx, p53null-Basa-
formed agar colonies much more efficiently (>100-fold) than lEx, and C3TagEx subtypes, each a model of BLBC (Pfefferle
CD44low cells (Figures S4H and S4I). This is consistent with et al., 2013) (Figures 5F and S5D).
DNA damage-associated transdifferentiation that appears after Employing an intraductal injection approach, p53 loss, alone in
BRCA1 depletion, representing a step toward BC development. Krt8+ luminal MECs resulted in Claudin-low mammary tumors,
possibly due to an acquisition of secondary mutations (e.g.,
Loss of BRCA1 Function in Luminal MECs Promotes 9qA1 [Yap1] or 6qA2 [Met] amplification; Tao et al., 2017). How-
Basal-like Mammary Tumorigenesis ever, genes from such regions in p53/BRCA1-deficient tumors
To understand how BRCA1 loss affects luminal MEC fate and lacked signs of amplification-driven overexpression (Figure S5E).
promotes BC development, we established a mouse BRCA1 Thus, co-induced loss of BRCA1 and p53 drove luminal MECs to
BC model. It involved the intraductal injection of a Cre-express- develop BLBC and not Claudin-low cancer. Others argue that
ing adenovirus under Krt8 promoter control (Ad-K8-Cre), leading human Claudin-low BC is not a prevalent BRCA1 disease (Mada-
to floxed Brca1 and/or Trp53 knockout and simultaneous activa- ras et al., 2016). Lastly, analysis of gene sets that reflect MaSC
tion of a conditional YFP-expressing Cre-reporter (Rosa26- (Lim et al., 2010) and EMT (Gotzmann et al., 2006) programs (Fig-
LSL-YFP [R26Y]) in luminal MECs (Tao et al., 2014, 2017) (Fig- ure 5G) suggests that these BRCA1-deficient tumor cells, being
ure 5A). Of note, Cre-mediated excision of floxed exons 22–24 derived from luminal MECs originally, acquired aberrant basal
of mouse Brca1 (McCarthy et al., 2007), resulted in disruption and mesenchymal (b&m) gene expression programs after
of the NUMB/CtIP/Abraxas/BACH1-interacting BRCA1 BRCT BRCA1 loss.
domains.
Speculating that a BRCA1-dependent MEC differentiation Loss of BRCA1 Function Elicits a Luminal to Basal/
program affects normal MG biology and that its loss will impair Mesenchymal Change
luminal MEC fate—e.g., during the early stage of BRCA1 BC Our mouse model revealed the emergence of premalignant
development—we studied YFP-marked MEC fate soon after basal-like MECs and mammary cancer from luminal MECs
Ad-K8-Cre injection (Figure 5B). This vector restricts Cre expres- following induced BRCA1 loss. To determine if these p53/
sion to luminal MECs (Tao et al., 2017); thus, YFP+ MECs were BRCA1-deficient MECs had acquired an abnormal in vivo differ-
detected solely in the LinCD24hiCD29lo luminal fluorescence- entiation state, we FACS-purified YFP+ MECs from the corre-
activated cell sorting (FACS) gate 3 days after injection, in sponding mice one month after Ad-K8-Cre injection. These cells
Trp53L/L;Brca1L/L;R26Y (PBY) females, as well as in R26Y (WT) then underwent RNA sequencing (RNA-seq). Several mesen-
and Trp53L/L;R26Y (PY) controls (Figure 5B). Three weeks after chymal/EMT genes (e.g., Vim, Zeb1, Zeb2) were upregulated
injection, we began to detect YFP+ MECs in the basal MEC (unlike in the case of p53-deficient or WT MECs) (Figure 6A).
gate in injected PBY females, but not in injected R26Y and PY Furthermore, GSEA revealed that multiple, mesenchymal/EMT-
controls (Figures 5B and 5C). Thus, induced BRCA1 loss in related gene sets were enriched, again only in the BRCA1/
luminal MECs likely led to a luminal-to-basal cell fate change p53-deficient MECs (Figures 6B and S5F), accompanied by
soon thereafter. enrichment of a mammary LP gene set (Lim et al., 2010) (Fig-
We also detected more YFP+ MECs in injected PBY mice than ure 6B). The latter may reflect the LP origin of BRCA1 BLBC
in controls (Figure 5B). Quantitation of YFP+ MECs in females (Lim et al., 2009; Molyneux et al., 2010; Proia et al., 2011).
2 weeks post injection confirmed the presence of more prolifer- To further assess the aberrant differentiation state of p53/
ating p53/BRCA1-deficient MECs compared to p53-deficient or BRCA1-deficient MECs, we purified YFP+ MECs by FACS
WT MECs (Figure S5A). These data imply that (1) BRCA1 main- 7 days after Ad-K8-Cre injection and cultured them as organo-
tains a luminal MEC fate in vivo and (2) a corresponding loss of ids. Although their organoid-forming efficiency was similar to
a luminal and the emergence of a basal-like cell fate accompa- that of p53-deficient and WT luminal MECs, the doubly deficient
nied BRCA1 loss. The latter correlated with enhanced BRCA1/ cells were larger when cultivated in the presence (rather than the
p53-deficient MEC proliferation. absence) of B27, which includes progesterone (Figures S5G–
All Ad-K8-cre-injected PBY female mice eventually developed S5I). When cultivated in B27-free medium, only the doubly (and
mammary tumors (first palpated 8–12 months post injection; Fig- the p53-deficient) MECs still formed organoids. Moreover, the
ures 5D and S5B). Immunofluorescence (IF) staining showed that organoid-forming efficiency and size of the doubly deficient
the resulting tumor cells expressed basal markers (e.g., K5, K14) MECs were greater than those of p53-deficient and WT MECs
(Figures 5E and S5C). By gene set enrichment analysis (GSEA) (Figures S5H and S5I). B27-independent growth of BRCA1-defi-
(Subramanian et al., 2005), we compared their microarray cient MECs was observed previously (Lim et al., 2009; Sau et al.,

(D) Kaplan-Meier tumor-free plot for PBY female mice (n = 8) injected with Ad-K8-Cre.
(E) IF staining showing expression of YFP (luminal origin), K5 (basal marker), and K8 (luminal marker) in a representative mammary tumor developed in mice in (D).
(F) GSEA for comparisons of microarray data from tumors in (D) to gene sets representing different human BC subtypes.
(G) GSEA plots showing significant enrichment of MaSC and EMT-related gene sets in tumors in (D) relative to WT Krt8+ luminal MECs.
See also Figure S5.

Cell 178, 135–151, June 27, 2019 143


Figure 6. Loss of BRCA1 Function Elicits a Luminal to Basal/Mesenchymal Fate Change and Accumulation of DNA Damage
(A) Expression (relative to that of WT cells, normalized = 1, from bulk RNA-seq) of select EMT-related genes in YFP+ cells sorted from MGs of PBY or PY or R26Y-
only females 1 month after Ad-K8-Cre injection.
(B) GSEA plots showing enrichment of EMT and LP-related gene sets in Trp53/Brca1 null MECs compared to WT or Trp53 null MECs based on RNA-seq data
as in (A).
(C) qRT-PCR data revealing an upregulation of selected b&m genes in Trp53/Brca1 null organoid cells compared to those obtained from PY or R26Y-only control
females following Ad-K8-Cre injection. Data represent mean ± SEM.
(D) IF staining confirming upregulation of Keratin 14 (K14) and Vimentin (VIM) expression in Trp53/Brca1 null (bottom) versus Trp53 null (top) organoid cells as in
(C). Scale bar, 100 mm.

(legend continued on next page)

144 Cell 178, 135–151, June 27, 2019


2016). The differentiation status of MECs in organoids, when many cells in the LP cluster at 5 months had begun to express
characterized by qRT-PCR analysis, also revealed an upregula- Sox4 (Figure 6F), a known EMT inducer (Tiwari et al., 2013),
tion of expression of b&m genes only in p53/BRCA1-deficient and several b&m genes initially found to be highly expressed in
cells (Figures 6C and 6D). the Basal-like cluster at 2 weeks (e.g., Sfn, Igfbp7, Serpinh1;
We also detected 53BP1+ cells in these p53/BRCA1-deficient compare Figure 6E cluster B and Figure 6F cluster LP). Overall,
organoids, a sign of ongoing DNA damage. Furthermore, 53BP1 these and bulk RNA-seq data from the YFP+ premalignant
foci were more abundant in cells that manifest aberrant differen- MECs suggest that (1) HR LPs are the main MEC population de-
tiation (i.e., exhibited robust K14 expression) (Figure S5J). Thus, tected following the loss of BRCA1 and p53 and that (2) YFP+,
aberrant differentiation of p53/BRCA1-deficient MECs was p53/BRCA1-deficient LPs, and basal-like cells continued to un-
accompanied by DNA damage, as in BRCA1-depleted human dergo b&m transdifferentiation over time.
MECs (Wang et al., 2016). In the two tumors analyzed, we observed 1–2 cluster(s) of tu-
mor epithelial cells, as well as a cluster of mesenchymal cells
Single-Cell Expression Analysis during the Evolution of (possibly reflecting stromal cells, though some of these cells
BRCA1-Deficient Tumors may be tumor epithelial cells undergoing more complete mesen-
To assess the fate change of p53/BRCA1-deficient luminal chymal transdifferentiation) (Figures 6G and 6H). The tumor
MECs in vivo, we performed single-cell (sc)RNA-seq analysis epithelial cell clusters (Figures 6G and 6H and Data S1) largely
(10X Genomics) on YFP+ MECs FACS-purified from injected reflect their LP origin, given that they expressed LP/alveolar
MGs and from mammary tumors that arose in Ad-K8-Cre-in- luminal genes (e.g., Sox9, Csn3, Wfdc18, Lcn2, Mfge8, Phlda1,
jected PBY females. We used the Seurat tool (Butler et al., Tm4sf1, Fxyd3). Furthermore, many cells in these clusters also
2018) to analyze the top 50 enriched genes in each cell cluster expressed mesenchymal (e.g., Sparc, Postn, Bgn, Mgp, Igfbp7,
(Data S1). Serpinh1, Aebp1) and EMT inducer genes such as Sox4, sug-
Two weeks after Ad-K8-Cre injection, we found that YFP- gesting that they might be undergoing an EMT.
marked MECs included five luminal MEC clusters. The largest Using another t-distributed stochastic neighbor embedding
four were composed of hormone receptor (HR)+ luminal cells (t-SNE) plotting approach (10X Genomics), we observed that tu-
that expressed Prlr, Pgr, and an estrogen receptor co-factor mor epithelial and mesenchymal cell clusters self-organized in a
gene, Cited1 (Figure 6E). There was also a cluster that expressed manner that reflected transdifferentiation from a luminal (purple-
Csn3 and Wfdc18 (Figure 6E and Data S1), likely reflecting HR lettered genes) toward a mesenchymal fate (red- and green-
MECs with an LP/alveolar luminal phenotype (Giraddi et al., lettered genes) (Figure S6A). These included clusters that
2018). There was also a small cluster of MECs that expressed manifested signs of an LP origin (purple lettering), as reflected
b&m genes (e.g., Acta2, Krt5, Krt14, Sparc, Postn, Igfbp7, by their expression of LP/alveolar luminal genes (e.g., Sox9,
Serpinh1, Sfn) and myoepithelial differentiation genes (e.g., Elf5, Kit, Csn3, Wfdc18). Yet other clusters seemed to be gradu-
Myl9, Mylk, and Tpm2) (B cluster; Figure 6E and Data S1). The ally losing expression of the pan-luminal gene Krt8, the EMT in-
presence of this cluster is consistent with that of a sizeable num- hibitor/LP gene Elf5 (Chakrabarti et al., 2012), and the epithelial
ber of YFP+ MECs in the basal FACS gate at this early post-Ad- gene Cdh1 while gaining expression of the basal gene (Krt14)
K8-Cre injection stage (Figures 5B and 5C). and eventually certain EMT-associated TF-encoding (e.g.,
5 months post injection, the YFP-marked MECs now included Zeb1, Twist1, Snai1, Sox4) and mesenchymal genes (e.g.,
a large LP cluster (HR, high expression of Csn3 and Wfdc18) Cdh2, Vim, Sparc, Postn, Aebp1). At the top of Figure S6A, in
and a much smaller HL cluster (again expressing Prlr and Cited 1; both tumors, an LP-like tumor cell cluster is on the left side, an
Figure 6F). There was also a distinct EMT cluster expressing EMT-like tumor cell cluster is in the middle/middle right, and a
mesenchymal genes (e.g., Sparc, Postn, Aebp1, Figure 6F and mesenchymal cell cluster is on the right.
Data S1). Cells in this cluster may be related to those in the Compared to cells in the mesenchymal clusters, which might
basal-like cluster observed at 2 weeks (Figure 6E; cluster B) be stromal cells, cells in the EMT clusters are tumor cells that
that subsequently underwent EMT and expressed mesenchymal manifest signs of an epithelial origin, as they expressed the
genes (e.g., Sparc, Postn) and retained expression of the myoe- above-noted LP/alveolar luminal genes (e.g., Csn3, Wfdc18,
pithelial gene, Tpm (Figures 6E and 6F). Sox9; Figure S6A). In tumor 1, cells in the EMT cluster also ex-
In addition, multiple highly enriched RNAs in the 5-month LP pressed Krt8 (Figure S6A, left).
cluster were the same as those in the LP cluster 2 weeks post in- Interestingly, cells in the B/M (basal/mesenchymal) and EMT
jection, possibly reflecting a common LP origin (Figures 6E and clusters expressed several genes, defined above, that appear
6F and Data S1). However, unlike MECs analyzed at 2 weeks, to reflect cell fate dynamics over time (e.g., basal: Sfn;

(E–H) Analysis of scRNA-seq data based on Seurat (top: t-SNE plots; bottom: heatmaps of select cell clusters). Data obtained 2 weeks after Ad-K8-Cre injection in
PBY females were based on sorted YFP+ MECs (E), data for 5-month chasing was based on sorted YFP+ MECs and CD45+ leukocytes (F), and data for tumors
were based on tissues (cells unfractionated) from two different mammary tumors (G and H). Heatmaps for the top 50 enriched genes in each cell cluster are shown
(epithelial and mesenchymal clusters only). HL, hormone receptor-positive luminal cell cluster; LP, luminal progenitor-like cell cluster; B, basal-like cell cluster;
EMT, epithelial-to-mesenchymal transition cell cluster; M, mesenchymal cell cluster. See also Data S1 for the top 50 enriched genes
(I) Representative IF staining of 53BP1(green arrowheads) in MGs or tumors from PBY female mice at different time points after Ad-K8-Cre injection. Scale bar,
50 mm. See also Figure S6D for quantification.
See also Figures S5 and S6 and Data S1.

