You are on page 1of 26

A RT I C L E S

Distinct E-cadherin-based complexes regulate cell


behaviour through miRNA processing or Src and
p120 catenin activity
Antonis Kourtidis1, Siu P. Ngok1,5, Pamela Pulimeno2,5, Ryan W. Feathers1, Lomeli R. Carpio1, Tiffany R. Baker1,
Jennifer M. Carr1, Irene K. Yan1, Sahra Borges1, Edith A. Perez3, Peter Storz1, John A. Copland1, Tushar Patel1,
E. Aubrey Thompson1, Sandra Citi4 and Panos Z. Anastasiadis1,6
E-cadherin and p120 catenin (p120) are essential for epithelial homeostasis, but can also exert pro-tumorigenic activities. Here,
we resolve this apparent paradox by identifying two spatially and functionally distinct junctional complexes in non-transformed
polarized epithelial cells: one growth suppressing at the apical zonula adherens (ZA), defined by the p120 partner PLEKHA7 and
a non-nuclear subset of the core microprocessor components DROSHA and DGCR8, and one growth promoting at basolateral areas
of cellcell contact containing tyrosine-phosphorylated p120 and active Src. Recruitment of DROSHA and DGCR8 to the ZA is
PLEKHA7 dependent. The PLEKHA7microprocessor complex co-precipitates with primary microRNAs (pri-miRNAs) and
possesses pri-miRNA processing activity. PLEKHA7 regulates the levels of select miRNAs, in particular processing of miR-30b, to
suppress expression of cell transforming markers promoted by the basolateral complex, including SNAI1, MYC and CCND1. Our
work identifies a mechanism through which adhesion complexes regulate cellular behaviour and reveals their surprising
association with the microprocessor.
p120 catenin (p120) was identified as a tyrosine phosphorylation
substrate of the Src oncogene1 and an essential component of the
cadherin complex2. The interaction with p120 stabilizes E-cadherin
junctional complexes by preventing E-cadherin endocytosis25.
p120 also regulates the activity of Rho-GTPases, and thus the
organization of the actomyosin cytoskeleton69. By stabilizing
E-cadherin, p120 is expected to act as a tumour suppressor, and
mouse knockout studies support this notion10. However, p120 also
exhibits tumour-promoting activities, as an essential mediator of
anchorage-independent growth and cell migration induced by EGFR,
HER2, Rac1 or Src (refs 1113). This was partly attributed to the
expression of different cadherin family members14,15; however, p120
can induce tumour growth even in the presence of E-cadherin13,16
and is the essential intermediate for E-cadherin-mediated Rac1
activation and subsequent proliferation induction17. Consistent

with this, E-cadherin is still expressed in several types of aggressive


and metastatic cancer1820. Therefore, despite their significance
in epithelial adhesion and cellular regulation, present knowledge
on the role of E-cadherin and p120 in cancer is conflicting
and inconclusive.
In the present study, we sought to reconcile the apparently
contradictory observations and clarify the roles of p120 and
E-cadherin in epithelial cell behaviour. Recently, the p120 binding
partner PLEKHA7 was shown to specifically localize at the apical
zonula adherens (ZA) but not along lateral surfaces of epithelial cells,
as for p120 or E-cadherin21,22. By using PLEKHA7 as a marker of the
apical ZA in mature epithelial cells, we characterize two distinct p120associated complexes with antagonistic functions and we describe a
microRNA (miRNA)-mediated mechanism through which the ZA
suppresses transformed cell growth.

Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Mayo Clinic, 4500 San Pablo Road, Jacksonville, Florida 32224, USA. 2Department of
Molecular Biology, University of Geneva, 30 quai Ernest-Ansermet, CH-1211, Geneva 4, Switzerland. 3Division of Hematology/Oncology, Mayo Clinic, 4500 San Pablo
Road, Jacksonville, Florida 32224, USA. 4Department of Cell Biology and Institute of Genetics and Genomics of Geneva, University of Geneva, 30 quai
Ernest-Ansermet, CH-1211, Geneva 4, Switzerland. 5Present addresses: Division of Hematology, Oncology, Stem Cell Transplantation and Cancer Biology, Department
of Pediatrics, Stanford School of Medicine, Stanford, California 94305-5457, USA (S.P.N.); Department of Genetics and Complex Diseases, Harvard T. Chan School of
Public Health, 665, Huntington Avenue, Boston, Massachusetts 02115, USA (P.P.).
6
Correspondence should be addressed to P.Z.A. (e-mail: panos@mayo.edu)
Received 19 March 2015; accepted 20 July 2015; published online 24 August 2015; DOI: 10.1038/ncb3227

NATURE CELL BIOLOGY ADVANCE ONLINE PUBLICATION

2015 Macmillan Publishers Limited. All rights reserved

A RT I C L E S
RESULTS
Two distinct p120-associated populations exist at epithelial
junctions
Double immunofluorescence (IF) carried out in intestinal (Caco2)
and renal (MDCK) polarized monolayers confirmed previous results
that PLEKHA7 co-localizes with p120 or E-cadherin only in a narrow
area apically at the junctions, whereas p120 and E-cadherin are also
found basolaterally (Fig. 1a and Supplementary Fig. 1ac; refs 21,
22). The ZA markers afadin, circumferential actin and myosin IIA
(refs 23,24) co-localized precisely with PLEKHA7 (Supplementary
Fig. 1d), as previously shown22, verifying that PLEKHA7 labels the ZA
in these monolayers.
Unlike PLEKHA7, tyrosine phosphorylation of p120 at the Srctargeted sites Tyr 96 and Tyr 228 (ref. 25), which has been associated
with cancer11,26,27, was abundant basolaterally but not apically
(Fig. 1b and Supplementary Fig. 1e,f). In contrast, phosphorylation
of p120 at the non-Src-targeted Thr 310 site was both apical and
basolateral (Supplementary Fig. 1g). Total Src was distributed both
basolaterally and apically (Fig. 1c), although active Src, denoted
by auto-phosphorylation at Tyr 416, was absent from the ZA but
present at basolateral areas of cellcell contact (Fig. 1d), mirroring
the distribution of tyrosine-phosphorylated p120. Furthermore,
p130CAS, a Src target associated with increased cell mobility and
decreased junction stability28, was excluded from the ZA and was
abundant basolaterally (Fig. 1e).
We also examined the localization of total and active Rho and
Rac by co-staining with PLEKHA7. Total RhoA and Rho-GTP
were restricted at the ZA, whereas total Rac1 and Rac-GTP were
excluded from the ZA and were detected along basolateral cell
cell contacts (Fig. 1f,g and Supplementary Fig. 1hj). p190RhoGAP,
a Src substrate that suppresses RhoA activity29, was also excluded
from the ZA (Fig. 1h), further indicating that Rac1 and RhoA
activities are mutually exclusive along areas of cellcell contact.
Together, the data suggest the presence of an apical p120 complex
at the ZA associated with PLEKHA7, and a basolateral complex
lacking PLEKHA7 and encompassing markers associated with a more
aggressive cellular phenotype.
Proteomics identify two distinct junctional complexes in
polarized epithelial cells
To obtain further evidence for the existence of two distinct
junctional complexes, we biochemically separated them on the basis
of the strictly ZA-specific PLEKHA7 localization and the p120
localization throughout cellcell contacts in polarized cells (Fig. 1a and
Supplementary Fig. 1ac; ref. 22). Fully polarized Caco2 monolayers
were subjected to intracellular chemical cross linking to maintain the
interactions and then to either direct p120 immunoprecipitation (IP)
to isolate total p120, or a two-step IP, initially with PLEKHA7 to isolate
the apical complex, followed by a sequential p120 IP to isolate the
remaining p120 complexes (Fig. 2a). Western blot and silver staining of
these IPs indicated an efficient separation of the immunoprecipitated
proteins into two fractions (Fig. 2b). Mass spectrometry analysis
revealed 114 putative interactions, of which 47 were uniquely
associated with the apical fraction, 35 with the basolateral and 32 were
common (Fig. 2c and Supplementary Table 1). The apical complex
was enriched in structural and cytoskeletal proteins, such as actin

(ACTB), CKAP5, IQGAP1, ANXA2, MYL6, FLNA and ACTN1. In


contrast, the basolateral complex contained proteins involved in cell
cycle progression (CCAR1, CDC7, CDK5RAP2), signalling (S100A7,
S100A8), endocytosis (VPS33B, VIPAR) and cancer (MCC; Fig. 2c and
Supplementary Table 1).
Western blot of the isolated lysates confirmed the efficient
depletion of PLEKHA7 from the basolateral complex and the
successful IP of p120 by PLEKHA7 in the apical fraction (Fig. 2d).
pY228-p120 was absent from the apical complex lysates but abundant
in the basolateral ones (Fig. 2d), in agreement with the IF data
(Fig. 1b and Supplementary Fig. 1e). Western blot of the separated
fractions also confirmed that E-cadherin, together with - and
-catenin, co-precipitates with the apical complex (Supplementary
Fig. 2a), as suggested by our IF data (Supplementary Fig. 1b,c)
and previously published results21. Indeed, PLEKHA7 localization to
the junctions is E-cadherin and p120 dependent, as indicated by
loss of junctional PLEKHA7 after E-cadherin or p120 knockdown
(Supplementary Fig. 2b,c). Furthermore, only full-length murine
mp120-1A (1A), but not the mp120-4A (4A) isoform that lacks
the amino-terminal PLEKHA7-binding domain2,21, was able to
rescue junctional localization of PLEKHA7 in p120-depleted cells
(Supplementary Fig. 2c).
In full agreement with the proteomics data, TRIM21, IQGAP1,
ACTN1 and HNRNPA2B1 were detected only in the apical or total
p120 lysates, EWS in all lysates and CCAR1 solely in the basolateral
(Fig. 2d). In addition, IF of polarized cells showed strict apical
junctional co-localization of IQGAP1 with PLEKHA7 (Fig. 2e) and
co-localization of G3BP1 with p120 along both apical and basolateral
junctions (Fig. 2f), further validating the proteomics results (Fig. 2c).
Together, the data confirm the existence of two distinct junctional
complexes in polarized epithelial cells.
PLEKHA7 suppresses anchorage-independent growth and
expression of transformation-related markers
To define the functional role of the apical complex, we carried
out short hairpin RNA (shRNA)-mediated knockdown of PLEKHA7
(Supplementary Fig. 3a). Impedance measurements during junction
assembly of PLEKHA7-depleted cells showed both a delay in resistance
development and an overall lower transepithelial resistance of the
mature monolayer (Fig. 3a). PLEKHA7 knockdown also resulted
in fragmented circumferential actin (Fig. 3b) and a more flattened
cellular phenotype (Supplementary Fig. 3b). These results show that
depletion of PLEKHA7 compromises the integrity of the apical ZA, as
reported earlier21.
We then examined the effects of disrupting the ZA on the
ability of Caco2 cells to form colonies on soft agar, an established
assay for anchorage-independent growth. PLEKHA7-depleted
cells showed a threefold increase in colony formation (Fig. 3c
and Supplementary Fig. 3c). Furthermore, PLEKHA7 knockdown
increased p120 phosphorylation at Tyr 96 and Tyr 228, Src
phosphorylation at Tyr 416, and p130CAS phosphorylation at
Tyr 165 (Fig. 3d,e and Supplementary Fig. 3d). To account for the
morphological changes and induction of growth on PLEKHA7
depletion, we also examined the levels of transformation-related
and mesenchymal markers15,30,31. PLEKHA7 knockdown resulted
in increased expression of SNAI1, cadherin 11 (Cad11), cyclin

NATURE CELL BIOLOGY ADVANCE ONLINE PUBLICATION

2015 Macmillan Publishers Limited. All rights reserved

A RT I C L E S
p120

Merged

Apical

PLEKHA7

PLEKHA7

p120: Tyr228

Merged

Apical

z
x

Basal

Basal

x
Src

Merged

Apical

PLEKHA7

PLEKHA7

Src: Tyr 416

Merged

Apical

z
x

Basal

Basal

x
p130CAS

Merged

Apical

PLEKHA7

PLEKHA7

Rho-GTP

Merged

Apical

Basal

Basal

Rac-GTP

Merged

PLEKHA7

p190

Merged

Apical

Apical

PLEKHA7

Basal

Basal

Figure 1 Polarized epithelial cells show distinct p120-associated populations


at the junctions. Caco2 cells were grown for 21 days to polarize and
subjected to IF for PLEKHA7 and (a) p120, (b) phosphorylated p120
Tyr 228, (c) Src, (d) phosphorylated Src Tyr 416; (e) p130CAS and
(h) p190RhoGAP. Also, Caco2 cells were transfected with (f) a green
fluorescent protein (GFP)rGBD (rhotekin RhoA-binding domain) construct
to detect active Rho (Rho-GTP) or (g) a yellow fluorescent protein (YFP)
PBD (PAK-binding domain) construct to detect active Rac (Rac-GTP), and

co-stained with PLEKHA7. In all cases, stained cells were imaged by


confocal microscopy and image stacks were acquired, covering the entire
polarized monolayer between the basal and the apical level. Representative
xy image stacks and merged composite xz images are shown. Enlarged
parts of merged images in f and g indicate areas of cellcell contact. Scale
bars for xy images, 20 m; for xz images, 5 m; for enlarged parts of f
and g, 3 m. PLEKHA7 background staining in f and g is an artefact of
paraformaldehyde fixation.

D1 (CCND1), MYC, FAK and VIM (Fig. 3f and Supplementary


Fig. 3d), whereas re-expression of PLEKHA7 reversed these effects
and suppressed p120 phosphorylation (Fig. 3g). Indeed, ectopic
overexpression of SNAI1 in Caco2 cells at modest levels was adequate
to induce a twofold increase in soft agar colony formation (Fig. 3h
and Supplementary Fig. 3e).
Knockdown of NEZHA, the intermediate link between PLEKHA7
and the microtubules21, retained PLEKHA7 at the ZA but failed to
phenocopy the effects of PLEKHA7 depletion on marker expression

(Supplementary Fig. 3f,g), suggesting that microtubules are not


involved in the observed effects. Next, we tested whether the effects of
PLEKHA7 knockdown are due to its loss from the ZA, by removing
PLEKHA7 from the junctions but retaining its total levels in the
cells. To do this, we transfected Caco2 cells with a p120 mutant
that cannot bind E-cadherin and is cytoplasmic, because it lacks
the first armadillo repeat (mp120-1ARM1; ref. 2), but can bind
PLEKHA7, because it contains the N-terminal PLEKHA7-binding
domain21. Expression of this construct sequestered PLEKHA7 to the

NATURE CELL BIOLOGY ADVANCE ONLINE PUBLICATION

2015 Macmillan Publishers Limited. All rights reserved

A RT I C L E S

199
116
79

p120 (total p120 to control)

52

AHNAK SHROOM3 MYO1D MCC


PPL
OSTM1
IQGAP1
VAV2
PLUNC
DSG1
ANXA2
ACTB ACTN1
TMEM128
ANXA4
FLG2
ANXA1 SNRPB
CKAP5
TTN
ODZ2
DPP4
FAT2
MYL6 ANXA5
LRP1
CLTC
CTNNB1
CES1
ODZ3
DSP
FZD2
CFTR
SPASTIN
SSR1
CTNNA1
JUP
HPCAL1
FLNA ARPC5
TRIM10
PITPNB
CCAR1
VIPAR
DYNC1H1
CDH1
TRIM21
HSP90A1
S100A8
ETAA1 PSPC1
HSPA8 EIF3D
VPS33B VPS35
G3BP1
SCYL2 AHCY
S100A7
HSPA1A
NOS1
Apical
Basolateral
G3BP2
CDC7
HSPA9 EIF3A
GLG1
SLC25A1
WDR67
PPP1CA
SFPQ CAPRIN1
HNRNPA2B1
MLEC
KPRP
DCP1B
KL
NCOA3
APOA4 SLC25A5
CLYBL
DDX1
PGLS
HNRNPH3
RELN
MSH5
FUS YWHAE
EWS
SERPINB12
NDUFV1 HNRNPA3
DDX4
CDK5RAP2
DNAH7
HNRNPA1L2
WDR82 SLC25A3
CYB5R3
DDX17 NONO THOC4
SYNE1
RALY
LKAP
FEM1A
ATP5C1 ALDH18A1 DLGAP1 GAPDH C14orf166 SELT

Silver stain

Mr (K)

37
29

20

p1

12

ra
l
la
te

Ig
G

17

A7

Lyse,
IP with:

p120:
basolateral
complex

lp

complexes

Then

ta

for 21 days
to polarize

PLEKHA7:
apical
complex

Ba

so

To

Crosslink

PL

Grow Caco2s

EK
H

IPs

WBs

PLEKHA7

IQGAP1

Merged

Apical

PLEKHA7

p120

Basal

p-p120: pY228
y

TRIM21
x

IQGAP1

p120

G3BP1

Merged

Apical

ACTN1
HNRNPA2B1

Basal

EWS

PL IgG
EK
H
A7
Ba
t
so al p
12
la
te
0
ra
lp
12
0

Caco2
x

To

In

pu

CCAR1

IPs

Figure 2 Biochemical separation of two distinct junctional complexes


by proteomics. (a) Outline of the methodology to isolate the apical and
basolateral complexes from polarized Caco2 cells. (b) SDSpolyacrylamide
gel electrophoresis (SDSPAGE) and silver stain of the isolated junctional
fractions; green arrowheads indicate examples of apical-specific bands,
whereas red arrowheads indicate basolateral-specific bands. (c) Schematic
representation of the proteins identified after mass spectrometry of the
isolated junctional fractions, identified in the apical (green outlines), the
basolateral (red) or both fractions (yellow). Grey connections indicate known

proteinprotein interactions. See Supplementary Table 1 for the list of


proteins and mass spectrometry peptide counts. (d) Western blot (WB) of
the lysates from the separated fractions for PLEKHA7, p120 and markers
identified in the apical (TRIM21, IQGAP1, ACTN1, HNRNPA2B1), the
basolateral (CCAR1) and both fractions (EWS). The isolated fractions were
also blotted for phosphorylated p120 Tyr 228 (p-p120: pY228), another
basolateral marker (Fig. 1b). See Supplementary Fig. 8 for unprocessed blot
scans. (e,f) Polarized Caco2 cells were stained and imaged as in Fig. 1 for
PLEKHA7 and IQGAP1 (e) and for p120 and G3BP1 (f). All scale bars: 20 m.

cytoplasm without altering its total levels and phenocopied the effects
of PLEKHA7 depletion on the levels of the same markers examined
above (Supplementary Fig. 3h,i). Therefore, it is the disruption of

the PLEKHA7-associated apical ZA complex that results in induction


of growth-related signalling. Consistent with this, examination of
the expression and localization of PLEKHA7, p120 and E-cadherin