Cell 178, 135–151, June 27, 2019 145


mesenchymal: Sparc, Postn, Aebp1) (Figures 6E–6H and S6A). etoposide exposure, which induces topoisomerase II-linked
These clusters may be clinically relevant, since, from public DSB as opposed to ICL (Baldwin and Osheroff, 2005), led to a
database sources, we found that among human BLBC cases, significant increase in mRNA expression of basal (Trp63 and
those with higher expression levels of any of these four genes Krt5), but not mesenchymal markers (Vim and Zeb1) (Figures
experienced a worse clinical outcome (Figure S6B). 7B, S7A, and S7B), in keeping with our previous observations
(Wang et al., 2016). Taken together, these data indicate that
Loss of BRCA1 Function Causes the Accumulation of ongoing DSB-like DNA damage triggers a luminal to basal cell
DNA Damage fate change in primary MECs and that ICL DNA damage induced
To determine whether the kinetics of cell fate change correlate by cisplatin or a loss of BRCA1 function phenocopy one another,
with an accumulation of DNA damage in BRCA1/p53-deleted leading to the acquisition by these transdifferentiated basal cells
MECs, we stained MGs from Ad-K8-Cre-injected PBY females of a mesenchymal fate.
for DNA damage markers. A small number of 53BP1+ cells could Next, we asked if certain subunits of BRCA1-containing multi-
already be detected 1 month after injection (Figure 6I). Fewer still protein complexes participate in DNA damage-associated dif-
were detected in p53-depleted MGs, and none were found in WT ferentiation maintenance in primary mouse luminal MECs. We
MGs (Figures S6C and S6D). disrupted NUMB expression in p53-deficient luminal organoid
5 months after BRCA1/p53 deletion, the abundance of cells, using CRISPR/Cas9 (Figure S7D). As a positive control,
53BP1+ cells had increased significantly in PBY MGs (Figures we similarly disrupted BRCA1 in another aliquot of the same or-
6I and S6D). Subsequently, in the resulting tumors, most tumor ganoid cells and confirmed the absence of BRCA1 nuclear foci in
epithelial cells exhibited nuclear 53BP1 staining (Figures 6I and proliferating organoid MECs (Figure S7C). As negative controls,
S6E, green arrowheads), whereas the non-epithelial cells (Fig- we targeted either the mouse Rosa26 locus (leading to DSB in a
ure S6E, white arrowheads) in these tumors did not. There was neutral genomic region) or an unrelated plant gene (which is not
an analogous pattern of g-H2AX accumulation in p53/BRCA1- expected to induce DNA breaks in mouse cells). Similar to
deficient MECs and tumor cells (Figure S6F). BRCA1-loss, NUMB loss resulted in larger organoids by com-
Collectively, these data imply that normal differentiation parison with controls (Figures 7C and S7E); qRT-PCR analysis
maintenance by BRCA1 in luminal MECs is a major suppressor revealed upregulation of both b&m gene expression in NUMB-
of BLBC development. By contrast, loss of BRCA1 (in a p53- deficient organoid cells, like what occurred in BRCA1-deficient
deficient background) in primary luminal MECs resulted in cells (Figure 7D).
the emergence of aberrantly differentiated, progesterone- In addition, loss of either NUMB or BRCA1 led to an accumu-
independent, hyperproliferating, b&m-like cells. BRCA1 defi- lation of DNA damage in aberrantly differentiated MECs, as
ciency-linked aberrant MEC differentiation gave way to BLBC measured by comet assay (Figures 7E, 7F, and S7F). More spe-
development, and both were accompanied by abundant DNA cifically, their DNA damage levels were significantly higher than
damage that likely arose, at least in part, from a defect in those of MECs exposed to a Rosa26 gRNA, which should only
BRCA1 function. have elicited a low level of DSB (Figure S7F). Thus, the DNA dam-
age associated with loss of either NUMB or BRCA1 was not due
Inadequate DNA Repair Promotes Aberrant to irrelevant CRISPR editing.
Transdifferentiation in Primary Luminal MECs Lastly, since HES1 links BRCA1 and NUMB to ICL DNA repair,
To further test if failed BRCA1 DNA repair function results in we also tested if loss of HES1 in luminal MECs phenocopies the
aberrant, ex vivo MEC transdifferentiation, we exposed Trp53/ outcome of either BRCA1-loss or NUMB-loss. HES1 expression
organoids derived from sorted YFP+ luminal MECs of Ad-K8- was disrupted in p53-deficient luminal organoid cells by
Cre-injected PY mice to cisplatin (0.2 mM, 1 week). After drug CRISPR/Cas9 (Figure S7G). Similar to BRCA1 or NUMB loss,
withdrawal, the cells were maintained in organoid culture for HES1 loss also resulted in larger organoids (Figures S7H and
14 days. By comparison with mock-treated cells, the cisplatin- S7I) and an upregulation of b&m gene expression (Figure S7J).
treated cells experienced a dramatic increase in basal-like orga- Therefore, both NUMB and HES1 recapitulated the role of
noids with upregulated K5 expression (Figures 7A and S7A). BRCA1 in suppressing a luminal to b&m transition of primary
When compared with mock-treated cells, 53BP1 staining re- mouse luminal MECs. Importantly, in human BCs, both NUMB
flected persistent cisplatin DNA damage in these cells (Figures and HES1 are expressed at significantly lower levels in BLBC
7A and S7B). than other subtypes (except the luminal B subtype; Figure S7K),
We also assayed for b&m marker gene expression and de- consistent with these two genes playing supporting roles as ICL-
tected increased expression of most of these genes in the repair proteins during the suppression of BLBC development.
cisplatin-treated organoids. Expression had risen to a level com-
parable to that detected in drug-free BRCA1/p53-deficient orga- DISCUSSION
noid cells (Figure 7B), suggesting that cisplatin treatment had, at
least in part, phenocopied BRCA1 loss in p53-deficient luminal BRCA1 prevents in vivo DNA damage from promoting transdif-
MECs. The same was true in human MECs (Wang et al., 2016). ferentiation of normal luminal MECs to basal-like cells that
To determine whether this cell fate change resulted from DNA manifest a mesenchymal phenotype. Aberrant luminal MEC dif-
damage in general or more specifically from ICL DNA damage, ferentiation likely contributes to BRCA1 BLBC development (Lim
we exposed p53-deficient luminal organoids to etoposide and et al., 2009; Molyneux et al., 2010; Proia et al., 2011). Others have
then subjected the cells to the same analyses. Unlike cisplatin, detected heterogeneity in the results of primary MEC analysis

146 Cell 178, 135–151, June 27, 2019


Figure 7. Inadequate ICL Repair Pro-
motes Luminal to Basal/Mesenchymal
Transdifferentiation
(A) IF staining for K5 and 53BP1 in organoids (as in
Figures 6C and 6D) with or without cisplatin or
etoposide treatment. Scale bar, 100 mm. Quanti-
tation of K5+ and 53BP1+ organoids is shown in
Figures S7A and S7B.
(B) The effect of cisplatin or etoposide treatment on
b&m gene expression in Trp53 null luminal orga-
noid cells by qRT-PCR analysis. Data represent
mean ± SEM.
(C) Size assessment of Trp53 null luminal organo-
ids following CRISPR/Cas9-mediated editing of
Brca1, Numb, or Rosa26 (control). Data represent
mean ± SEM. **p % 0.01.
(D) The effect of CRISPR/Cas9-mediated editing of
Brca1 or Numb on b&m gene expression in Trp53
null luminal organoid cells compared to that of
Rosa26 editing (control).
(E and F) Quantitation of the incidence of cells
with comet tails (E) and relative comet tail length
(F) (following the indicated CRISPR/Cas9-medi-
ated gene editing). Data represent mean ± SD;
***p % 0.005, ****p % 0.001.
(G) A proposed model of the ICL-dependent dif-
ferentiation program in MECs.
See also Figure S7.

steps that lead to BRCA1-associated


BC. This process operates in humans
and in mice and is summarized in
Figure 7G.
Aberrant LPs exist both in human
BRCA1 mutation carriers and in mouse
models (Lim et al., 2009; Molyneux et al.,
2010; Proia et al., 2011). However, the
mechanism by which a BRCA1 functional
defect results in aberrant luminal MEC dif-
ferentiation followed by BLBC has been
unclear.
Four generic questions were posed
during this study. First, does an inte-
grated, BRCA1-containing, ICL repair-
promoting process contribute to MEC
differentiation control? If so, are any of
performed on cells of BRCA1+/+ and tumor-free BRCA1 mutation its participants newly detected, and how do they operate?
carriers (Nakshatri et al., 2015). Therefore, we undertook a pro- Does such a process sustain normal MEC differentiation (1) in
spective, comparative analysis of primary MECs from BRCA1- multiple mammalian species, (2) both in vitro and in vivo, and
mutated and relevant control mice to determine if aberrant (3) by executing the same steps in both instances? Lastly, is its
MEC differentiation accompanies BRCA1 loss in tumor-free failure material to BRCA1 BC development?
mammary tissue. The participants in this process are, at a minimum, BRCA1 and
Evidence of such a process was detected. Its central out- four BRCA1-associated proteins—two established partners,
comes mimicked the aberrant DNA damage-associated trans- BRG1 and FANCD2, a formerly suspected one, HES1 (Buckley
differentiative phenotypes induced in human MECs by BRCA1 et al., 2013), and a new one, NUMB. Yet others may exist.
p220 depletion or depletion of certain p220-associated proteins Some contribute to DSB repair (Houghtaling et al., 2005; Scully
(Wang et al., 2016). Where tested, these DNA damage-associ- et al., 1997), and all contribute to ICL repair (Kim and D’Andrea,
ated outcomes developed rapidly after in vivo BRCA1 luminal 2012; Tremblay et al., 2008; Wang et al., 2016). Depletion of any
cell depletion and persisted during BRCA1 mouse mammary member of this group not only compromised ICL repair, but
tumorigenesis. Therefore, they may be signs of molecular mis- also led to the mesenchymal transdifferentiation of cultured,

Cell 178, 135–151, June 27, 2019 147


basal-like human and primary mouse luminal MECs analyzed model, the tumor may be polyclonal, with cells in the LP and
during mouse BRCA1 mammary tumorigenesis. Basal-like clusters progressing independently and then jointly
All of these associated proteins not only contribute to ICL forming a tumor (Figure S7N).
repair, they likely do so in an organized, possibly even integrated This notwithstanding, both LPs and basal-like cells revealed
manner. In particular, much of the BRCA1 binding to the above- increasing mesenchymal gene expression that correlated with
noted partner proteins depended upon it also binding to each and possibly even depended upon an accumulation of endoge-
subunit individually. Leaving one out triggered a defect in multi- nous ICL or ICL-like DNA damage. Taken together, these find-
partner binding to BRCA1. BRCA1/multisubunit complex forma- ings suggest that linked defects in BRCA1-driven multiprotein
tion is, therefore, subunit interdependent, as if intact complex complex formation, repair of ICL or ICL-like DNA damage, and
formation represents a well-organized and cooperative process. MEC transdifferentiation suppression conspire to promote
Each subunit that was tested also appeared to be necessary for BRCA1 tumorigenesis. Our results also provide the first longitu-
the maintenance of normal, cultured basal-like human MEC dinal mechanism-related picture of certain key events that give
differentiation and proper ICL repair. Since ICL damage alone rise to BRCA1 mammary cancer.
triggered aberrant MEC transdifferentation, we suspect that sub-
unit-interdependent BRCA1 complex formation and ICL repair STAR+METHODS
contribute to efficient MEC differentiation maintenance.
The studies reported here also revealed that the nature of the Detailed methods are provided in the online version of this paper
breakdown in MEC differentiation differed when cells were and include the following:
exposed to ICL- versus DSB-inducing compounds. Defective
DSB repair led to a luminal-to-basal change, while ICL damage d KEY RESOURCES TABLE
contributed to a luminal-to-b&m change. Therefore, the quality d LEAD CONTACT AND MATERIALS AVAILABILITY
of DNA damage affected the nature of its biological outcome. d EXPERIMENTAL MODEL AND SUBJECT DETAILS
Better still, the mammary cancers that emerged from BRCA1- B Cell lines and Culture conditions
deficient luminal MECs were preceded by both advancing DNA B Mouse models
damage and aberrant luminal transdifferentiation. These findings d METHOD DETAILS
imply that BRCA1 elimination-dependent premalignant DNA B Immunoblotting and Immunoprecipitation
damage and its associated luminal-to-b&m phenotypic changes B GST –BRCA1 Fragment Binding experiments
represent vital contributions to BRCA1 mammary tumorigenesis. B Mammary gland cell preparation and flow cytometry
NUMB and HES1, both BRCA1 partners, contribute to ICL B Mammary organoid culture and manipulation
repair-driven MEC differentiation and communicate with one B Immunofluorescence staining
another (Flores et al., 2014; Buckley et al., 2013). HES1 also posi- B Confocal microscope analysis
tively influences Fanconi core complex ICL repair function B Soft Agar Assay
(Tremblay et al., 2008). Therefore, a reasonable speculation is B Clonogenic and Cell Growth Assays
that they co-participate in BRCA1 BC suppression. B CRISPR/Cas9 lentiviral vectors
In both p53/BRCA1-deficient tumors that were analyzed, B Plasmid Construction
scRNA-seq data showed that most tumor cells expressed LP/al- B siRNA and shRNA
veolar luminal markers, consistent with them having an LP origin B Comet Assay
(Lim et al., 2009; Molyneux et al., 2010; Proia et al., 2011). Impor- B Anaphase bridges
tantly, a subset of tumor cells (e.g., the B/M and EMT clusters in B Microarray expression profiling and analysis
tumor 1, the EMT cluster in tumor 2; Figure S6A) expressed both B RNA isolation and Bulk RNA sequencing
LP/alveolar luminal markers and basal and/or mesenchymal B Single-cell RNA sequencing (scRNaseq)
genes. The premalignant single-cell data and that derived B scRNaseq data preprocessing
from both tumors reflect the operation of an overarching luminal B Quantitative RT–PCR
to mesenchymal transdifferentiation process. It involved an B Statistics
enlarging LP population and/or a rare basal-like population that d DATA AND CODE AVAILABILITY
then underwent mesenchymal transdifferentiation (Figures
S7L–S7N). SUPPLEMENTAL INFORMATION

The subsequent development of BRCA1-deficient mammary


Supplemental Information can be found online at https://doi.org/10.1016/j.
tumor cells can be explained by at least three models (Figures cell.2019.06.002.
S7L–S7N). In one, LP cells represent the origin of each primary
tumor (Figure S7L), and basal-like, EMT, and mesenchymal tu- ACKNOWLEDGMENTS
mor cells are all derived from them. In another model, the cells
in the Basal-like cluster originated from the earlier growth of a We thank all members of the Li and Livingston groups for their many helpful
once relatively rare Ad-K8-Cre-targeted basal cell species that suggestions. We are also grateful to Dr. Joan Brugge and her group and to
Drs. Myles Brown, Kornelia Polyak, and Robert Weinberg for valuable input
subsequently underwent EMT; alternatively, LP cells underwent
along the way. We also thank Dr. Steve McCarroll’s lab for access to their
aberrant expansion, gradually acquired b&m features, and 10X Genomics and deep-well PCR machines. This work was supported by
became EMT cells. In both cases, the EMT cells may manifest grants from the National Cancer Institute (NCI) (2PO1CA80111-16; D.M.L.),
multipotent cancer stem cell activity (Figure S7M). In the third (5R01CA136512-05; D.M.L.) and from the Susan B. Komen Foundation for

148 Cell 178, 135–151, June 27, 2019


the Cure, the Breast Cancer Research Foundation, the Murray Winsten Foun- Chen, C.C., Feng, W., Lim, P.X., Kass, E.M., and Jasin, M. (2018). Homology-
dation, the BRCA Foundation, and the Gray Family Foundation (D.M.L.). This Directed Repair and the Role of BRCA1, BRCA2, and Related Proteins in
work was also supported by a Seed Grant (SG-0062-10; Z.L.) and a Cancer Genome Integrity and Cancer. Annu. Rev. Cancer Biol. 2, 313–336.
Program Pilot Grant (DP-0137-13-00; Z.L.) from the Harvard Stem Cell Insti- Dhami, G.K., Liu, H., Galka, M., Voss, C., Wei, R., Muranko, K., Kaneko, T.,
tute, by Breakthrough Awards from the Department of Defense (W81XWH- Cregan, S.P., Li, L., and Li, S.S. (2013). Dynamic methylation of Numb by
15-1-0100 and W81XWH-18-1-0037; Z.L.), and by NCI/NIH grants R21 Set8 regulates its binding to p53 and apoptosis. Mol. Cell 50, 565–576.
CA205825 (Z.L.) and R01 CA222560 (Z.L.).
Dho, S.E., French, M.B., Woods, S.A., and McGlade, C.J. (1999). Character-
ization of four mammalian numb protein isoforms. Identification of cytoplasmic
AUTHOR CONTRIBUTIONS and membrane-associated variants of the phosphotyrosine binding domain.
J. Biol. Chem. 274, 33097–33104.
H.W., Z.L., and D.M.L. conceived the project. H.W. and H.J.R. performed most Drost, J., Karthaus, W.R., Gao, D., Driehuis, E., Sawyers, C.L., Chen, Y., and
cultured cell studies, and D.X. performed most in vivo studies and organoid ex- Clevers, H. (2016). Organoid culture systems for prostate epithelial and cancer
periments with help from A.H., L.T., and Y.X.; D.X. performed the scRNA-seq tissue. Nat. Protoc. 11, 347–358.
and bioinformatic analysis with help from K.X.Z. and Q.C.; and B.L. and N.D.
Duarte, A.A., Gogola, E., Sachs, N., Barazas, M., Annunziato, S., de Ruiter,
helped construct and analyze plasmids. A.P.C. and V.V.B. performed soft
J.R., Velds, A., Blatter, S., Houthuijzen, J.M., van de Ven, M., et al. (2018).
agar assays and confocal microscope analysis. A.D. contributed to the EMT
BRCA-deficient mouse mammary tumor organoids to study cancer-drug
analysis. N.S. and H.C. generously provided an unpublished protocol that
resistance. Nat. Methods 15, 134–140.
made primary mammary organoid cultures possible before its publication
Escribano-Dı́az, C., Orthwein, A., Fradet-Turcotte, A., Xing, M., Young, J.T.,
and advised us on its optimal use. Z.L. and D.M.L. supervised all experiments.
, J., Cook, M.A., Rosebrock, A.P., Munro, M., Canny, M.D., et al.
Tkác
H.W., D.X., Z.L. and D.M.L. wrote the manuscript. All authors read and were
(2013). A cell cycle-dependent regulatory circuit composed of 53BP1-RIF1
invited to comment on the manuscript.
and BRCA1-CtIP controls DNA repair pathway choice. Mol. Cell 49, 872–883.
Flores, A.N., McDermott, N., Meunier, A., and Marignol, L. (2014). NUMB inhi-
DECLARATION OF INTERESTS bition of NOTCH signalling as a therapeutic target in prostate cancer. Nat. Rev.
Urol. 11, 499–507.
D.M.L. is a consultant to Constellation Pharma and the Novartis Institute for
Follenzi, A., Ailles, L.E., Bakovic, S., Geuna, M., and Naldini, L. (2000). Gene
Biomedical Research and a scientific advisor to NextechInvest; he sits on
transfer by lentiviral vectors is limited by nuclear translocation and rescued
external advisory boards of the Johns Hopkins (Kimmel), MIT, and Rutgers
by HIV-1 pol sequences. Nat. Genet. 25, 217–222.
Cancer Centers. K.X.Z. is an employee of Merck. None of the other authors
have any conflicts of interest to declare. Friedmann-Morvinski, D., and Verma, I.M. (2014). Dedifferentiation and re-
programming: origins of cancer stem cells. EMBO Rep. 15, 244–253.
Received: June 18, 2018 Garcia-Higuera, I., Taniguchi, T., Ganesan, S., Meyn, M.S., Timmers, C.,
Revised: March 4, 2019 Hejna, J., Grompe, M., and D’Andrea, A.D. (2001). Interaction of the Fanconi
Accepted: May 31, 2019 anemia proteins and BRCA1 in a common pathway. Mol. Cell 7, 249–262.
Published: June 27, 2019 Gerloff, D.L., Woods, N.T., Farago, A.A., and Monteiro, A.N. (2012). BRCT do-
mains: A little more than kin, and less than kind. FEBS Lett. 586, 2711–2716.
REFERENCES Giraddi, R.R., Chung, C.Y., Heinz, R.E., Balcioglu, O., Novotny, M., Trejo, C.L.,
Dravis, C., Hagos, B.M., Mehrabad, E.M., Rodewald, L.W., et al. (2018). Sin-
Baldwin, E.L., and Osheroff, N. (2005). Etoposide, topoisomerase II and can- gle-Cell Transcriptomes Distinguish Stem Cell State Changes and Lineage
cer. Curr. Med. Chem. Anticancer Agents 5, 363–372. Specification Programs in Early Mammary Gland Development. Cell Rep. 24,
Bu, P., Chen, K.Y., Chen, J.H., Wang, L., Walters, J., Shin, Y.J., Goerger, J.P., 1653–1666.
Sun, J., Witherspoon, M., Rakhilin, N., et al. (2013). A microRNA miR-34a- Gotzmann, J., Fischer, A.N., Zojer, M., Mikula, M., Proell, V., Huber, H., Jech-
regulated bimodal switch targets Notch in colon cancer stem cells. Cell linger, M., Waerner, T., Weith, A., Beug, H., and Mikulits, W. (2006). A crucial
Stem Cell 12, 602–615. function of PDGF in TGF-beta-mediated cancer progression of hepatocytes.
Buckley, N.E., Nic An tSaoir, C.B., Blayney, J.K., Oram, L.C., Crawford, N.T., Oncogene 25, 3170–3185.
D’Costa, Z.C., Quinn, J.E., Kennedy, R.D., Harkin, D.P., and Mullan, P.B. Guo, M., Jan, L.Y., and Jan, Y.N. (1996). Control of daughter cell fates during
(2013). BRCA1 is a key regulator of breast differentiation through activation asymmetric division: interaction of Numb and Notch. Neuron 17, 27–41.
of Notch signalling with implications for anti-endocrine treatment of breast Houghtaling, S., Newell, A., Akkari, Y., Taniguchi, T., Olson, S., and Grompe,
cancers. Nucleic Acids Res. 41, 8601–8614. M. (2005). Fancd2 functions in a double strand break repair pathway that is
Bunting, S.F., Callén, E., Wong, N., Chen, H.T., Polato, F., Gunn, A., Bothmer, distinct from non-homologous end joining. Hum. Mol. Genet. 14, 3027–3033.
A., Feldhahn, N., Fernandez-Capetillo, O., Cao, L., et al. (2010). 53BP1 inhibits Huen, M.S., Sy, S.M., and Chen, J. (2010). BRCA1 and its toolbox for the main-
homologous recombination in Brca1-deficient cells by blocking resection of tenance of genome integrity. Nat. Rev. Mol. Cell Biol. 11, 138–148.
DNA breaks. Cell 141, 243–254. Jardé, T., Lloyd-Lewis, B., Thomas, M., Kendrick, H., Melchor, L., Bougaret,
Butler, A., Hoffman, P., Smibert, P., Papalexi, E., and Satija, R. (2018). Inte- L., Watson, P.D., Ewan, K., Smalley, M.J., and Dale, T.C. (2016). Wnt and Neu-
grating single-cell transcriptomic data across different conditions, technolo- regulin1/ErbB signalling extends 3D culture of hormone responsive mammary
gies, and species. Nat. Biotechnol. 36, 411–420. organoids. Nat. Commun. 7, 13207.
Chakrabarti, R., Hwang, J., Andres Blanco, M., Wei, Y., Lukac isin, M., Jiang, X., Xing, H., Kim, T.M., Jung, Y., Huang, W., Yang, H.W., Song, S., Park,
Romano, R.A., Smalley, K., Liu, S., Yang, Q., Ibrahim, T., et al. (2012). Elf5 in- P.J., Carroll, R.S., and Johnson, M.D. (2012). Numb regulates glioma stem cell
hibits the epithelial-mesenchymal transition in mammary gland development fate and growth by altering epidermal growth factor receptor and Skp1-Cullin-
and breast cancer metastasis by transcriptionally repressing Snail2. Nat. F-box ubiquitin ligase activity. Stem Cells 30, 1313–1326.
Cell Biol. 14, 1212–1222. Kim, H., and D’Andrea, A.D. (2012). Regulation of DNA cross-link repair by the
Chen, J., Silver, D.P., Walpita, D., Cantor, S.B., Gazdar, A.F., Tomlinson, G., Fanconi anemia/BRCA pathway. Genes Dev. 26, 1393–1408.
Couch, F.J., Weber, B.L., Ashley, T., Livingston, D.M., and Scully, R. (1998). Kim, H., Huang, J., and Chen, J. (2007). CCDC98 is a BRCA1-BRCT domain-
Stable interaction between the products of the BRCA1 and BRCA2 tumor sup- binding protein involved in the DNA damage response. Nat. Struct. Mol. Biol.
pressor genes in mitotic and meiotic cells. Mol. Cell 2, 317–328. 14, 710–715.