NATURE CELL BIOLOGY ADVANCE ONLINE PUBLICATION

2015 Macmillan Publishers Limited. All rights reserved

A RT I C L E S

Junctional resistance
(normalized units)

b PLEKHA7

Junctional integrity

14

12

Apical actin
ring intensity

NT

Actin
120
Percentage change

10
8

NT

PLEKHA7 shRNA

4
2

60
40
20

12

24

36

48
h

60

72

84
PLEKHA7 shRNA

PL NT
E
sh KHA
RN 7
A

Fold change

80

100

PLEKHA7

PLEKHA7

p-p120: Y228

p-Src: Y416

p-p120: Y96

Src

p120

p-p130CAS: Y165

Actin

p130CAS
NT No. 8 No. 10

Actin

PLEKHA7 shRNA

NT No. 8 No. 10

NT No. 8 No. 10

PLEKHA7 shRNA

PLEKHA7 shRNA

g
PLEKHA7

PLEKHA7

2.5

p-p120: pY228

SNAI1

2.0

CCND1

Fold change

p-p120: pY96

MYC
p120
Cad11

1.5
1.0
0.5

VIM

SNAI1

FAK

CCND1

0
SNAI1

MYC

-tubulin

Actin

PLEKHA7 shRNA

ad
G
FP
ad
SN
AI
1

Cad11

NT No. 8 No. 10

Actin
PLEKHA7 shRNA
LZRS-PLEKHA7

+
+

Figure 3 PLEKHA7 suppresses anchorage-independent growth and


expression of transformation-related markers. (a) Caco2 non-target control
shRNA (NT) and PLEKHA7 shRNA knockdown cells were measured for
cell impedance using the ECIS apparatus to assess junctional resistance
(mean s.d. from n = 3 independent experiments calculated for 96 h and
7,000 continuous time points from 40,000 cells per condition, NT or
PLEKHA7 shRNA, per experiment; P < 0.03 is shown for every 12 h interval,
Students two-tailed t-test). (b) Caco2 control (NT) and PLEKHA7 knockdown
(PLEKHA7 shRNA) cells co-stained for PLEKHA7 and actin (phalloidin) and
quantified (right panel) for apical ring phalloidin intensity (mean s.d. from
n = 3 independent experiments, assessing 40 cells in total per condition,
NT or PLEKHA7 shRNA, P = 0.01, Students two-tailed t-test; PLEKHA7
background staining is an artefact of paraformaldehyde fixation for actin).
Scale bar for full images, 20 m; for enlarged parts, 10 m. (c) Caco2 control
(NT) and PLEKHA7 knockdown cells using two shRNAs (PLEKHA7 shRNA

no. 8, no. 10) were quantified for colony formation on soft agar (mean s.d.
from n = 3 independent experiments; P < 0.003, Students two-tailed t-test;
see Supplementary Fig. 3c for representative scan). (df) Caco2 control
(NT) and PLEKHA7 knockdown cells using two shRNAs (no. 8, no. 10)
analysed by western blot for the indicated markers. Phosphorylation sites are
denoted by p-. Actin is the loading control. (g) Western blot of PLEKHA7
knockdown Caco2 cells (PLEKHA7 shRNA) after ectopic re-expression of
PLEKHA7 (LZRS-PLEKHA7) for the markers shown. Actin is the loading
control. (h) Western blot and soft agar assay quantification of Caco2 cells
infected with either vector control (adGFP) or a SNAI1-expressing construct
(adSNAI1) (mean s.d. from n = 3 independent experiments; P = 0.006
Students two-tailed t-test; see Supplementary Fig. 3e for representative
scan). -tubulin is the loading control. Source data for ac,h are provided
in Supplementary Table 2. See Supplementary Fig. 8 for unprocessed blot
scans of dg.

in normal and cancer samples of breast and kidney revealed that


PLEKHA7 is either mislocalized or lost in the tumour tissues, although
p120 and E-cadherin are still expressed and junctional to a high

percentage in the same samples (Supplementary Fig. 4a,b). These


data indicate the selective disruption of the ZA in tumour tissues, in
agreement with our in vitro findings.

NATURE CELL BIOLOGY ADVANCE ONLINE PUBLICATION

2015 Macmillan Publishers Limited. All rights reserved

A RT I C L E S
The basolateral complex promotes cell growth and expression
of related markers
Next, we examined whether the events induced by disruption of the
apical complex are due to the activity of the basolateral complex.
In agreement, both the increased growth and the elevated levels
of transformation-related markers in PLEKHA7-depleted cells were
abolished when p120 was simultaneously knocked down (Fig. 4a,b
and Supplementary Fig. 4c). PLEKHA7 knockdown did not affect
E-cadherin levels; however, p120 depletion reduced E-cadherin levels,
as expected2,5 (Supplementary Fig. 4d). Interestingly, simultaneous
knockdown of E-cadherin decreased the levels of SNAI1, CCND1,
MYC and phosphorylation of p130CAS in PLEKHA7-depleted cells
(Fig. 4c), indicating that the effects observed in the absence of
PLEKHA7 depend on E-cadherin-based junctions. Furthermore,
suppression of Src activity using the PP2 inhibitor abolished p120
and p130CAS phosphorylation and also reversed the increased
expression of SNAI1, CCND1, MYC and cadherin 11 after PLEKHA7
knockdown (Fig. 4d and Supplementary Fig. 4e). Re-expression of
wild-type murine p120 (mp120-1A; ref. 25) in p120 knockdown
cells partially restored p120 phosphorylation as well as SNAI1 and
cadherin 11 levels, whereas expression of a non-phosphorylatable
p120 construct (mp120-8F; ref. 25) at similar levels failed to reproduce
these effects (Fig. 4e).
Depletion of cadherin 11, which was previously implicated in
p120-mediated growth and migration signalling14,15, also reversed the
increased levels of SNAI1, CCND1 and MYC on PLEKHA7 depletion
(Fig. 4f). As E-cadherin levels are unaltered in the PLEKHA7-depleted
cells (Fig. 4c and Supplementary Fig. 4d), the data do not suggest a
cadherin switch, but rather that the effects of PLEKHA7 depletion are
mediated by the cadherin-based basolateral junctions. Indeed, p120
tyrosine phosphorylation occurs only at the junctions (Fig. 1b and
Supplementary Figs 1e,f and 4e; refs 32,33), and cadherin 11 exhibits
strong junctional staining after PLEKHA7 knockdown (Fig. 4g). Collectively, the results indicate that the basolateral complex acts through
E-cadherin, cadherin 11, Src and p120 to promote transformationrelated signalling, after disruption of the apical complex.
PLEKHA7 suppresses expression of SNAI1, MYC and CCND1
through miRNAs
PLEKHA7 knockdown did not significantly alter the messenger RNA
(mRNA) levels of the examined markers (Supplementary Fig. 5a),
did not prolong protein stability after cycloheximide treatment
(Supplementary Fig. 5b) and did not alter the rate of stabilization after
MG-132 treatment (Supplementary Fig. 5c). PLEKHA7 knockdown
also did not affect the phosphorylation of GSK3B, a dominant
regulator of SNAI1 stability34,35 (Supplementary Fig. 5d). These
results suggest that PLEKHA7 is not affecting the examined markers
transcriptionally or post-translationally, but imply that it may suppress
mRNA expression. A core such mechanism is RNA interference
(RNAi), which involves the miRNA-mediated silencing of mRNA
expression3639 and is commonly deregulated in cancer40,41. To
examine this, we used the NanoString platform and found significant
changes in the levels of 29 miRNAs out of 735 examined in PLEKHA7depleted cells: 15 of them were decreased and 14 miRNAs increased
(Fig. 5a). The strongest decrease was observed for miR-24, miR-30a
and let-7g, and the strongest increase for miR-19a. We also focused on

the most abundant of the affected miRNAs, namely miR-30b, which


was among those decreased (Fig. 5a). Quantitative PCRs with reverse
transcription (qRTPCRs) confirmed the NanoString results (Fig. 5b),
whereas PLEKHA7 re-expression rescued the altered miRNA levels
(Supplementary Fig. 5e,f).
We then used anti-miRNAs to target miR-24, miR-30a, miR-30b
and let-7g in wild-type PLEKHA7-containing cells, to mimic the
effect of PLEKHA7 knockdown on their expression. Indeed, the antimiRNA transfections resulted in increased levels of SNAI1, MYC and
CCND1 (Fig. 5c), similar to the effect of PLEKHA7 depletion (Fig. 3f).
The miR-30 and let-7 miRNA families are validated suppressors of
SNAI1 (refs 4244), whereas miR-24 and the let-7 family are also
verified suppressors of MYC (refs 4547). Re-expression of miR-30b in
PLEKHA7-depleted cells using a miR-30b mimic construct reversed
the increase of SNAI1, MYC and CCND1 (Fig. 5d), confirming that
miR-30b is a mediator of PLEKHA7s growth-suppressing signalling.
Interestingly, anti-miR-30a and anti-miR-30b also increased Src and
p120 phosphorylation, whereas the miR-30b mimic reversed their
increase in PLEKHA7-depleted cells (Supplementary Fig. 5g,h). In
addition, ectopic SNAI1 overexpression (Fig. 3h) induced p120
phosphorylation at Tyr 228 (Supplementary Fig. 5i), suggesting that
the effect of miR-30b on p120 phosphorylation is mediated by SNAI1.
In summary the data reveal that a specific subset of miRNAs, and in
particular miR-30b, are regulated by PLEKHA7 and mediate its effects
on proteins involved in transformed cell growth.
The ZA associates with the microprocessor complex
Although PLEKHA7 knockdown decreased levels of mature miR-30b
(Fig. 5a,b), it did not affect expression of the primary miR-30b
(pri-miR-30b) transcript (Fig. 6a); however, it resulted in decreased
levels of the next precursor, pre-miR-30b (Fig. 6a and Supplementary
Fig. 6a). This suggested that PLEKHA7 affects pri-miR-30b processing
to pre-miR-30b, a step in miRNA biogenesis catalysed by the
microprocessor complex and its two essential components DROSHA
and DGCR8 (refs 4851). We tested whether PLEKHA7 depletion
affects either the total levels of DROSHA or DGCR8, or their
localization, because the microprocessor is thought to function in
the nucleus. Whereas both the overall levels of the two proteins and
their subcellular distribution remained unchanged, cell fractionation
assays revealed the existence of a non-nuclear fraction of both
DROSHA and DGCR8 (Fig. 6b). IF and confocal microscopy
confirmed their presence outside the nucleus, particularly at areas
of cellcell contact (Fig. 6c), and revealed their co-localization with
PLEKHA7 and p120 strictly at the apical ZA of polarized epithelial
cells (Fig. 6dg and Supplementary Fig. 6be). The use of alternative
antibodies for DROSHA and DGCR8 (Supplementary Fig. 6b,c)
as well as elimination of their staining after knockdown by short
interfering RNAs (siRNAs) (Supplementary Fig. 6f,g) confirmed the
specificity of their junctional localization. In agreement, DROSHA
and DGCR8 co-precipitated with PLEKHA7 (Fig. 6h,i), p120 (Fig. 6i
and Supplementary Fig. 6h) and E-cadherin (Supplementary Fig. 6h).
Notably, the interaction of PLEKHA7 with the microprocessor is RNA
independent (Fig. 6j), excluding the possibility that these interactions
are due to non-specific isolation of large ribonucleoprotein granules
from the cells. Proximity ligation assay confirmed that the interaction
of DROSHA and PLEKHA7 occurs only at the junctions (Fig. 6k).