Cell 178, 135–151, June 27, 2019 149


Lim, E., Vaillant, F., Wu, D., Forrest, N.C., Pal, B., Hart, A.H., Asselin-Labat, Schwank, G., Andersson-Rolf, A., Koo, B.K., Sasaki, N., and Clevers, H.
M.L., Gyorki, D.E., Ward, T., Partanen, A., et al.; kConFab (2009). Aberrant (2013). Generation of BAC transgenic epithelial organoids. PLoS ONE 8,
luminal progenitors as the candidate target population for basal tumor devel- e76871.
opment in BRCA1 mutation carriers. Nat. Med. 15, 907–913. Scully, R., Chen, J., Ochs, R.L., Keegan, K., Hoekstra, M., Feunteun, J.,
Lim, E., Wu, D., Pal, B., Bouras, T., Asselin-Labat, M.L., Vaillant, F., Yagita, H., and Livingston, D.M. (1997). Dynamic changes of BRCA1 subnuclear loca-
Lindeman, G.J., Smyth, G.K., and Visvader, J.E. (2010). Transcriptome ana- tion and phosphorylation state are initiated by DNA damage. Cell 90,
lyses of mouse and human mammary cell subpopulations reveal multiple 425–435.
conserved genes and pathways. Breast Cancer Res. 12, R21. Shalem, O., Sanjana, N.E., Hartenian, E., Shi, X., Scott, D.A., Mikkelson, T.,
Liu, S., Ginestier, C., Charafe-Jauffret, E., Foco, H., Kleer, C.G., Merajver, S.D., Heckl, D., Ebert, B.L., Root, D.E., Doench, J.G., and Zhang, F. (2014).
Dontu, G., and Wicha, M.S. (2008). BRCA1 regulates human mammary stem/ Genome-scale CRISPR-Cas9 knockout screening in human cells. Science
progenitor cell fate. Proc. Natl. Acad. Sci. USA 105, 1680–1685. 343, 84–87.
Lukas, C., Savic, V., Bekker-Jensen, S., Doil, C., Neumann, B., Pedersen, Silver, D.P., and Livingston, D.M. (2012). Mechanisms of BRCA1 tumor sup-
R.S., Grøfte, M., Chan, K.L., Hickson, I.D., Bartek, J., and Lukas, J. (2011). pression. Cancer Discov. 2, 679–684.
53BP1 nuclear bodies form around DNA lesions generated by mitotic Subramanian, A., Tamayo, P., Mootha, V.K., Mukherjee, S., Ebert, B.L., Gil-
transmission of chromosomes under replication stress. Nat. Cell Biol. 13, lette, M.A., Paulovich, A., Pomeroy, S.L., Golub, T.R., Lander, E.S., and Me-
243–253. sirov, J.P. (2005). Gene set enrichment analysis: a knowledge-based approach
Madaras, L., Balint, N., Gyorffy, B., Tokes, A.M., Barshack, I., Yosepovich, A., for interpreting genome-wide expression profiles. Proc. Natl. Acad. Sci. USA
Friedman, E., Paluch-Shimon, S., Zippel, D., Baghy, K., et al. (2016). BRCA 102, 15545–15550.
Mutation-Related and Claudin-Low Breast Cancer: Blood Relatives or Step- Tao, L., van Bragt, M.P., Laudadio, E., and Li, Z. (2014). Lineage tracing of
sisters. Pathobiology 83, 1–12. mammary epithelial cells using cell-type-specific cre-expressing adenovi-
Manke, I.A., Lowery, D.M., Nguyen, A., and Yaffe, M.B. (2003). BRCT repeats ruses. Stem Cell Reports 2, 770–779.
as phosphopeptide-binding modules involved in protein targeting. Science Tao, L., Xiang, D., Xie, Y., Bronson, R.T., and Li, Z. (2017). Induced p53 loss in
302, 636–639. mouse luminal cells causes clonal expansion and development of mammary
McCarthy, A., Savage, K., Gabriel, A., Naceur, C., Reis-Filho, J.S., and Ash- tumours. Nat. Commun. 8, 14431.
worth, A. (2007). A mouse model of basal-like breast carcinoma with meta- Tiwari, N., Tiwari, V.K., Waldmeier, L., Balwierz, P.J., Arnold, P., Pachkov, M.,
plastic elements. J. Pathol. 211, 389–398. Meyer-Schaller, N., Schübeler, D., van Nimwegen, E., and Christofori, G.
Molyneux, G., Geyer, F.C., Magnay, F.A., McCarthy, A., Kendrick, H., Natrajan, (2013). Sox4 is a master regulator of epithelial-mesenchymal transition by con-
R., Mackay, A., Grigoriadis, A., Tutt, A., Ashworth, A., et al. (2010). BRCA1 trolling Ezh2 expression and epigenetic reprogramming. Cancer Cell 23,
basal-like breast cancers originate from luminal epithelial progenitors and 768–783.
not from basal stem cells. Cell Stem Cell 7, 403–417. Tosoni, D., Zecchini, S., Coazzoli, M., Colaluca, I., Mazzarol, G., Rubio, A.,
Murina, O., von Aesch, C., Karakus, U., Ferretti, L.P., Bolck, H.A., Hänggi, K., Caccia, M., Villa, E., Zilian, O., Di Fiore, P.P., and Pece, S. (2015). The
and Sartori, A.A. (2014). FANCD2 and CtIP cooperate to repair DNA interstrand Numb/p53 circuitry couples replicative self-renewal and tumor suppression
crosslinks. Cell Rep. 7, 1030–1038. in mammary epithelial cells. J. Cell Biol. 211, 845–862.

Nakatani, Y., and Ogryzko, V. (2003). Immunoaffinity purification of mamma- Tremblay, C.S., Huang, F.F., Habi, O., Huard, C.C., Godin, C., Lévesque, G.,
lian protein complexes. Methods Enzymol. 370, 430–444. and Carreau, M. (2008). HES1 is a novel interactor of the Fanconi anemia
core complex. Blood 112, 2062–2070.
Nakshatri, H., Anjanappa, M., and Bhat-Nakshatri, P. (2015). Ethnicity-Depen-
dent and -Independent Heterogeneity in Healthy Normal Breast Hierarchy Im- Uemura, T., Shepherd, S., Ackerman, L., Jan, L.Y., and Jan, Y.N. (1989). numb,
pacts Tumor Characterization. Sci. Rep. 5, 13526. a gene required in determination of cell fate during sensory organ formation in
Drosophila embryos. Cell 58, 349–360.
Orthwein, A., Noordermeer, S.M., Wilson, M.D., Landry, S., Enchev, R.I.,
Sherker, A., Munro, M., Pinder, J., Salsman, J., Dellaire, G., et al. (2015). A Unno, J., Itaya, A., Taoka, M., Sato, K., Tomida, J., Sakai, W., Sugasawa, K.,
mechanism for the suppression of homologous recombination in G1 cells. Na- Ishiai, M., Ikura, T., Isobe, T., et al. (2014). FANCD2 binds CtIP and regulates
ture 528, 422–426. DNA-end resection during DNA interstrand crosslink repair. Cell Rep. 7,
1039–1047.
Park, S.Y., Lee, H.E., Li, H., Shipitsin, M., Gelman, R., and Polyak, K. (2010).
Heterogeneity for stem cell-related markers according to tumor subtype and Van Keymeulen, A., Lee, M.Y., Ousset, M., Brohée, S., Rorive, S., Giraddi,
histologic stage in breast cancer. Clin. Cancer Res. 16, 876–887. R.R., Wuidart, A., Bouvencourt, G., Dubois, C., Salmon, I., et al. (2015). Reac-
tivation of multipotency by oncogenic PIK3CA induces breast tumour hetero-
Pfefferle, A.D., Herschkowitz, J.I., Usary, J., Harrell, J.C., Spike, B.T., Adams, geneity. Nature 525, 119–123.
J.R., Torres-Arzayus, M.I., Brown, M., Egan, S.E., Wahl, G.M., et al. (2013).
Transcriptomic classification of genetically engineered mouse models of Verdi, J.M., Bashirullah, A., Goldhawk, D.E., Kubu, C.J., Jamali, M., Meakin,
breast cancer identifies human subtype counterparts. Genome Biol. 14, R125. S.O., and Lipshitz, H.D. (1999). Distinct human NUMB isoforms regulate differ-
entiation vs. proliferation in the neuronal lineage. Proc. Natl. Acad. Sci. USA 96,
Proia, T.A., Keller, P.J., Gupta, P.B., Klebba, I., Jones, A.D., Sedic, M., Gil- 10472–10476.
more, H., Tung, N., Naber, S.P., Schnitt, S., et al. (2011). Genetic predisposi-
tion directs breast cancer phenotype by dictating progenitor cell fate. Cell Visvader, J.E. (2011). Cells of origin in cancer. Nature 469, 314–322.
Stem Cell 8, 149–163. Visvader, J.E., and Stingl, J. (2014). Mammary stem cells and the differentia-
Sachs, N., de Ligt, J., Kopper, O., Gogola, E., Bounova, G., Weeber, F., tion hierarchy: current status and perspectives. Genes Dev. 28, 1143–1158.
Balgobind, A.V., Wind, K., Gracanin, A., Begthel, H., et al. (2018). A Living Wang, B., Matsuoka, S., Ballif, B.A., Zhang, D., Smogorzewska, A., Gygi, S.P.,
Biobank of Breast Cancer Organoids Captures Disease Heterogeneity. Cell and Elledge, S.J. (2007). Abraxas and RAP80 form a BRCA1 protein complex
172, 373–386. required for the DNA damage response. Science 316, 1194–1198.
Sau, A., Lau, R., Cabrita, M.A., Nolan, E., Crooks, P.A., Visvader, J.E., and Wang, H., Bierie, B., Li, A.G., Pathania, S., Toomire, K., Dimitrov, S.D., Liu, B.,
Pratt, M.A. (2016). Persistent Activation of NF-kB in BRCA1-Deficient Mam- Gelman, R., Giobbie-Hurder, A., Feunteun, J., et al. (2016). BRCA1/FANCD2/
mary Progenitors Drives Aberrant Proliferation and Accumulation of DNA Dam- BRG1-Driven DNA Repair Stabilizes the Differentiation State of Human Mam-
age. Cell Stem Cell 19, 52–65. mary Epithelial Cells. Mol. Cell 63, 277–292.

150 Cell 178, 135–151, June 27, 2019


Wu, Q., Jubb, H., and Blundell, T.L. (2015). Phosphopeptide interactions with binding partner of BRCA1 C-terminal domains. Mol. Cell. Biol. 24,
BRCA1 BRCT domains: More than just a motif. Prog. Biophys. Mol. Biol. 117, 9478–9486.
143–148.
Yu, X., Chini, C.C., He, M., Mer, G., and Chen, J. (2003). The BRCT domain is a
Xia, B., Sheng, Q., Nakanishi, K., Ohashi, A., Wu, J., Christ, N., Liu, X., Jasin, phospho-protein binding domain. Science 302, 639–642.
M., Couch, F.J., and Livingston, D.M. (2006). Control of BRCA2 cellular and
clinical functions by a nuclear partner, PALB2. Mol. Cell 22, 719–729. Yu, X., Fu, S., Lai, M., Baer, R., and Chen, J. (2006). BRCA1 ubiquiti-
Xu, X., Wagner, K.U., Larson, D., Weaver, Z., Li, C., Ried, T., Hennighausen, L., nates its phosphorylation-dependent binding partner CtIP. Genes Dev.
Wynshaw-Boris, A., and Deng, C.X. (1999). Conditional mutation of Brca1 in 20, 1721–1726.
mammary epithelial cells results in blunted ductal morphogenesis and tumour Yu, M., Smolen, G.A., Zhang, J., Wittner, B., Schott, B.J., Brachtel, E., Ram-
formation. Nat. Genet. 22, 37–43. aswamy, S., Maheswaran, S., and Haber, D.A. (2009). A developmentally regu-
Yu, X., and Chen, J. (2004). DNA damage-induced cell cycle lated inducer of EMT, LBX1, contributes to breast cancer progression. Genes
checkpoint control requires CtIP, a phosphorylation-dependent Dev. 23, 1737–1742.

Cell 178, 135–151, June 27, 2019 151


STAR+METHODS

KEY RESOURCES TABLE

REAGENT or RESOURCE SOURCE IDENTIFIER


Antibodies
BRCA1 (D9) Santa Cruz Cat#SC-6954; RRID: AB_626761
BRCA1 Millipore Cat#07-434; RRID: AB_2275035
NUMB Cell Signaling Cat#2761; RRID: AB_2267391
NUMB Novus Biologicals Cat#NB500-178; RRID: AB_922130
NUMB Bethyl labs Cat#A301-715A; RRID: AB_1211245
NUMB Bethyl labs Cat#A301-714A; RRID: AB_1211242
53BP1 Fisher Scientific Cat#BDB612522; RRID: AB_2206766
53BP1 Santa Cruz Cat#SC-22760; RRID: AB_2256326
Cyclin A Fisher Scientific Cat#BDB611268; RRID: AB_398796
HA Covance Cat#MMS-101P; RRID: AB_2314672
Flag Sigma Aldrich Cat#F1804(M2); RRID: AB_262044
Myc Millipore Cat#05-419; RRID: AB_309725
Keratin 5 (K5) Covance Cat#PRB-160P: RRID: AB_291581
Keratin 14 (K14) Covance Cat#PRB155P; RRID: AB_292096
Keratin 14 (K14) Covance Cat#SIG-3476; RRID: AB_107180
Keratin 8 (K8) Covance Cat#MMS-162P; RRID: AB-291334
GFP Abcam Cat#ab290; RRID: AB_303395
GFP Abcam Cat#ab6673; RRID: AB_305643
b-Actin Sigma Aldrich Cat#A5441; RRID: AB_476744
Zeb1 Santa Cruz Cat#SC-25388; RRID: AB_2217979
E-cadherin BD-Transductions Cat#610182; RRID: AB_397581
Vimentin (VIM) Santa Cruz Cat#SC-5565; RRID: AB_793999
HES1 Cell Signaling Cat#D6P2U; RRID: AB_2728766
HES1 Santa Cruz Cat#SC-166410; RRID: AB_2117960
DNp63 Santa Cruz Cat#SC-8431; RRID: AB_628091
FANCD2 (polyclonal antibody, IF) Novus Cat#NB100-182; RRID: AB_10002867
FANCD2 Bethyl labs Cat#A302-174A; RRID: AB_1659803
FANCD2 (monoclonal antibody, WB) Santa Cruz Cat#sc20022; RRID: AB_2278211
CtIP Bethyl labs Cat#A300-488A; RRID: AB_2175262
BRG1 Bethyl labs Cat#A300-813A; RRID: AB_2191850
Tubulin Sigma Aldrich Cat#T5168: RRID: AB_477579
Histone 3 Cell Signaling Cat#D1H2; RRID: AB_10544537
PALB2 Bethyl labs Cat#A301-246A; RRID: AB_890607
BRG1 Santa Cruz Cat#SC-17796; RRID: AB_626726
BRCA1 (anti-mouse) homemade NA
CD24-FITC BD Cat#555427; RRID: AB_395821
CD44-PE BD Cat#555479; RRID: AB_395871
CD24-PE eBiosciences Cat#12-0242-83: RRID: AB_465603
CD29-APC eBiosciences Cat#12-0291-82; RRID: AB_763478
biotinylated CD31 eBiosciences Cat#13-0311-85; RRID: AB_466421
biotinylated CD45 eBiosciences Cat#17-0495-82: RRID: AB_2016694
biotinylated TER119 eBiosciences Cat#13-5921-85: RRID: AB_466798
(Continued on next page)

e1 Cell 178, 135–151.e1–e8, June 27, 2019


Continued
REAGENT or RESOURCE SOURCE IDENTIFIER
Bacterial and Virus Strains
Max efficiency DH5a competent cells Life Technologies Cat#18258012
Adenovirus K8-Cre (Ad-K8-Cre) University of Iowa NA
Chemicals, Peptides, and Recombinant Proteins
B27 Thermo Fisher Cat#17504044
A83-01 Tocris Bioscience Cat#2939
EGF PeproTech Cat#AF-100-15
Nrg1 R&D Cat#396-HB
Y-27632 (ROCK inhibitor) Sigma Aldrich Cat#Y0503
Cisplatin Sigma Aldrich Cat#P4394-100mg
Etoposide Sigma Aldrich Cat#E1383-100mg
Noggin PeproTech Cat#120-10C
R-spondin1 Peprotech Cat#120-38
TrypLE Thermo Fisher 12604-013
Critical Commercial Assays
CometAssay kit TREVIGEN Cat#4250-050-K
QuickChange II Site-Directed Mutagenesis Kit Agilent Cat#200523
Deposited Data
Gene expression data GEO database GSE114787, GSE126761,
GSE130453
Experimental Models: Cell Lines
HMEC Yu et al., 2009 NA
MCF10A ATCC Cat#CRL-10317
MCF7 ATCC Cat#HTB-22
293T ATCC Cat#CRL-3216
U2OS ATCC Cat#HTB-96
MDA-MB231 ATCC Cat#HTB-26
Experimental Models: Organisms/Strains
B6.129P2-Trp53tm1Brn/J (Trp53L) The Jackson Laboratory Stock No#008462
Brca1tm1Aash/J (Brca1L) The Jackson Laboratory Stock No#017835
B6.129X1-Gt(ROSA)26Sortm1(EYFP)Cos/J (R26Y) The Jackson Laboratory Stock No#006148
Oligonucleotides
gNumb-1: CGTGGCCGAGGTACTTAACT This paper NA
gNumb-2: GATGAAGAAGGAGTCCGCAC This paper NA
gBrca1-1: GGCGTCGATCATCCAGAGCG This paper NA
gBrca1-2: GCTACCGGAACCGTGTCAGA This paper NA
gHes1-1: GTCCGGTGTCGTGTTGACAC This paper NA
gHes1-2: GAGGCCGTCTTTGGTTTGTC This paper NA
gPlant: GGTTCGTACGTACACTGTTC This paper NA
gROSA26: GACTCCAGTCTTTCTAGAAGA This paper NA
siBRCA1:50 UAUAAGACCUCUGGCAUGAAUUU30 This paper NA
Si53BP1: GAGAGCAGAUGAUCCUUUA This paper NA
siHES1: 50 GAGAAAAGACGAAGAGCAAUU30 This paper NA
siNUMB(both):50 CAGACUUUGUCUCCUGAUUUU30 This paper NA
siNumb (long isoform, exon 9, PRR region): Dharmacon NA
50 UAGCAAUGCCUGUGCGUGAUU30
siNumb (short isoform specific): GAAAGCUACUGGAAAGAAAUU Dharmacon NA
(Continued on next page)

Cell 178, 135–151.e1–e8, June 27, 2019 e2


Continued
REAGENT or RESOURCE SOURCE IDENTIFIER
shNUMB-A: CCGGGCATCACCTTTCCAAGGGAATCTCGAGATTCCC Sigma-Aldrich Cat#TRCN0000007227
TTGGAAAGGTGATGCTTTTT
shNUMB-B: CCGGGCAGCTTTCAATGGTGTAGATCTCGAGATCT Sigma-Aldrich Cat#TRCN0000007225
ACACCATTGAAAGCTGCTTTTT
shHES1: CCGGTGACTGACCATGCACTATATTCTCGAGAATATAGT Sigma-Aldrich Cat#TRCN0000229656
GCATGGTCAGTCATTTTTG
Gapdh: forward: 50 -GGTGAAGGTCGGTGTGAACG-30 ; reverse: This paper NA
50 -CTCGCTCCTGGAAGATGGTG-30
Trp53: forward: 50 - GCGTAAACGCTTCGAGATGTT 30 ; reverse: This paper NA
50 - TTTTTATGGCGGGAAGTAGACTG 30
Brca1: forward: 50 -CTGCCGTCCAAATTCAAGAAGT-30 ; reverse: This paper NA
50 -CTTGTGCTTCCCTGTAGGCT-30
Krt5: forward:50 -TGGCGATGACCTTCGAAACA-30 ; reverse: This paper NA
50 -GGTTGGCACACTGCTTCTTG-30
Krt14: forward: 50 -CGGACCAAGTTTGAGACGGA-30 ; reverse: This paper NA
50 -GCCACCTCCTCGTGGTTC-30
Trp63 (DNp63): forward:50 -GGAAAACAATGCCCAGACTC-30 ; This paper NA
reverse: 50 -GTGGAATACGTCCAGGTGGC-30
Vim: forward:50 -CGGCTGCGAGAGAAATTGC 30 ; reverse: This paper NA
50 - CCACTTTCCGTTCAAGGTCAAG 30
Zeb1: forward:50 -GCTGGCAAGACAACGTGAAAG-30 ; reverse: This paper NA
50 - GCCTCAGGATAAATGACGGC 30
Tcf4: forward:50 -TGAGGGGGAAAATGAGGTGC-30 ; reverse: This paper NA
50 -GAGGCGAAAACATCGCACTG-30
Hes1: forward:50 -AGGCGGACATTCTGGAAATG-30 ; reverse: This paper NA
50 - CGGTACTTCCCCAGCACACTT 30
hNumb (total)-forward:50 -CCCCAGACAGGAACTTTGATAG-30 This paper NA
reverse:50 -GCTCTAAACAGGCTGCAAAAG-30
hNumb (long, Exon9) This paper NA
forward: 50 -CTCTAGCACCCGTAGCAATG-30
reverse: 50 -GGCTGCAATTTCCTTGTTAGC-30
hNumb (short, at junction of exon 8+exon10) This paper NA
forward: 50 -CTTCCAAGGGACCGAGTG-30
reverse: 50 -AGGGAGTACGTCTATGACCG-30
S361A forward: This paper NA
50 -CAGTGGTGGCACCACAAGCTCCTACCTTCCAAG-30 reverse:
50 -CTTGGAAGGTAGGAGCTTGTGGTGCCACCACTG-30
S634A forward: This paper NA
50 -CAGCGTACTAATCCCGCCCCTACCAACCCTTTC-30 reverse:
50 -GAAAGGGTTGGTAGG GGCGGGATTAGTACGCTG-30
Recombinant DNA
lentiCRISPR-v2 Addgene Cat#52961
PHAGE-CMV-dsRed-UBC-GFP-NUMB Bu et al., 2013 NA
Numb4-GFP Jiang et al., 2012 NA
POZ-FH-N Nakatani and Ogryzko, 2003 NA
Software and Algorithms
ImageJ NIH NA
Other
glutathione-Sepharose beads GE healthcare Cat#17075601
RNeasy Micro Kit QIAGEN Cat# 74004
Chromium Single Cell 30 Library and Gel Bead Kit v2, 4 rxns 10x Genomics Cat# 120267
Chromium Single Cell A Chip Kit, 16 rxns 10x Genomics Cat# 1000009
Chromium i7 Multiplex Kit, 96 rxns 10x Genomics Cat# 120262

e3 Cell 178, 135–151.e1–e8, June 27, 2019


LEAD CONTACT AND MATERIALS AVAILABILITY

Further information and requests for resources and reagents should be directed to and will be fulfilled by the Lead Contact, David M.
Livingston (david_livingston@dfci.harvard.edu).

EXPERIMENTAL MODEL AND SUBJECT DETAILS

Cell lines and Culture conditions


293T, MCF7, MDA-MB231and U2OS cells were grown in 10% FBS DMEM medium at 37 C in a 10% CO2-containing atmosphere.
Telomerase-immortalized human mammary epithelial cells (MECs) including CD44low and CD44high MECs and MCF10A cells were
maintained in MEGM medium (Lonza). The source of cell lines is listed in the key resource tables. All cells were cultivated in a humid-
ified incubator. All cell lines and primary cells are from female origin.