NATURE CELL BIOLOGY ADVANCE ONLINE PUBLICATION

2015 Macmillan Publishers Limited. All rights reserved

A RT I C L E S
a

b
2.0

Fold change

PLEKHA7

PLEKHA7

PLEKHA7

p120

Ecad

1.5

p-p130CAS: Y165

p-p130CAS: Y165

1.0

p130CAS

p130CAS

p-p120: Y228
p120
p-p130CAS: Y165

SNAI1
0.5

p130CAS

SNAI1

SNAI1

CCND1

CCND1

CCND1
1

PLEKHA7 shRNA

p120 shRNA

Cad11

MYC

+
+

MYC

MYC

Actin

Actin

PLEKHA7 shRNA
p120 shRNA

p120
p-p120: Y228

N
T
RN
A

Cad11
+
+

PLEKHA7 shRNA
Ecad shRNA

Actin

PLEKHA7

PLEKHA7 shRNA
Src inh: PP2

+
+

PLEKHA7

+
+

Cad11

Cad11

p1
20

sh

SNAI1
SNAI1

NT

CCND1

Cad11

MYC
Actin

Actin

+
+

PLEKHA7 shRNA

Ve
ct
p1 or
20
m
p1 1A
20
-8
F

PLEKHA7 shRNA
Cad11 shRNA

Figure 4 The basolateral junctional complex promotes anchorageindependent growth and expression of transformation-related markers.
(a) Caco2 control, PLEKHA7 knockdown (PLEKHA7 shRNA) and PLEKHA7
p120 double knockdown (PLEKHA7 shRNA, p120 shRNA) cells were
grown on soft agar and quantified for colony formation (mean s.d.
from n = 3 independent experiments; P = 0.0008, Students two-tailed
t-test; see Supplementary Fig. 4c for representative scan). Source
data are provided in Supplementary Table 2. (b) Control, PLEKHA7
knockdown (PLEKHA7 shRNA) and PLEKHA7p120 double knockdown
(PLEKHA7 shRNA, p120 shRNA) Caco2 cells were analysed by western blot
for the markers shown. Actin is the loading control (see also Supplementary
Fig. 4d). (c) Control, PLEKHA7 knockdown (PLEKHA7 shRNA) and
PLEKHA7/E-cadherin (PLEKHA7 shRNA, Ecad shRNA) double knockdown
Caco2 cells were analysed by western blot for the markers shown. Actin

is the loading control. (d) Western blot of Caco2 control or PLEKHA7


knockdown (PLEKHA7 shRNA) cells after treatment with vehicle control
(dimethylsulphoxide, DMSO) or 10 M Src inhibitor PP2; actin is the
loading control (see also Supplementary Fig. 4e). (e) Western blot of p120
knockdown Caco2 cells (p120 shRNA) after ectopic expression of either
vector control (vector), murine full-length (isoform 1A) wild-type p120
(mp120-1A) or murine full-length non-phosphorylatable p120 (mp1208F) for the markers shown. Actin is the loading control. (f) Western blot
of single and double PLEKHA7 (PLEKHA7 shRNA) and cadherin 11
(Cad11 shRNA) knockdown Caco2 cells for the markers shown. Actin
is the loading control. (g) IF of control (NT) or PLEKHA7 knockdown
(PLEKHA7 shRNA) cells for PLEKHA7 and cadherin 11 (Cad11). Scale
bar: 20 m. See Supplementary Fig. 8 for unprocessed blot scans
of bf.

The junctional localization of DROSHA and DGCR8 was disrupted


in PLEKHA7-depleted cells, indicating that it is PLEKHA7 dependent
(Fig. 7a,b). Removal of PLEKHA7 from the junctions by the
mp120-1ARM1 mutant also resulted in compromised junctional
localization of DROSHA and DGCR8 (Supplementary Fig. 7ac).
Furthermore, depletion of p120, which is required for PLEKHA7
recruitment to the junctions (Supplementary Fig. 2c), also negatively
affected DROSHA junctional localization (Supplementary Fig. 7d)
and decreased the levels of the examined miRNAs (Supplementary
Fig. 7e), mimicking the effects of PLEKHA7 depletion. Conversely,
strengthening of the junctions by Src inhibition33 increased junctional
localization of PLEKHA7, DROSHA and DGCR8 (Fig. 7ce). As
PLEKHA7 tethers the ZA to the microtubules21, we examined
whether the localization of DROSHA and DGCR8 at the ZA
is microtubule dependent. Calcium switch assays showed that
the junctional localization of PLEKHA7, DROSHA and DGCR8

recovers rapidly after re-addition of calcium, and is not affected


by dissolution of microtubules by nocodazole (Fig. 7fi). Similarly,
neither prolonged nocodazole treatment (Supplementary Fig. 7f), nor
NEZHA knockdown (Supplementary Fig. 7g,h) affected the junctional
localization of PLEKHA7, DROSHA or DGCR8. Although both
miR-30b (Fig. 5d) and p120 or cadherin 11 knockdown (Fig. 4b,f)
rescue the levels of pro-tumorigenic markers in PLEKHA7-depleted
cells, simultaneous knockdown of either p120 or cadherin 11 with
PLEKHA7 did not reverse either the decreased miRNA levels
(Supplementary Fig. 7i) or the compromised junctional localization
of DROSHA and DGCR8 (Supplementary Fig. 7jm), indicating that
these events are PLEKHA7 specific and that p120 and cadherin 11
induce cell transforming markers through a separate pathway. Taken
together, the data reveal the association of the microprocessor complex
with the apical ZA in non-transformed epithelial cells, in a PLEKHA7dependent manner.

NATURE CELL BIOLOGY ADVANCE ONLINE PUBLICATION

2015 Macmillan Publishers Limited. All rights reserved

A RT I C L E S
a

NanoString: PLEKHA7 shRNA versus NT

29

16

38

34

21

94

36

27

24

46

39
11
0

61

31

12

29

9
59

85

31
32

23

16

17

3
35

19

10

14

55

19

Relative abundance

34

Fold change (P < 0.05)

Relative abundance

a-

hs

hs

a-

hs

ahs

let

-7

83

iR

-1

-7
let

ahs

a-

hs

0a

-2

-3

iR

iR
m

m
ahs

iR
am 30
hs iR- d
4
am 50a
iR
14
hs
8b
am
iR
-3
hs
01
aa
m
hs iR-3
a0
b
m
hs iR-3
a0e
m
iR
-1
hs
5b
am
i
R
hs
-1
03
am
iR
-2
hs
21
a
hs
a- -miR
m
-2
iR
0
-4
hs
50 3
abm
5p
iR
-2
9
6hs
5p
ahs miR
-2
ahs miR 23
a-1
m
iR 30b
-4
23
hs
-3
ap
m
iR
hs
-6
a19
m
iR
-1
27
hs
4b
am
iR
hs
a93
4
hs miR
am -124
iR
6
-3
hs
31
a-3
m
p
iR
-3
hs
02
am
b
iR
hs
a- -30
2a
m
iR
-3
hs
02
ad
m
iR
-1
9a

2.2

NT

2.0

PLEKHA7 shRNA no. 8

1.8

PLEKHA7 shRNA no. 10

PLEKHA7 shRNA

PLEKHA7

1.6
Fold change

NT

1.4

SNAI1

SNAI1

MYC

MYC

CCND1

CCND1

Actin

Actin

1.2
1.0

0.8

0.6

0.4

0.2

0b
-3
iR

on

tro

0b
-3
iR

on

tro

C
on
tro
l
am
iR
a24
m
iR
-3
0a
am
iR
-3
0b
ale
t-7
g

9a
-1
m
iR

m
iR

-3

0b

g
t-7
le

-3
0a
iR
m

iR

-2
4

Figure 5 PLEKHA7 suppresses protein expression through miRNAs. (a) Caco2


control (NT) and PLEKHA7 knockdown (PLEKHA7 shRNA) cells analysed
for miRNA expression using NanoString. Results shown are statistically
significant changes (mean s.d. from n = 3 independent experiments;
P < 0.05, Students two-tailed t-test) shown as average fold change of
PLEKHA7 shRNA versus control cells. Numbers on each bar are NanoString
counts indicating relative abundance of the respected miRNAs (see also
Methods). Source data are deposited at Gene Expression Omnibus, accession
number GSE61593. (b) qRTPCR analysis of Caco2 control (NT) or PLEKHA7

knockdown (PLEKHA7 shRNA no. 8, no. 10) cells for the indicated miRNAs
(mean s.d. from n = 3 independent experiments; P < 0.01, Students
two-tailed t-test). Source data are provided in Supplementary Table 2.
(c) Western blot of Caco2 cells transfected with the indicated anti-miRNAs
for the markers shown; actin is the loading control. (d) Caco2 control (NT)
and PLEKHA7 knockdown (PLEKHA7 shRNA) cells were transfected with
either control or miR-30b mimic and blotted for the markers shown; actin
is the loading control. See Supplementary Fig. 8 for unprocessed blot scans
of bd.

PLEKHA7 regulates processing of pri-miR-30b at the junctions


Subcellular fractionation revealed the existence of at least three
pri-miRNAs, pri-miR-30b, pri-let-7g and pri-miR-19a, outside the
nucleus (Fig. 8a), and RNA-IPs indicated that PLEKHA7 is enriched
in these pri-miRNAs (Fig. 8b). In vitro processing assays using
pri-miR-30b as a template showed that the PLEKHA7-associated
complex possesses pri-miRNA processing activity (Fig. 8c). PLEKHA7
depletion decreased in vitro processing of pri-miR-30b by about 30%
(Fig. 8d,e), similar to the decrease observed in endogenous pri-miR30b processing after PLEKHA7 knockdown (Fig. 6a). Finally, in situ
hybridization (ISH) confirmed the localization of pri-miR-30b at
the junctions (Fig. 8f). Collectively, our results demonstrate that the
ZA-localized microprocessor complex is active and that PLEKHA7
regulates processing at least of pri-miR-30b at the ZA.

In summary, the data support a model whereby the apical ZA


complex recruits the microprocessor through PLEKHA7 to suppress
growth-related signalling promoted by the basolateral junctional
complex, hence maintaining the normal epithelial architecture and
behaviour (Fig. 8g).

DISCUSSION
Our work shows that the dual p120 and E-cadherin behaviour
in epithelial cell growth can be explained by the existence of
two distinct p120 complexes with opposing functions: an apical
complex at the ZA that suppresses growth-related signalling
through PLEKHA7-mediated regulation of select miRNAs and
a basolateral complex that promotes anchorage-independent
growth and increased expression of markers such as SNAI1, MYC

NATURE CELL BIOLOGY ADVANCE ONLINE PUBLICATION

2015 Macmillan Publishers Limited. All rights reserved

A RT I C L E S
a

b
DROSHA

1.0
Fold change

Nonnuclear Nuclear

Total

DGCR8

0.8

Non-polarized

1.2

NT
PLEKHA7 shRNA no. 8

0.6

Lamin A/C

0.4

Histone H3

DROSHA

DGCR8

DAPI

Merged

0.2
GAPDH

0b

e-

iR
-3

A7 N
s T
PL hR
EK NA
H
A7
PL
sh NT
EK
RN
H
A
A7
sh NT
RN
A

Actin

WBs
PLEKHA7
p120

PL
EK
H

pr

pr

i-m

iR
-3

0b

DROSHA
DGCR8

DAPI

PLEKHA7

DROSHA

In
pu
t
PL Ig
EK G
H
A
p1 7
20

IPs

Merged

Apical

i
x

WBs
PLEKHA7

Basal

p120
DROSHA

Dr IgG
o
DG sha
C
R8

In
pu
t

DGCR8

IPs

j
PLEKHA7

DGCR8

PLEKHA7

Merged

Apical

DROSHA
DGCR8

Basal

In
pu

IPs

k
x

DROSHA
p120

DGCR8
p120

z
x

PLEKHA7+
DROSHA

PLEKHA7+
p120

Neg. control

Pr
ox
lig imi
at ty
io
n

PL I
E gG
PL KH
A
+ EK 7
RN HA
As 7
e

DAPI

WBs

pr

ox Pha
im se
ity D
lig AP
at I
io
n

Figure 6 PLEKHA7 associates with DROSHA and DGCR8 at the ZA.


(a) qRTPCR analysis of pri-miR-30b and pre-miR-30b in Caco2 control (NT)
or PLEKHA7 knockdown (PLEKHA7 shRNA) cells (mean s.d. from n = 3
independent experiments; P = 0.02, Students two-tailed t-test). Source
data are provided in Supplementary Table 2. (b) Western blot of either
total cell lysates or of nuclear and non-nuclear lysates after subcellular
fractionation of Caco2 NT or PLEKHA7 shRNA cells for the indicated
markers. Lamin A/C and histone H3 are the nuclear and GAPDH the
cytoplasmic markers; actin is the total loading control. (c) IF of nonpolarized Caco2 cells, co-stained for DROSHA and DGCR8; 4,6-diamidino2-phenylindole (DAPI) is the nuclear stain. (d,e) IF of polarized Caco2
cells co-stained for PLEKHA7 and DROSHA or DGCR8. Enlarged details in
boxes are shown above the apical fields; a composite xz merged image is
shown at the side of the xy image stack (antibodies used here: DROSHA,

Abcam; DGCR8, Abcam; see also Supplementary Fig. 6be for a second
set of antibodies used and Supplementary Table 3 for antibody details).
DAPI is the nuclear stain. (f,g) Composite xz images of polarized Caco2
cells co-stained by IF for p120 and DROSHA or DGCR8 (see Supplementary
Fig. 6b,c, respectively, for xy set of image stacks). (h,i) Western blots of
PLEKHA7, p120, DROSHA and DGCR8 IPs for the markers shown. IgG is
the negative IP control. (j) Western blot of PLEKHA7 IPs with and without
RNAse treatment; IgG is the negative control. (k) Proximity ligation assay
using PLEKHA7 and DROSHA antibodies, PLEKHA7 and p120 antibodies
(assay positive control) or a p120 antibody only (assay negative control); the
composite phase contrastDAPIproximity ligation images are also shown.
Scale bars for xy images, 20 m; for xz images, 5 m; for enlarged parts
of d and e, 3 m. See Supplementary Fig. 8 for unprocessed blot scans
of b,hj.

NATURE CELL BIOLOGY ADVANCE ONLINE PUBLICATION

2015 Macmillan Publishers Limited. All rights reserved

A RT I C L E S
a PLEKHA7

c p120

PLEKHA7

b p120

DROSHA

DGCR8

NT

NT

PLEKHA7 shRNA

PLEKHA7 shRNA

d p120

e p120

DROSHA

DGCR8

Control

Control

Control

PP2

PP2

PP2

-tubulin

f
Ca2+: 0 min

Ca2+: 30 min

g
Ca2+: 60 min

Ca2+: 0 min

Control

Nocodazole
DROSHA

Ca2+: 0 min

Ca2+: 60 min

Control

Nocodazole

PLEKHA7
Ca2+: 30 min

Ca2+: 30 min

DGCR8

i
Ca2+: 60 min

Ca2+: 0 min

Control

Control

Nocodazole

Nocodazole

Ca2+: 30 min

Ca2+: 60 min

Figure 7 Localization of DROSHA and DGCR8 at the ZA is PLEKHA7


dependent. (a,b) IF of control (NT) or PLEKHA7 knockdown
(PLEKHA7 shRNA) Caco2 cells for DROSHA and DGCR8, co-stained for
PLEKHA7 or p120. PLEKHA7 background intracellular staining in a is an
artefact of paraformaldehyde fixation. (ce) Caco2 cells treated either with
vehicle control (DMSO) or the Src inhibitor PP2 (10 M) were stained by IF
for PLEKHA7, DROSHA and DGCR8 and co-stained with p120. (fi) Calcium

switch assay of Caco2 cells for the indicated times after Ca2+ re-addition
(Ca2+ : 0 min indicates Ca2+ depleted cells immediately before Ca2+ readdition), treated with vehicle (control) or with 10 M nocodazole, and
stained by IF for -tubulin, PLEKHA7, DROSHA and DGCR8. Enlarged
image details are shown in yellow boxes on the right-hand side of the image
stacks in a, b, d and e. Scale bars for full images, 20 m; for enlarged
parts, 8 m.

and CCND1 through activated Src and tyrosine-phosphorylated


p120 (Fig. 8g).
DROSHA and DGCR8 are thought to regulate miRNA processing
in the nucleus4850. However, these proteins have not been studied
in the context of non-transformed, polarized mammalian epithelial
cells. Our data reveal that, in these cells, a functional subset of the
microprocessor is recruited by PLEKHA7 to the ZA (Figs 6dk and
7a,b). PLEKHA7 does not globally regulate miRNA expression in
the cells examined, but affects only a specific subset of miRNAs
(29 out of 735 examined; Fig. 5a) in a confluence-independent
manner, because all experiments herein were carried out in fully

confluent epithelial monolayers. Importantly, the interaction of


PLEKHA7 with DROSHA (Fig. 6k) or pri-miR-30b (Fig. 8b,f)
occurs exclusively at the ZA, whereas depletion of PLEKHA7
dissociates DROSHA and DGCR8 from the ZA and reduces miR30b processing, without affecting nuclear DROSHA and DGCR8
levels (Figs 6b, 7a,b and 8d,e). Therefore, the present study does
not dispute the general model of miRNA biogenesis, but uncovers
a mechanism whereby PLEKHA7 recruits a subpopulation of the
microprocessor to the ZA and regulates processing of select miRNAs
to suppress the expression of markers involved in transformed
cell growth.

10

NATURE CELL BIOLOGY ADVANCE ONLINE PUBLICATION

2015 Macmillan Publishers Limited. All rights reserved

A RT I C L E S

Lamin A/C
Histone H3

1,000

100

10

i-m
pr

pri-miR-30b
pri-let-7g
pri-miR-19a

on
-

nu

cl
ea
uc r
le
ar

GAPDH

Nuclear
Non-nuclear

Ig
G
PL
EK
H
A7
DG
C
R8

DGCR8

40
38
36
34
32
30
28
26
24
22
20
18
16

Fold enrichment (log10)

qPCR: Ct values

DROSHA

iR
pr -30
i-l b
pr et
i-m -7
iR g
-1
9a
AC
TB

50

NT
IPs:
nt:
1,000
500
300
150

PLEKHA7 shRNA

1.2
primiR-30b

80
premiR-30b

Relative
pre-miR-30b

premiR-30b

50

1.0
0.8

0.6
0.4
0.2
0

DG IgG
C
DG R8
C
R8

80

primiR-30b

Fold change

nt:
1,000
500
300
150

Ig

IPs:

G
PL
E
D G KH
C A7
R
Ig 8
G
PL
EK
DG HA
C 7
R8

Low exposure

Ig
G
DG
C
DG R8
C
R8

21

21

NT PLEKHA7
shRNA

pri-miR-30b
Ecad
DAPI !