Mouse models
Trp53L (B6.129P2-Trp53tm1Brn/J, Stock No: 008462), Brca1L (STOCK Brca1tm1Aash/J, Stock No: 017835), R26Y (B6.129X1-Gt(ROSA)
26Sortm1(EYFP)Cos/J, Stock No: 006148) mice were obtained from The Jackson Laboratory. To target luminal MECs in Trp53L/L;R26Y,
Trp53L/L;Brca1L/L;R26Y, or R26Y-only adult female mice (2-3 months of age), animals were anaesthetized; and Ad-K8-Cre adeno-
virus [diluted in injection medium (DMEM supplemented with 0.1% Bromophenol blue and 0.01 M CaCl2)] was introduced into ducts
of the #4 inguinal mammary gland (MG) via intraductal injection (Tao et al., 2014). Female mice were then monitored weekly for any
sign of mammary tumor development. Once tumors were detected via palpation or visual inspection, animals were monitored 2–3
times per week for subsequent tumor growth. All animal experiments were approved by the Institutional Animal Care and Use Com-
mittee (IACUC) of the Brigham and Women’s Hospital.

METHOD DETAILS

Immunoblotting and Immunoprecipitation


Immunoblotting was carried out using standard methods. In brief, cells were lysed for 2 h on ice in NETN-250 lysis buffer
(150mM NaCl, 20mM Tris-HCl pH 7.5, 0.5mM EDTA, 0.5% NP40 and 10% Glycerol) with proteinase inhibitors (Roche), and the lysate
was cleared of residual cells by centrifugation. 20-30 mg of extract were electrophoresed in each lane of a 4%–12% Bis-Tris gel
(Invitrogen) using MOPS buffer. Electrophoresed proteins were transferred to nitrocellulose membranes at 350 mA for 3 h at 4 C.
Membranes were blocked in 3% non-fat milk in PBS + 0.2% Tween 20 (PBST) and incubated overnight at 4 C in primary antibody
diluted in blocking solution. After a brief wash with PBST, the membranes were incubated with Horseradish peroxidase (HRP)-con-
jugated secondary antibodies (GE) for 2 h at room temperature. The membranes were developed by ECL chemiluminescence
(Thermo fisher). Primary antibodies are listed in the key resource table.
To generate different subcellular fractions, cells were lysed for 5 min on ice in NETN-100 lysis buffer (100mM NaCl) and centrifuged
at 8,000rpm for 5 min. The resulting pellets were suspended in the NETN 400 (400 mM NaCl) for 1 h on ice and centrifuged at 13,200
for 10 min. The insoluble chromatin pellet was then suspended in 0.2N HCL for 15 min on ice and neutralized with an identical volume
of 1M Tris-HCl (pH = 8) and labeled p400. After clearance by centrifugation, the resulting fractions (S100/S400/P400) were subjected
to immunoblotting assays.
Immunoprecipitation was performed with NETN 250-based (250mM NaCl) soluble extracts, unless otherwise specified. To probe
the BRCA1 and NUMB interaction, 293T cells were transiently transfected with HA-BRCA1 or myc-BRCA1 or Flag-NUMB plasmids
using Lipofectamine 2000 (Invitrogen). 72 h after transfection, cells were collected and lysed with NETN 250. Cellular extracts were
then incubated with the indicated antibodies and protein A-coupled Sepharose beads (GE) at 4 C overnight with continuous rocking.
The resulting immunobeads were washed 4 times in NETN 100 buffer and suspended in SDS-Laemmli sample buffer (BIO-RAD) and
analyzed by immunoblotting as described above.
To analyze endogenous BRCA1/NUMB interactions, 239T or MCF7 cell extracts were incubated with BRCA1 antibody (D9) or
NUMB antibody (Bethyl lab) and the resulting complex were analyzed by immunoblotting.
To analyze the subunit interdependence of the BRCA1/HES1/NUMB/FANCD2/CTIP complex formation, 293T cells were trans-
fected with both myc-BRCA1 and the indicated siRNA species. Transfected cells were collected after 72 h, extracted, and the ex-
tracts incubated with myc antibody (9E10) at 4 C overnight. myc-BRCA1 associated protein complexes were then analyzed by
immunoblotting.
The antibodies used in these experiments were listed in the key resource tables.

GST –BRCA1 Fragment Binding experiments


GST-BRCA1 fragments (#1-#6) have been described (Chen et al., 1998; Scully et al., 1997). These GST-BRCA1 fusion proteins
were synthesized in E.Coli and purified on glutathione-Sepharose beads (GE healthcare 17075601). Cell extract from 293T cells

Cell 178, 135–151.e1–e8, June 27, 2019 e4


was incubated with bead-bound GST fusion proteins or GST beads only. The bound complexes were then analyzed by SDS gel elec-
trophoresis and immunoblotting, as described above. Coomassie blue staining that revealed the relative abundance of each GST
fusion protein is indicated above each arrow.

Mammary gland cell preparation and flow cytometry


Inguinal MGs injected with Ad-K8-Cre were dissected, minced, and incubated for 1–1.5 h in digestion medium (DMEM/F12 with 2%
Penicillin/ Streptomycin, 0.1 mg ml1 Gentamicin, 0.6% Nystatin, 2 mg ml1 Collagenase A, 0.096 mg ml1 Hyaluronidase) at 37  C
with continuous shaking. After digestion, the cells/tissues were treated sequentially with 0.25% trypsin/EDTA (37  C, 2 min),
5 mg ml1 dispase with DNaseI (0.1 mg ml1, Sigma, St Louis, MO; 37  C, 5 min), and cold red blood cell (RBC) lysis buffer (on
ice, 2–3 min). Between each treatment step, cells/tissues were washed with 1 3 PBS containing 5% FCS. After treatment with
RBC lysis buffer, cells/tissues were filtered through a 40 mm cell strainer and washed with 1 3 PBS/5% FCS, to obtain a single-
cell suspension (Tao et al., 2017). The resulting MECs are depleted with the CD45, CD31 and TER119 lineage markers and only
YFP positive cells were used for analysis of CD24 and CD29. Flow cytometric (FACS) analysis and cell sorting were performed
with a FACSAria sorter (BD Biosciences). Antibodies used in FACS analysis and cell sorting were purchased from eBiosciences
(San Diego, CA) and included, CD24-PE (clone M1/69, 12-0242-83; 1:250), CD29-APC (clone eBioHMb1-1, 12-0291-82; 1:250),
and biotinylated lineage (Lin) markers: CD31 (clone 390, 13-0311-85; 1:100), CD45 (clone 30-F11, 17-0495-82; 1:100) and
TER119 (clone Ter-119, 13-5921-85; 1:100).

Mammary organoid culture and manipulation


The long-term organoid culture system used in this study was developed with minor optimization based on the original formulation
from (Drost et al., 2016) and adapted from (Jardé et al., 2016), and (Sachs et al., 2018), and (Duarte et al., 2018). Sorted YFP+ MECs
were embedded in 15-20 mL Matrigel (Corning, 356231) in a 48-well plate, allowed to solidify for 5-10 min, and then overlaid with
DMEM/F12 advance media, supplemented with HEPES (1:100, GIBCOTM, 15630-080), GlutaMAX (1:100, GIBCOTM, 35050-061),
B27 (1:50, Thermo Fisher, 17504044), 100 ng/mL A83-01 (Tocris Bioscience, 2939), 50 ng/mL EGF (PeproTech, AF-100-15),
100 ng/mL Noggin (PeproTech, 120-10C), 500 ng/mL commercial R-spondin1 (Peprotech, 120-38) or conditioned R-spondin1 me-
dium from RSPO cells (Cultrex, 3700-100-01), 100 ng/mL Nrg1 (R&D, 396-HB), 10 mM Y-27632 (ROCK inhibitor, Sigma Aldrich,
Y0503) which is only necessary when organoids are first prepared, thawed, or when organoids are dissociated to single cells. The
organoid culture medium including the supplementaries described above will be referred to as ‘‘full medium’’ for the subsequent
work. The procedure for organoid transfection was developed with modifications mainly based on the protocol described by
(Schwank et al., 2013). Briefly, following one day of culture in full medium, as described above, organoids were infected with lentivirus
for 2-3 days before selection was initiated in full medium containing puromycin (1 mg/mL) for one week. Stably infected and edited
organoid cells were expanded in full medium.
For drug treatment of organoids, cisplatin (final concentration = 0.2 mM, Sigma) and etoposide (final concentration = 0.1 mM, Sigma)
were added to relevant full organoid culture medium for 7 days. Drug-treated organoids were then digested for single cell suspen-
sions by TrypLE (Thermo Fisher Scientific, 12604-013) for 15 min in the cell culture incubator. Single organoid cells were re-plated in
a 48-well plate and cultured in full organoid medium without cisplatin or etoposide treatment for another 14 days. At this point, orga-
noids were digested and collected for RNA extraction, or single organoid cells were cultured in an 8-well chamber slide for at least
24 h followed by co-immunofluorescent staining for K5, 53BP1 and DAPI. In all these experiments, 250 ml of full organoid culture
medium was added per well, and these organoids were maintained in a 37  C humidified atmosphere under 5% CO2.

Immunofluorescence staining
Prior to embedding organoids in paraffin, organoids suspended in Matrigel were entirely embedded in Specimen Processing
HistoGel (Thermo Fisher, HG-4000-012), followed by fixation in 4% paraformaldehyde (PFA, Fisher Scientific, Hampton, NH) for
2 h and then overnight in 70% ethanol at room temperature. Immunofluorescent (IF) staining was performed on sections from
MGs, mammary tumors and organoids. The IF staining was performed as described (Tao et al., 2017). Primary antibodies used
included: anti-GFP (ab290, 1:500 or ab6673, 1:200, Abcam, Cambridge, UK), anti-Keratin 14 (K14) (PRB-155P, 1:400 or SIG-
3476, 1:200, Covance, Dedham, MA), anti-Keratin 5 (K5) (PRB-160P, 1:500, Covance), anti-Keratin 8 (K8) (MMS-162P, 1:200, Cova-
nce), anti-53BP-1 (BD 612522, 1:1000). For IF staining, the secondary antibodies used were: goat anti-mouse IgG conjugated with
AF488 (A11029, 1:250) or with AF647 (A31571, 1:250), donkey anti-goat IgG conjugated with AF488 (Ab150129, 1:250), chicken anti-
goat IgG conjugated with AF647 (A21469, 1:250), goat anti-rabbit IgG conjugated with AF488 (A11008, 1:250) or with AF594 (A11037,
1:250), and goat anti-chicken IgG conjugated with AF594 (A11042, 1:250) or with AF488 (A11039, 1:250) (all from Molecular Probes,
Eugene, OR). Slides were counterstained with DAPI (1 mg/mL).
To stain mouse BRCA1 in the CRISPR/Cas9-affected organoids, organoids stably edited with gPlant/gRosa26/gBrca1/gNumb/ or
gHes1 were first collected and resuspended as single cells and then seeded onto coverslips with overnight incubation. Cells were
then fixed with 4% paraformaldehyde for 15 min, washed with PBS 3 times and permeablized with cold 0.5% Triton X-100 for
5 min. Cells were incubated with primary antibodies for 3 h and then with secondary antibodies for 1 h, both at room
temperature. After washing with PBST (PBS containing 0.5% Tween 20), cells were then mounted with medium containing
4’,6-diamidino-2-phenylindole (DAPI, Vector lab H1500) and sealed with clear nail polish.

e5 Cell 178, 135–151.e1–e8, June 27, 2019


Confocal microscope analysis
Confocal images were captured as z stacks (0.265mm thickness) using a Nikon Ti inverted microscope equipped with a CSU-X1 spin-
ning disk head (Yokogawa), 100x oil objective (NA1.4; Nikon), and an iXon DU-897 EM-CCD camera (Andor). Confocal images were
captured using Andor iQ2 software. HME cells for confocal imaging were cultured asynchronously on 1.5mm cover glasses at 37 C/
5% CO2, fixed with 4% paraformaldehyde and permeabilized with PBS+0.5% Triton X-100. Cells were stained with mouse mono-
clonal a-BRCA1 antibody at 1:500 dilution in 1% BSA/PBS (sc-6954, Santa Cruz Biotechnology) and rabbit polyclonal a-NUMB anti-
body at 1:50 dilution in 1% BSA/PBS (A301-715A, Bethyl Laboratories) Secondary antibodies were FITC AffiniPure goat anti-rabbit
(1:200 in PBS; Jackson ImmunoResearch Laboratories, 111-095-003) and Rhodamine Red-X AffiniPure goat anti-mouse (1:400 in
PBS; Jackson ImmunoReseach Laboratories, 115-295-146). Image analysis was performed using the Fiji processing software pack-
age of ImageJ.

Soft Agar Assay


Bottom layers were prepared in six well dishes with 0.6% noble agar (Difco 214220) in DMEM. HMECs were plated in a top layer
composed of 0.3% agar in MEGM at a density of 12,500 cells per well. Each well was fed weekly with 1ml of MEGM. After three
weeks, the media was aspirated and plates were fixed in a solution of 5% methanol and 5% acetic acid for 20-30 min, then stained
overnight with 0.005% crystal violet in 10% ethanol. Crystal violet solution was aspirated prior to imaging and counting colonies.
Counts represent the average of three wells.