Apical
ZA

ZA PLEKHA7
DROSHADGCR8 pri-miR-30b
p120Ecad
PPIB
Ecad
DAPI

Lateral

DapB
Ecad
DAPI

p120Ecad
p120Cad11

miR-30b

p-p120

SNAI1
MYC
CCND1

p-Src
Negative cont.

Positive cont.

AIG

p130CAS
p190RhoGAP
Basal

Figure 8 PLEKHA7 regulates pri-miR-30b processing at the junctions.


(a) qRTPCR of isolated nuclear and non-nuclear subcellular fractions
of Caco2 cells for the indicated pri-miRNAs (right panel). Successful
fractionation is demonstrated by western blot for the indicated markers
(left panel). Ct values are shown (mean s.d. from n = 3 independent
experiments); actin (ACTB) is the positive qRTPCR control. Lamin A/C and
histone H3 are the nuclear and GAPDH the cytoplasmic markers for the
western blot. See Supplementary Fig. 8 for unprocessed blot scans. (b) qRT
PCR of the eluates from the IgG (negative control), PLEKHA7 and DGCR8
(positive control) RNA-IPs for the indicated pri-miRNAs (a representative
is shown from two independent experiments). (c) In vitro pri-miR-30b
processing activity assay of Caco2 lysates for PLEKHA7, DGCR8 (positive
control) and IgG (negative control) IPs. A lower exposure image of the pri-miR30b template is presented to show the actual loading amount. nt, nucleotides.
(d) In vitro pri-miR-30b processing activity assay for DGCR8 IPs of NT or

PLEKHA7 shRNA Caco2 cells; IgG is the negative control. (e) Quantification
of pre-miR-30b band intensities from n = 3 independent in vitro processing
activity assays, carried out as in d (mean s.d.; P < 0.001, Students twotailed t-test). (f) ISH of Caco2 cells for pri-miR-30b; a bacterial DapB RNA
probe was used as the negative and the endoplasmic-reticulum-specific PPIB
as the positive control. Merged images are shown. An enlarged area in the
yellow box in the top right panel is shown for the pri-miR-30b ISH. Arrows
indicate hybridization signals. Scale bars for full images, 20 m; for enlarged
part, 12 m. Source data are provided in Supplementary Table 2 for a, b and
e. (g) Schematic diagram summarizing the findings of the present study. Two
distinct junctional complexes exist in polarized epithelial cells: a basolateral
complex lacking PLEKHA7 promotes growth signalling and anchorageindependent growth (AIG) through cadherins, Src and p120 activity, whereas
an apical ZA complex suppresses these events through PLEKHA7, by locally
recruiting the microprocessor at the ZA to regulate processing of miR-30b.

NATURE CELL BIOLOGY ADVANCE ONLINE PUBLICATION

2015 Macmillan Publishers Limited. All rights reserved

11

A RT I C L E S
The miRNAs regulated by PLEKHA7 provide a mechanistic
link to the essential function of the ZA in maintaining epithelial
homeostasis. The miR-30 and let-7 families are well established
guardians of the epithelial phenotype and epithelialmesenchymal
transition suppressors4244. In contrast, the most strongly increased
miR-19a after PLEKHA7 depletion is a key component of the
polycistronic miR-17-92 miRNA oncogene52,53. Our data support a
model whereby individual cells within an epithelial monolayer sense
the status of their apical ZA by regulating the levels of a subset of
miRNAs to accordingly suppress or promote growth (Fig. 8g).
Co-localization of phosphorylated p120 with activated Src and
Rac1 was observed specifically at the basolateral complex, similar
to the case of p190RhoGAP, which suppresses RhoA signalling and
induces transformed cell growth in a Rac1- and Src-dependent
manner7,13 (Figs 1b,d,g,h, 2d, and Supplementary Fig. 1e,f,h,i).
Therefore, it is the basolateral complex that exerts the previously
described Src- and Rac1-induced transforming activity of p120
(refs 13,15). In agreement, we show that E-cadherin, Src activation
and p120 tyrosine phosphorylation are required for increased growth
signalling in the absence of PLEKHA7 (Fig. 4be). Therefore,
the growth-suppressing or growth-promoting functions of p120
and E-cadherin are fine-tuned by binding to PLEKHA7. Further
interactions of PLEKHA7 with paracingulin54, afadin55, ZO-1 and
cingulin56, may also play a role in PLEKHA7 function, although the
interaction with p120 is essential for modulating cell behaviour by
regulating miRNA processing.
Taken together, our data untangle the complicated roles of
E-cadherin and p120 in the context of distinct junctional complexes,
spatially separating their functions and providing an explanation for
their conflicting behaviour in cell growth. In addition, they identify
PLEKHA7 as a specific marker of ZA that mediates suppression of
growth-related signalling. Finally, they reveal an interaction of the ZA
with the microprocessor complex, and uncover a mechanism whereby
the ZA regulates a set of miRNAs to suppress cellular transformation
and maintain the epithelial phenotype.

METHODS
Methods and any associated references are available in the online
version of the paper.
Note: Supplementary Information is available in the online version of the paper
ACKNOWLEDGEMENTS
This work was supported by NIH R01 CA100467, R01 NS069753, P50 CA116201
(P.Z.A.); NIH R01 GM086435, Florida Department of Health, Bankhead-Coley
10BG11 (P.S.); NIH/NCI R01CA104505, R01CA136665 (J.A.C.); BCRF (E.A.P.); the
Swiss Cancer League (S.C., Project KLS-2878-02-2012). A.K. is supported by the Jay
and Deanie Stein Career Development Award for Cancer Research at Mayo Clinic.
We thank Mayo Clinics Proteomics Core and B. Madden for assistance with mass
spectrometry, B. Edenfield for immunohistochemistry, M. Takeichi, D. Radisky and
M. Cichon for constructs, and D. Radisky, L. Lewis-Tuffin, J. C. Dachsel, B. Necela
and the late G. Hayes for suggestions and comments.
AUTHOR CONTRIBUTIONS
A.K. designed the study, conceived and designed experiments, carried out all
experiments except those described below, analysed the data and wrote the
manuscript. S.P.N. made constructs. P.P. and S.C. developed antibodies and
constructs. R.W.F. provided technical support. L.R.C. assisted with IF. T.R.B., J.M.C.
and E.A.T. carried out the NanoString experiment and assisted with qRTPCRs.
I.K.Y. and T.P. assisted with the ISH assay. E.A.P., P.S., S.B. and J.A.C. developed and
provided tissue micro-arrays. P.Z.A. conceived and designed the study, conceived
and designed experiments and wrote the manuscript.

12

COMPETING FINANCIAL INTERESTS


The authors declare no competing financial interests.
Published online at http://dx.doi.org/10.1038/ncb3227
Reprints and permissions information is available online at www.nature.com/reprints