Clonogenic and Cell Growth Assays


To measure cell growth, Cells were transfected with siLuc/siBRCA1/siNumb/ or siHes1 for 48 h. 300 cells/well were seeded in six-
well plastic plates and treated with indicated drugs and cultivated for 10 days. Cells were fixed and visualized with crystal violet.
To rescue NUMB depletion-associated ICL drug sensitivity, siRNA-resistant NUMB cDNA (WT or 2A) or empty vector was stably
introduced into MCF10A cells by retrovirus infection followed by puromycin-based selection. The stable MCF10A-vector and
MCF10A-NUMB (WT or 2A, both siRNA-resistant) -transduced cells were then transfected with siNUMB to deplete endogenous
NUMB proteins and plated in 96 well plates (1,000 cells/well). These cells were exposed overnight to increasing concentrations of
cisplatin. We measured the relative growth of NUMB-WT or NUMB-2A expressing MCF10A after siNUMB depletion by CellTiter-
Glo Luminescent Cell Viability Assay (Promega) following the manufacture instructions.

CRISPR/Cas9 lentiviral vectors


The lentiCRISPR-v2 plasmid was from Addgene (No. 52961, a gift from Dr, Feng Zhang). Two individual guide RNAs (gRNAs) target-
ing Numb or Brca1 or Hes1 were designed based on http://crispr.mit.edu (with a high quality score of above 90). LentiGuide vectors
constructed with gRNAs targeting the mouse Rosa26 region of mouse (gRosa26) and targeting a plant region (gPlant) were kind gifts
from Prof. Richard Mass [Division of Genetics, Brigham and Women’s Hospital (BWH)]. Sequences for all of these gRNAs are listed in
the key resource table. Cloning of gRNAs in lentiCRISPR-v2 plasmid was performed as described (Shalem et al., 2014). All vectors
were validated by Sanger sequencing. We produced concentrated lentiviral stocks, pseudotyped with the VSV-G envelope protein,
by transient transfection of their corresponding plasmids into 293T cells as previously described (Follenzi et al., 2000).

Plasmid Construction
The NUMB long isoform was amplified from the PHAGE-CMV-dsRed-UBC-GFP-NUMB plasmid provided by Pengcheng Bu
(Bu et al., 2013) and the NUMB short isoform was amplified from the Numb4-GFP plasmid provided by Dr. Hongyan Xing
(Jiang et al., 2012). The amplified PCR products were then cloned into a vector with 3XHA or 3xFlag or an HA/Flag double tag.
The NUMB S361A /S634A mutant was generated with the quickchange II site-directed mutagenesis Kit (Agilent 200523) followed
by the manual instruction with following primers. All plasmids were sequenced to confirm the mutations.

siRNA and shRNA


All siRNAs were synthesized at Dharmacon Inc, and the sequences are listed in the key resource table. CD44low MECs were plated at
50% confluence, and the siRNAs were transfected with RNA MAX(Invitrogen) following the Manual’s instructions.
To test the effect of siRNA transfection on BRCA1/NUMB/53BP1/Cyclin A staining, cells were transfected with the indicated
siRNAs for 48 h, plated on coverslips, and allowed to incubate overnight. Cells were then fixed with 4% PFA, permeabilized with
0.5% Triton, and then incubated with antibodies at room temperature.
To test the effect of overexpression of short NUMB on differentiation, CD44low MECs stably infected with empty vector or short
NUMB cDNA retrovirus were then transfected with SiLuc (Luc: luciferase) or SiNUMB. SiNUMB targeting its 30 UTR depleted both
endogenous short and long NUMB isoforms but had no effect on short NUMB cDNA.
ShRNAs were delivered by lentivirus infection. Briefly, the shRNA and packaging plasmids (VSV-G and D8.9) were cotransfected
into 293T cells using Lipfectomine 2000. Viral particles were collected at 48 and 72 h after transfection. The supernatants were used
for infection after filtration through 0.45 mM syringe filters. Infected cells were selected with puromycin (2 mg/mL) and maintained in

Cell 178, 135–151.e1–e8, June 27, 2019 e6


puromycin-containing MEGM. To produce retroviruses expressing HA/Flag-tagged NUMB, viral vectors were cotransfected with
pMD-MLV and pMD-G using Lipfectomine 2000 in 293T cells. Supernatants were collected 48 or 72 h after transfection and centri-
fuged to concentrate virus for infection.

Comet Assay
Alkaline comet assays were performed on mouse MECs engineered with CRISPR/Cas9 (gPlant/gRosa26/gNumb/ or gBrca1) using
the CometAssay kit (Trevigen, 4250-050-K) according to the manufacturer’s instructions. 100-300 cells were analyzed in each
experiment.

Anaphase bridges
Proliferating CD44high cells following HES1 depletion were plated at 60% confluence overnight, fixed with 4% paraformaldehyde for
15 min, and stained with 4’,6-diamidino-2-phenylindole (DAPI; Vector Labs) prior to microscopy.

Microarray expression profiling and analysis


Total RNAs from YFP+ MECs sorted from R26Y females 2 weeks after intraductal induction of Ad-K8-Cre or from Trp53/Brca1 null
mammary tumors were prepared using an RNeasy kit (QIAGEN, Valencia, CA). The microarray experiment was performed in Dana-
Faber Cancer Institute Core facility as described in (Tao et al., 2017). All arrays were normalized and processed in GenePattern
(https://www.broadinstitute.org/cancer/software/genepattern/). Gene Set Enrichment Analysis (GSEA) was performed using the Mo-
lecular Signatures Database v5.2 (http://software.broadinstitute.org/gsea/msigdb) and the standalone GSEA program (http://
software.broadinstitute.org/gsea/login.jsp). Gene sets for mouse or human intrinsic breast cancer subtypes were extracted from
a recent study (Pfefferle et al., 2013) by using upregulated genes (SAM fold change > 1) for each subtype, with SAM q-value of 0.
A Multiple Experiment Viewer (MeV) program (http://www.tm4.org/) was used to visualize the expression data.

RNA isolation and Bulk RNA sequencing


Single-cell suspensions of mammary glands (MGs, from Trp53L/L;Brca1L/L;R26Y or Trp53L/L;R26Y or R26Y female mice 1 month post
Ad-K8-Cre injection were obtained as described in the ‘‘Mammary gland cell preparation and flow cytometry’’ procedure. Cell preps
were stained with DAPI and DAPI-YFP+ cells were sorted in a FACS Aria II (BD Biosciences). Total RNA was extracted using the
RNeasy Micro Kit (QIAGEN, Cat# 74004). Libraries were prepared by the Dana-Farber Cancer Institute (DFCI) Molecular Biology
Cores, using the Low Input mRNA Library (Clontech SMARTer) v4, following the vendor’s protocol. Library quality was checked using
High Sensitivity D1000 reagents in an Agilent Tape Station. Libraries were pooled and sequenced in an Illumina NextSeq500 (Single-
End 75bp reads per sample). A Visualization Pipeline for RNaseq (Viper) analysis tool, developed by the Center for Functional Cancer
Epigenetics (CFCE) at DFCI, was used to generate standard outputs.

Single-cell RNA sequencing (scRNaseq)


Single-cell suspensions were generated as described in the ‘‘Mammary gland cell preparation and flow cytometry’’ procedure. YFP+
premalignant MECs or mammary tumor cells from Trp53L/L;Brca1L/L;R26Y mice post Ad-K8-Cre injection (at the indicated time
points) were sorted by FACS into Dulbecco’s PBS + 0.04% BSA and retained on ice. Barcoded scRNaseq libraries were generated
according to the manufacturer’s instructions, using a 10 3 Chromium Single Cell 30 v2 reagent kit (10 3 Genomics, Pleasanton, CA,
USA). Basically, cell suspensions were loaded onto a Chromium Single-Cell Chip along with the reverse transcription (RT) master mix
and single cell 30 gel beads, aiming for maximum of 10,000 cells per channel. Following the generation of single-cell gel bead-in-emul-
sions (GEMs), reverse transcription was performed using a C1000 Touch Thermal Cycler with a Deep Well Reaction Module (Bio-Rad
Laboratories, Hercules, CA, USA). Amplified cDNA was purified using SPRIselect beads (Beckman Coulter, Lane Cove, NSW,
Australia) and sheared to approximately 200 bp with a Covaris S2 instrument (Covaris, Woburn, MA, USA) following the manufac-
turer’s recommended parameters. Sequencing libraries were generated with unique sample indices (SI) for each sample. Libraries
were sequenced on an Illumina HiSeq-RAPID High Output Mode at cycles of 26x98 bps.

scRNaseq data preprocessing


Following sequencing, demultiplexing of cellular barcodes, mapping of reads to the mouse genome (build mm10), and production of
matrices of gene counts were performed by the MIT Genome Technology Core or BWH Single Cell Genomics Core. We next pro-
cessed the unique molecular identifier (UMI) count matrix using the Seurat R package (v3.0) (Butler et al., 2018). Cells that have either
a unique gene count over 2,500 or less than 200, and genes detected in less than three cells, were filtered out. Seurat methodology
uses principle component analysis (PCA) to visualize and explore these datasets and further summarize PCA with tSNE dimension-
ality using the default settings of the RunTSNE function. Cell clusters in the resulting two-dimensional representation were annotated
to known biological cell types using FindAllMarkers for all clusters. For tumor single cell data, tSNE two-dimensional plots were also
generated by the Loupe Cell Browser (10X Genomics) by using .cloupe files generated from the expression matrices derived from the
Cell Ranger pipeline (10X Genomics).

e7 Cell 178, 135–151.e1–e8, June 27, 2019


Quantitative RT–PCR
Organoids were dissociated with TrypLE and pelleted by centrifugation for 5 min at 1000 rpm, followed by RNA extraction with either
TRIzol (Thermo Fisher Scientific, 15596026) or the Allprep DNA/RNA mini/micro kits (QIAGEN) according to the manufacturer’s pro-
tocols. cDNA was generated with an iScript cDNA synthesis kit (Bio-RAD, 170-8891). Quantitative RT–PCR was performed using
FastStart SYBR Green Master (Roche, 04913850001, IN).

Statistics
Student’s t test (two-tailed) and Two-way ANOVA methods were used to calculate P values. Data were reported as Mean ± SEM or
Mean ± SD. Statistical methods were not used to pre-determine mouse sample sizes. No randomization or blinding was used in the
in vivo studies.

DATA AND CODE AVAILABILITY

The accession numbers for gene expression reported in this paper are GEO: GSE114787, GEO: GSE126761 and GEO: GSE130453.

Cell 178, 135–151.e1–e8, June 27, 2019 e8


Supplemental Figures

Figure S1. NUMB Is a BRCA1-Interacting Nuclear Protein, Related to Figures 1 and 2