1. Reynolds, A. B., Roesel, D. J., Kanner, S. B. & Parsons, J. T. Transformationspecific tyrosine phosphorylation of a novel cellular protein in chicken cells
expressing oncogenic variants of the avian cellular src gene. Mol. Cell. Biol. 9,
629638 (1989).
2. Ireton, R. C. et al. A novel role for p120 catenin in E-cadherin function. J. Cell Biol.
159, 465476 (2002).
3. Ishiyama, N. et al. Dynamic and static interactions between p120 catenin and Ecadherin regulate the stability of cellcell adhesion. Cell 141, 117128 (2010).
4. Yap, A. S., Niessen, C. M. & Gumbiner, B. M. The juxtamembrane region of the
cadherin cytoplasmic tail supports lateral clustering, adhesive strengthening, and
interaction with p120ctn. J. Cell Biol. 141, 779789 (1998).
5. Davis, M. A., Ireton, R. C. & Reynolds, A. B. A core function for p120-catenin in
cadherin turnover. J. Cell Biol. 163, 525534 (2003).
6. Anastasiadis, P. Z. p120-ctn: A nexus for contextual signaling via Rho GTPases.
Biochim. Biophys. Acta 1773, 3446 (2007).
7. Wildenberg, G. A. et al. p120-catenin and p190RhoGAP regulate cellcell adhesion
by coordinating antagonism between Rac and Rho. Cell 127, 10271039 (2006).
8. Smith, A. L., Dohn, M. R., Brown, M. V. & Reynolds, A. B. Association of Rhoassociated protein kinase 1 with E-cadherin complexes is mediated by p120-catenin.
Mol. Biol. Cell 23, 99110 (2012).
9. Schackmann, R. C. et al. Cytosolic p120-catenin regulates growth of metastatic
lobular carcinoma through Rock1-mediated anoikis resistance. J. Clin. Invest. 121,
31763188 (2011).
10. Stairs, D. B. et al. Deletion of p120-catenin results in a tumor microenvironment
with inflammation and cancer that establishes it as a tumor suppressor gene. Cancer
Cell 19, 470483 (2011).
11. Mariner, D. J., Davis, M. A. & Reynolds, A. B. EGFR signaling to p120-catenin through
phosphorylation at Y228. J. Cell Sci. 117, 13391350 (2004).
12. Johnson, E. et al. HER2/ErbB2-induced breast cancer cell migration and invasion
require p120 catenin activation of Rac1 and Cdc42. J. Biol. Chem. 285,
2949129501 (2010).
13. Dohn, M. R., Brown, M. V. & Reynolds, A. B. An essential role for p120-catenin
in Src- and Rac1-mediated anchorage-independent cell growth. J. Cell Biol. 184,
437450 (2009).
14. Yanagisawa, M. & Anastasiadis, P. Z. p120 catenin is essential for mesenchymal
cadherin-mediated regulation of cell motility and invasiveness. J. Cell Biol. 174,
10871096 (2006).
15. Soto, E. et al. p120 catenin induces opposing effects on tumor cell growth depending
on E-cadherin expression. J. Cell Biol. 183, 737749 (2008).
16. Silvera, D. et al. Essential role for eIF4GI overexpression in the pathogenesis of
inflammatory breast cancer. Nat. Cell Biol. 11, 903908 (2009).
17. Liu, W. F., Nelson, C. M., Pirone, D. M. & Chen, C. S. E-cadherin engagement
stimulates proliferation via Rac1. J. Cell Biol. 173, 431441 (2006).
18. Lewis-Tuffin, L. J. et al. Misregulated E-cadherin expression associated with an
aggressive brain tumor phenotype. PLoS ONE 5, e13665 (2010).
19. Rodriguez, F. J., Lewis-Tuffin, L. J. & Anastasiadis, P. Z. E-cadherins dark side:
Possible role in tumor progression. Biochim. Biophys. Acta 1826, 2331 (2012).
20. Kuphal, S. & Bosserhoff, A. K. E-cadherin cellcell communication in melanogenesis
and during development of malignant melanoma. Arch. Biochem. Biophys. 524,
4347 (2012).
21. Meng, W., Mushika, Y., Ichii, T. & Takeichi, M. Anchorage of microtubule minus
ends to adherens junctions regulates epithelial cellcell contacts. Cell 135,
948959 (2008).
22. Pulimeno, P., Bauer, C., Stutz, J. & Citi, S. PLEKHA7 is an adherens junction
protein with a tissue distribution and subcellular localization distinct from ZO-1 and
E-cadherin. PLoS ONE 5, e12207 (2010).
23. Miyoshi, J. & Takai, Y. Structural and functional associations of apical junctions with
cytoskeleton. Biochim. Biophys. Acta 1778, 670691 (2008).
24. Smutny, M. et al. Myosin II isoforms identify distinct functional modules that support
integrity of the epithelial zonula adherens. Nat. Cell Biol. 12, 696702 (2010).
25. Mariner, D. J. et al. Identification of Src phosphorylation sites in the catenin p120ctn.
J. Biol. Chem. 276, 28006-28013 (2001).
26. Ma, L. W., Zhou, Z. T., He, Q. B. & Jiang, W. W. Phosphorylated p120-catenin
expression has predictive value for oral cancer progression. J. Clin. Pathol. 65,
315319 (2012).
27. Kourtidis, A., Ngok, S. P. & Anastasiadis, P. Z. p120 catenin: an essential regulator
of cadherin stability, adhesion-induced signaling, and cancer progression. Prog. Mol.
Biol. Transl. Sci. 116, 409432 (2013).
28. Tikhmyanova, N. & Golemis, E. A. NEDD9 and BCAR1 negatively regulate E-cadherin
membrane localization, and promote E-cadherin degradation. PLoS ONE 6,
e22102 (2011).
29. Chang, J. H., Gill, S., Settleman, J. & Parsons, S. J. c-Src regulates the simultaneous
rearrangement of actin cytoskeleton, p190RhoGAP, and p120RasGAP following
epidermal growth factor stimulation. J. Cell Biol. 130, 355368 (1995).

NATURE CELL BIOLOGY ADVANCE ONLINE PUBLICATION

2015 Macmillan Publishers Limited. All rights reserved

A RT I C L E S
30. Wang, Y. et al. Synergistic effect of cyclin D1 and c-Myc leads to more aggressive and
invasive mammary tumors in severe combined immunodeficient mice. Cancer Res.
67, 36983707 (2007).
31. Eiseler, T. et al. Protein kinase D1 mediates anchorage-dependent and -independent
growth of tumor cells via the zinc finger transcription factor Snail1. J. Biol. Chem.
287, 3236732380 (2012).
32. Thoreson, M. A. et al. Selective uncoupling of p120(ctn) from E-cadherin disrupts
strong adhesion. J. Cell Biol. 148, 189-202 (2000).
33. Ozawa, M. & Ohkubo, T. Tyrosine phosphorylation of p120(ctn) in v-Src transfected
L cells depends on its association with E-cadherin and reduces adhesion activity.
J. Cell Sci. 114, 503512 (2001).
34. Zhou, B. P. et al. Dual regulation of Snail by GSK-3-mediated phosphorylation in
control of epithelial-mesenchymal transition. Nat. Cell Biol. 6, 931940 (2004).
35. Yook, J. I. et al. A Wnt-Axin2-GSK3 cascade regulates Snail1 activity in breast
cancer cells. Nat. Cell Biol. 8, 1398-1406 (2006).
36. Fabian, M. R., Sonenberg, N. & Filipowicz, W. Regulation of mRNA translation and
stability by microRNAs. Annu. Rev. Biochem. 79, 351379 (2010).
37. Krol, J., Loedige, I. & Filipowicz, W. The widespread regulation of microRNA
biogenesis, function and decay. Nat. Rev. Genet. 11, 597610 (2010).
38. Meijer, H. A. et al. Translational repression and eIF4A2 activity are critical for
microRNA-mediated gene regulation. Science 340, 8285 (2013).
39. Pillai, R. S. et al. Inhibition of translational initiation by Let-7 MicroRNA in human
cells. Science 309, 15731576 (2005).
40. Croce, C. M. Causes and consequences of microRNA dysregulation in cancer. Nat.
Rev. Genet. 10, 704714 (2009).
41. Adams, B. D., Kasinski, A. L. & Slack, F. J. Aberrant regulation and function of
MicroRNAs in cancer. Curr. Biol. 24, R762-R776 (2014).
42. Joglekar, M. V. et al. The miR-30 family microRNAs confer epithelial phenotype to
human pancreatic cells. Islets 1, 137147 (2009).
43. Watanabe, S. et al. HMGA2 maintains oncogenic RAS-induced epithelialmesenchymal transition in human pancreatic cancer cells. Am. J. Pathol. 174,
854868 (2009).

44. Zhang, J. et al. miR-30 inhibits TGF-1-induced epithelial-to-mesenchymal


transition in hepatocyte by targeting Snail1. Biochem. Biophys. Res. Commun. 417,
11001105 (2012).
45. Buechner, J. et al. Tumour-suppressor microRNAs let-7 and mir-101 target the protooncogene MYCN and inhibit cell proliferation in MYCN-amplified neuroblastoma. Br.
J. Cancer 105, 296303 (2011).
46. Lan, F. F. et al. Hsa-let-7g inhibits proliferation of hepatocellular carcinoma cells
by downregulation of c-Myc and upregulation of p16(INK4A). Int. J. Cancer 128,
319331 (2011).
47. Lal, A. et al. miR-24 Inhibits cell proliferation by targeting E2F2, MYC, and other
cell-cycle genes via binding to "seedless" 30 UTR microRNA recognition elements.
Mol. Cell 35, 610625 (2009).
48. Ha, M. & Kim, V. N. Regulation of microRNA biogenesis. Nat. Rev. Mol. Cell. Biol.
15, 509524 (2014).
49. Gregory, R. I. et al. The microprocessor complex mediates the genesis of microRNAs.
Nature 432, 235240 (2004).
50. Lee, Y. et al. The nuclear RNase III Drosha initiates microRNA processing. Nature
425, 415419 (2003).
51. Han, J. et al. The Drosha-DGCR8 complex in primary microRNA processing. Genes
Dev. 18, 30163027 (2004).
52. He, L. et al. A microRNA polycistron as a potential human oncogene. Nature 435,
828833 (2005).
53. Olive, V. et al. miR-19 is a key oncogenic component of mir-17-92. Genes Dev. 23,
28392849 (2009).
54. Pulimeno, P., Paschoud, S. & Citi, S. A role for ZO-1 and PLEKHA7 in recruiting
paracingulin to tight and adherens junctions of epithelial cells. J. Biol. Chem. 286,
1674316750 (2011).
55. Kurita, S., Yamada, T., Rikitsu, E., Ikeda, W. & Takai, Y. Binding between the
junctional proteins afadin and PLEKHA7 and implication in the formation of adherens
junction in epithelial cells. J. Biol. Chem. 288, 2935629368 (2013).
56. Paschoud, S., Jond, L., Guerrera, D. & Citi, S. PLEKHA7 modulates epithelial tight
junction barrier function. Tissue Barriers 2, e28755 (2014).

NATURE CELL BIOLOGY ADVANCE ONLINE PUBLICATION

2015 Macmillan Publishers Limited. All rights reserved

13

METHODS

DOI: 10.1038/ncb3227

METHODS
Cell culture, reagents and chemicals. In all comparisons, cells were used strictly at
the same confluences. All cell lines were obtained from ATCC, used at low passage
(<20), and tested negative for mycoplasma contamination. Caco2 colon epithelial
cells were cultured in MEM (Cellgro) supplemented with 10% FBS (Invitrogen),
1 mM sodium pyruvate (Invitrogen) and 1 non-essential amino-acid supplement
(Mediatech). MDCK canine kidney epithelial cells, HEK 293FT human embryonic
kidney cells, and Phoenix-Ampho cells were cultured in DMEM supplemented
with heat-inactivated 10% FBS. PP2 was obtained from Calbiochem; nocodazole,
cycloheximide and MG-132 from Sigma.