(A) Sequence analysis of NUMB from different species emphasizing (in red type) two, putative BRCT-binding motifs SPTF and SPTXXF. Analogous motifs are also
depicted in three, other BRCA1 BRCT motif- binding proteins.
(B) A mutation in a BRCT motif (e.g., M1775R) reduced the interaction between BRCA1 and NUMB. 293T cells were transiently transfected with HA-BRCA1
(WT/M1775R/C61G), and cell extracts were immunoprecipitated with HA antibody and analyzed by immunobloting.
(C) Phosphatase exposure reduced the interaction between NUMB and BRCA1. Cell extracts from HA-NUMB (Long isoform)-expressing 293T cells were IP’d
with HA antibody and, where indicated, the IPs we exposed or not to phosphatase and subjected to immunobloting analysis.
(D-F) western blotting for NUMB and BRCA1 in cytoplasmic (S100), soluble nuclear (S400), and insoluble nuclear fractions (P400). U2OS or CD44low MECs or
MCF10A cells were fractioned at the different indicated salt concentrations in the experimental scheme (D) and analyzed by immunoblotting (E-F).
(G-H) western blotting for NUMB and BRCA1 of different cellular fractions from U2OS or MCF7 cells before and after cisplatin treatment.
Figure S2. NUMB Forms Distinct DNA Damage Foci, Related to Figure 2
(A-B) Immunofluorescent (IF) staining of BRCA1 and NUMB (A) or 53BP1 and NUMB (B) in MCF7 cells. Scale bar = 20 mm.
(C) Quantitation of 53BP1 foci in Cyclin A negative (G1/G0) CD44low MECs before and after NUMB or BRCA1 depletion.
(D) Quantitation of Cyclin A positive (S/G2 cells) and Cyclin A negative (G1/G0) cells before and after NUMB or BRCA1 depletion in CD44low MECs. Data in C and D
represent Mean ± S.D., and a Student’s t test was used to calculate statistical significance. N.S = Not Significant; *p < 0.05; ****p < 0.0001.
(E-F) Co-staining of 53BP1 and NUMB after transfection of siluc (E) or siBRCA1 (F) in CD44low MECs. Scale bar = 10 mm.
Figure S3. NUMB Sustains Differentiation and ICL Repair in MECs, Related to Figure 3
(A) Representative image of FANCD2 staining of SiLuc- or SiNUMB- transfected CD44low MECs after cisplatin treatment (1 mM, overnight).
(B) Quantitation of FANCD2 positive cells (i.e., that contain > 10 FANCD2 foci/cell) in SiLuc- or SiNUMB- transfected CD44low MECs after cisplatin treatment
(1 mM, overnight). More than 280 cells were analyzed, and the data represent Mean ± SD. A student’ t test was again used to calculate statistical significance.
****p < 0.0001.
(C) Effect of NUMB depletion on monoubiquitinaiton of FANCD2 after MMC treatment in MCF7 cells.
(D) qPCR analysis of relative expression of total NUMB, long NUMB isoform or short NUMB isoform mRNAs in CD44low MECs or BRCA1-depleted
CD44high MECs.
(E) Immunoblotting for NUMB after CD44low MECs were transfected with SiNUMB targeting both the long and short isoforms, the long isoform only, or the short
isoform only.
(legend continued on next page)
(F) Phase-contrast images from CD44low MECs stably infected with empty vector or short NUMB cDNA retrovirus after siluc or siNumb transfection. siNUMB
targeted the NUMB 30 UTR depleting both endogenous Short and Long NUMB. In contrast, it had no effect on the exogenously expressed short NUMB cDNA.
(G) CD24 and CD44 profiles of CD44low MECs stably infected with empty vector or an siRNA-resistant Short NUMB-encoding retrovirus after SilLuc or SiNUMB
transfection.
(H) Quantitation of CD44high cells derived from CD44low MECs expressing empty vector or NUMB-Short after SiLuc or SiNUMB transfection. Data represent
Mean ± S.D., and a Student’s t test was used to calculate statistical significance. **p < 0.01.
(I) Immunoblotting for NUMB/BRCA1/E-cadherin in basal breast cancer cells, MDA-MB231 and SUM149PT, and CD44low WT MECs. SUM149PT is a line derived
from a germline BRCA1 mutant patient.
Figure S4. Loss of HES1 Promotes Aberrant Differentiation and ICL Damage, Related to Figure 4
(A) Quantitation of FANCD2 positive cells (that contain > 10 foci/nucleus) in siLuc or siHes1- transfected CD44low MECs after cisplatin treatment (1 mM, over-
night). The data represent Mean ± SD. A Student’s t test was used to calculate statistical significance. ****p < 0.0001.
(B) Quantitation of HES1 mRNA expression in CD44low and BRCA-depleted CD44high cells, each labeled by its clone number. The figure depicts HES1 mRNA
expression in each of several BRCA1-depleted CD44high clones and dox-treated and untreated CD44low cells. Doxycycline (Dox) induction was performed to elicit
synthesis of a BRCA1 hairpin.
(C) Immunoblotting for HES1 in CD44low and BRCA-depleted CD44high cells. Doxycycline induction was performed to elicit synthesis of a BRCA1 hairpin.
(D) CD24 and CD44 profile of cells transfected with sh-control or shHES1 (clone-1).
(E) Phase-contrast images of FACS-sorted CD44low and CD44high cells after stable HES1 depletion.
(F) Immunoblotting for EMT markers in HES1-depleted CD44high and CD44low MECs (clone-1).
(G) Representative images of anaphase bridges in HES1-depleted CD44high cells. Scale bar = 10 mm.
(H) Representative images of soft agar assay results obtained with CD44low or BRCA1-depleted CD44high MECs.
(I) Statistical analysis of the effects on soft agar formation in clonal CD44low or CD44high cells. The data represent Mean ± SD. A Student’s t test was used to
calculate statistical significance. ***p < 0.001.
Figure S5. Induced Loss of BRCA1 and p53 Promotes Luminal Cell Origination of Basal-like Mammary Tumor Development, Related to
Figures 5 and 6
(A) Quantitation of percentages of YFP-marked MECs found in mammary glands (MGs) from virgin females with the indicated genotypes 2 weeks after intraductal
injection of Ad-K8-Cre. The data represent Mean ± SEM, and statistics were calculated using a Student’s t test: ***p % 0.005.
(B) Representative images showing H&E-stained sections of mouse cancers that developed in the cohort in Figure 5D. Scale bar = 50 mm.
(C) Co-IF staining showing expression of YFP (i.e., a lineage marker), K14 (a basal marker), and K8 (a luminal marker) in a representative mammary tumor
developed in the cohort depicted in Figure 5D. Scale bar = 50 mm.
(D) GSEA results showing comparisons of microarray data from tumors that developed in mice described in Figure 5D and from WT Krt8+ luminal MECs (cells of
origin) to gene sets representing mouse mammary tumor intrinsic subtypes. The gene sets were extracted from Pfefferle et al. In the plot, normalized enrichment
score (NES) and nominal P values (NOM p-val) are shown for each gene set. Gene sets for mouse subtypes most closely resembling human basal-like breast
cancer are highlighted in red (indicating the strongest similarities).

(legend continued on next page)


(E) No evidence of genomic amplification of a 1.15Mb amplicon on chromosome 9qA1 that encompasses multiple Mmp and Birc genes, the Hippo pathway-
related Yap1 gene, as well as another genomic amplicon spanning chromosome 6qA1-6qA2) that includes Cav2, Cav1 and Met was detected in Trp53/Brca1 null/
null tumors that developed in mice in the cohort depicted in Figure 5D. In both heatmaps, expression values of each gene (based on microarray results) were
normalized to the mean of WT luminal MEC samples.
(F) GSEA plots showing enrichment of a mesenchymal-related gene set in Trp53/Brca1 null/null MECs by comparison with Trp53 null or WT MECs, based on
RNaseq data as in Figure 6A. NES and nominal P values are shown.
(G) Representative images of H&E-stained sections of organoids generated with sorted YFP+ MECs from Trp53L/L;R26Y females or Trp53L/L;Brca1L/L;R26Y
females following intraductal injection of Ad-K8-Cre. Scale bar = 50 mm.
(H) Representative images showing organoids (left: YFP; right: phase contrast) generated with sorted YFP+ MECs from R26Y females, Trp53L/L;R26Y females,
and Trp53L/L;Brca1L/L;R26Y females, upon intraductal injection of Ad-K8-Cre. Organoids were cultured in the presence (left) or absence (right) of B27. Scale
bar = 50 mm.
(I) Quantitation of sizes (left) and organoid-forming efficiencies (i.e., number of organoids formed per 5,000 seeded cells) for organoids formed in (S5H) in the
presence (left) or absence (right) of B27. Data represent Mean ± SEM. *p % 0.05, **p % 0.01, ****p % 0.001.
(J) IF staining showing cells with K14 expression and those with ongoing DNA damage (i.e., 53BP1 positive) in Brca1/Trp53 null/null versus Trp53 null organoids
(luminal origin). Scale bar = 100 mm.
Figure S6. Loss of BRCA1 in Luminal MECs Leads to Aberrant Transdifferentiation and Accumulation of DNA Damage, Related to Figure 6
(A) Analysis of scRNaseq data based on Cell Ranger (10X Genomics). In t-SNE plots, arrows indicate in each tumor that tumor epithelial and mesenchymal cell
clusters exhibit a trend of transdifferentiation from LP-like cells (left) to EMT-like cells (middle), whose clusters are closest to mesenchymal cell clusters (right). LP:
luminal progenitor-like cells; B/M: cells with basal/mesenchymal transdifferentiation; EMT: epithelial-to-mesenchymal transition cells; M: mesenchymal cells.
Relative expression levels of select genes (black: epithelial genes; purple: LP/alveolar luminal genes; red: EMT TF/inducer genes; green: mesenchymal genes) are
shown for cells in the indicated tumor epithelial and mesenchymal clusters.
(B) Kaplan-Meier curves showing higher expression levels of SPARC, POSTN, AEBP1 and SFN correlate with worse patient outcomes [any event (AE)-free
survival, based on patients with basal-like breast cancers; for each gene, left: Hu’s SSP (Single Sample Predictor); right: Sorlie’s SSP], based on Breast Cancer
Gene-Expression Miner v4.2 (bc-GenExMiner v4.2) online tool.

(legend continued on next page)


(C) IF staining for 53BP1+ cells (red) in mammary glands from R26Y-only (WT) or Trp53L/L;R26Y control female mice one month after intraductal injection of Ad-
K8-Cre. Scale bar = 50 mm.
(D) Quantification for percentages of 53BP1+ cells in Figures 6I and (C). The percentage was assessed by quantification of 53BP1+ nuclei against DAPI+ nuclei
within the selected region of interest using Image Pro software (Rockville, USA). The number of 53BP1+ nuclei from 20 random fields in tissue sections was
counted up to 300 cells per region. 1 m or 5 m: 1 month or 5 months post Ad-K8-Cre intraductal injection, respectively. Statistics: ****p % 0.001.
(E) Representative IF staining shows that almost all Trp53/Brca1 null/null tumor epithelial cells stained positive for 53BP1 (green arrowheads), whereas non-
epithelial cells (e.g., stromal cells) were 53BP1 negative (white arrowheads). Scale bar = 50 mm.
(F) IF staining for g-H2AX + cells (red) in mammary glands from R26Y-only (WT) or Trp53L/L;R26Y control female mice performed one month after intraductal
injection of Ad-K8-Cre. IF staining for g-H2AX + cells was also performed on mammary glands of Trp53L/L;Brca1L/L;R26Y female mice at the indicated times after
intraductal injection of Ad-K8-Cre; staining was also performed on tumor cells of these animals that were detected > 8 months after Ad-K8-Cre injection.
Figure S7. Accumulation of ICL Damage Promotes a Luminal to Basal/Mesenchymal Cell Fate Change, Related to Figure 7
(A-B) Quantitation of K5 positive (A) or 53BP1 positive (B) organoids after Trp53 or Trp53/Brca1 knockout or exposure of Trp53 null mouse organoid MECs to
cisplatin or etoposide. Data represent Mean ± SEM. ****p % 0.001.
(C) IF images that depict a loss of BRCA1 foci in Trp53 null luminal organoid cells after CRISPR/Cas9-mediated editing of Brca1 (gBrca1), compared to those that
underwent editing of either Numb (gNumb) or the neutral Rosa26 genomic region (gRosa26). Scale bar = 10 mm
(D) western blots showing a loss of NUMB expression in Trp53 null luminal organoid cells after CRISPR/Cas9-mediated editing of Numb.
(E) Representative images depicting Trp53 null luminal organoids (left: phase contrast; right: YFP expressing) following CRISPR/Cas9-mediated editing of either
the neutral Rosa26 genomic region (gRosa26), Brca1 (gBrca1), or Numb (gNumb). Scale bar = 100 mm

(legend continued on next page)


(F) Representative microscopic images showing Trp53 null luminal organoid cells with comet tails upon CRISPR/Cas9-mediated editing of either Brca1 or Numb,
by comparison with cells that underwent editing of a neutral genomic region (gRosa26) or no editing (gPlant). Quantitation of the comet tails is shown in Figures 7E
and 7F. Scale bar = 10 mm.
(G) western blot showing a loss of HES1 protein expression in Trp53 null luminal organoid cells after CRISPR/Cas9-mediated editing of Hes1.
(H) Representative images depicting Trp53 null luminal organoids (left: phase contrast; right: YFP) following CRISPR/Cas9-mediated editing of either the neutral
Rosa26 genomic region (gRosa26) or Hes1 (gHes1). Scale bar = 100 mm.
(I) Quantitation of organoid size from H. Statistics: *** = p % 0.005.
(J) qRT-PCR analysis showing upregulation of selected basal (e.g., Krt5) and mesenchymal (e.g., Vim, Snai1, Zeb1) genes in Trp53 null luminal organoid cells
following CRISPR/Cas9-mediated editing of Hes1. By comparison, results in cells after editing of a neutral genomic region (Rosa26, control) are shown.
(K) Lower expression levels of NUMB and HES1 in BLBC in comparison to those of other human breast cancer subtypes, based on the Breast Cancer Gene-
Expression Miner (bc-GenExMiner) online tool. P values shown here are based on Dunnett-Tukey-Kramer’s test; NS = not significant.
(L-N) Proposed models based upon the results of scRNaseq to explain the cellular origin and progression of mammary tumors that developed in Trp53L/L;
Brca1L/L;R26Y female mice after intraductal injection of Ad-K8-Cre. See Discussion for details. LP*: refers to luminal progenitor-like premalignant cells that are in
the course of expressing basal/mesenchymal genes; LP**: refers to luminal progenitor-like tumor cells that express both basal and mesenchymal genes; B*:
refers to cells that express basal-like genes. B/M refers to cells expressing basal and mesenchymal genes. EMT are tumor cells that manifest a mesenchymal
gene expression phenotype.

You might also like