Constructs. Full-length human PLEKHA7 was cloned in the XbaIHindII sites


of pcDNA3.1mycHis and then the PLEKHA7Myc fragment was cut out by
PmeI and sub-cloned to the AfeI site of the retroviral LZRS vector to obtain the
LZRS-PLEKHA7Myc construct. The PLEKHA7GFP construct was a gift of the
M. Takeichi laboratory (RIKEN, Japan; ref. 21). The mp120-1A, mp120-4A, mp1208F and mp120-1ARM1 constructs have been previously described2,25. The adSNAI1
and adGFP adenoviruses were a gift of the D. Radisky laboratory (Mayo Clinic, FL,
USA). The YFPPBD (plasmid 11407) and GFPrGBD (plasmid 26732) constructs
were obtained from Addgene.

shRNAs, siRNAs, anti-miRNAs and miR-mimics. Cells were transfected using


Lipofectamine 2000 (Invitrogen) or Lipofectamine RNAiMAX (Invitrogen) for
anti-miRNA, miR-mimic and siRNA transfection, according to the manufacturers
protocols. Lentiviral shRNAs were derived from the pLKO.1-based TRC1
(SigmaRNAi Consortium) shRNA library (pLKO.1-puro non-target shRNA
control, SHC016; PLEKHA7 no. 8, TRCN0000146289; PLEKHA7 no. 10,
TRCN0000127584; Cad11, TRCN0000054334; Ecad no. 21, TRCN0000039664;
Ecad no. 23, TRCN0000039667) and the pLKO.5-based TRC2 library (pLKO.5-puro
non-target shRNA control, SHC216; NEZHA no. 72, TRCN0000268676; NEZHA,
no. 73: TRCN0000283758). Lentiviruses were produced in HEK 293FT cells and
used to infect cells according to standard protocols. Retroviruses were prepared in
Phoenix-Ampho cells, as described previously15. SMARTpool (Dharmacon) siRNAs
used: DROSHA, M-016996-02-0005; DGCR8, M-015713-01-0005; non-target,
D-001206-14-05 mirVana (Life Technologies catalogue no. 4464084). Anti-miRNAs
used: anti-hsa-miR-30a, ID MH11062; anti-hsa-let-7g, ID MH11758; anti-hsamiR-30b, ID MH10986; anti-hsa-miR-24, ID MH10737; negative control no. 1,
catalogue no. 4464076. MISSION (Sigma) miR-mimics used: miR-30b, catalogue
no. HMI0456; negative control 1 (catalogue no. HMC0002).

were removed and tissue samples were de-identified. The Mayo Clinic Institutional
Review Board assessed the protocol 09-001642 as minimal risk and waived the
need for further consent. Renal tissue samples were collected for the Mayo Clinic
Renal Tissue Registry under protocol 14-03, with the approval of the Mayo Clinic
Institutional Review Board. Written informed consent for the use of these tissues
in research was obtained from all participants to the Registry. Renal tissue samples
used here were obtained from the Renal Tissue Registry under the Mayo Clinic
Institutional Review Board protocol 675-05. All unique patient identifiers and
confidential data were removed and tissue samples were de-identified. The Mayo
Clinic Institutional Review Board assessed the 675-05 protocol as minimal risk
and waived the need for further consent. All data were analysed anonymously.
Paraffin-embedded tissues on slides were placed in xylene and rehydrated in a
graded ethanol series, rinsed in water, subjected to heat antigen retrieval according
to the manufacturers instructions (Dako), incubated with primary antibody and
finally with either anti-rabbit (for PLEKHA7) or anti-mouse (for Ecad, p120)
labelled polymer horseradish peroxidase (Dako). Slides were scanned using Aperio
ScanScope XT and viewed using Aperio ImageScope v11.1.2.752.

Junction resistance measurements. ECIS Z (Electric Cell-substrate Impedance


System, Applied Biophysics) was used to measure impedance of Caco2 cells. 4 104
cells were plated on electrode arrays (8W10E, Applied Biophysics). Measurements
were made every 180 s at the 4,000 Hz frequency that measures junctional impedance
and at 64,000 Hz that corresponds to cell density. The 4,000 Hz values were
normalized to the 64,000 Hz values to account for cell density variations.

Soft agar assay. Six-well plates (Costar 3516) were first covered with a layer of
0.75% agarose, prepared by mixing 1.5% of sterile agarose (Seakem) in a 1:1 ratio
with 2 concentrated Caco2 culture medium made from powder MEM (Invitrogen,
catalogue no. 61100061) and double addition of each supplement (see above). The
basal layer was left to solidify for 30 min. Cells were trypsinized, counted and
resuspended in 2 Caco2 medium. The cell suspension was mixed in a 1:1 ratio
with 0.7% sterile agarose to make a final layer of 0.35% agarose, which was added
on top of the basal layer. 5 104 Caco2 cells were seeded per well. The top layer was
left to solidify for another 30 min and was covered with 1 Caco2 culture medium.
Cultures were maintained for 4 weeks with medium renewal on top of agar every
three days until colonies were visible, and then stained with 0.02% crystal violet
(Fisher) in 20% ethanol and PBS, washed three times with distilled H2 O, scanned
and counted for colonies.

Calcium switch assay. Caco2 cells were grown on coverslips until confluency, preAntibodies. For detailed information on all the antibodies used in the study, as well
as working dilutions for each assay, see Supplementary Table 3.

Immunofluorescence. MDCK and Caco2 cells were grown on Transwell inserts


(Costar 3413) for 7 or 21 days respectively, until they polarized, or on sterile glass
coverslips until they reached full confluence. Cells were washed once with PBS
and then fixed with either 100% methanol (Fisher) for 7 min; or 4% formaldehyde
(EMS) for 20 min, followed by 0.02% Triton X-100 permeabilization for 10 min;
or 10% TCA (Sigma) for 15 min on ice, particularly for RhoA co-staining, and
permeabilized as above. Cells were blocked with either 3% non-fat milk (Carnation)
in PBS or Protein-Block reagent (Dako, X090930-2) for 30 min and stained with
primary antibodies diluted either in milk or antibody diluent (Dako, S302281-2)
for 1 h. Cells were then washed three times with PBS, stained with the fluorescentlabelled secondary antibodies for 1 h, washed three times with PBS, co-stained with
DAPI (Sigma) to visualize the nuclei, mounted (Aqua Poly/Mount, Polysciences)
and imaged using a Zeiss LSM 510 META laser confocal microscope, under a 63
objective, with a further 1.6 zoom. z-stacks were acquired at 0.5 m intervals.
Images, stacks and xz representations of the image stacks were processed using
the Zen software (Zeiss).

Immunoblotting. Whole-cell extracts were obtained using RIPA buffer (Tris,


pH 7.4, 50 mM NaCl, 150 mM, 1% NP-40, 0.5% deoxycholic acid, 0.1% SDS) supplemented with protease (Cocktail III, RPI) and phosphatase inhibitors (Pierce). Lysates
were homogenized through a 29 g needle and cleared by full-speed centrifugation
for 5 min. Protein extracts were mixed with LSB and separated by SDSPAGE,
transferred to nitrocellulose membranes (Bio-Rad), blotted according to standard
protocols, detected by luminescence using ECL (GE Healthcare) and imaged using
X-ray films (Pierce). See Supplementary Fig. 8 for original scans of western blots.

Immunohistochemistryethics statements. Breast tissue samples were initially


collected under protocol MC0033, with the approval of the Mayo Clinic Institutional
Review Board. Written informed consent for the use of these tissues in research was
obtained from all participants. Generation of the tissue micro-arrays was carried
out under protocol 09-001642. All unique patient identifiers and confidential data

treated with either DMSO (control) or 10 M nocodazole for 1 h to dissolve microtubules, then washed three times with calcium-free PBS and incubated in calciumfree Caco2 medium (Life, 11380-037, supplemented with glutamine, 10%FBS,
sodium pyruvate and MEM) containing 4 mM EGTA for 30 min, until cells were
rounded, while being kept in either DMSO or 10 M nocodazole, respectively. Cells
were then washed three times with PBS, returned to regular Caco2 medium, again
with DMSO or nocodazole respectively, and fixed for IF, for the indicated times.

IPs. Cells were grown on 10 cm plates until fully confluent, washed twice with PBS
and lysed using a Triton X-100 lysis buffer (150 mM NaCl, 1 mM EDTA, 50 mM
Tris pH 7.4, 1% Triton X-100) containing twice the amount of protease (Cocktail III,
RPI) and phosphatase inhibitors (Pierce). Four 10 cm plates (34 106 cells) were
used per IP. In parallel, 4 g of antibody or IgG control (Jackson ImmunoResearch,
011-000-003) was incubated with 40 l Protein G Dynabeads (Invitrogen) overnight
and then washed three times with IP lysis buffer. Cell lysates were incubated with
the bead-conjugated antibodies overnight. Beads were then washed three times with
IP lysis buffer and eluted using 50 mM dithiothreitol (Sigma) in lysis buffer at 37 C
for 45 min, with constant agitation. For RNAse-treated IPs, beads were distributed
equally after the final wash into two tubes with either 1.5 ml PBS or PBS containing
100 mg ml1 RNAse A (Sigma). After incubation at 4 C for 1.5 h, beads were washed
three times with PBS, and proteins were eluted as above.

Proteomics. Caco2 cells were grown on 10 cm Transwell inserts (Costar 3419)


for 21 days to polarize. Cells were then washed twice with PBS and proteins were
cross linked as previously described57, using 0.75 mM of the reversible cross linker
DSP (Lomants reagent; Pierce) for 3 min at room temperature, which was then
neutralized using 20 mM Tris pH 7.5 for 15 min. Cells were finally lysed using
RIPA (see recipe above, immunoblotting section) containing twice the amount of
protease (Cocktail III, RPI) and phosphatase inhibitors (Pierce) and 1 mM EDTA.
Four 10 cm Transwells (34 106 cells) were used per IP. In parallel, 4 g of
antibody or IgG control (Jackson ImmunoResearch, 011-000-003) was incubated
with 40 l protein G Dynabeads (Invitrogen) overnight and cross linked to the beads
using 5 mM BS (ref. 3; Pierce) according to the manufacturers protocol. Cross linked
lysates were incubated with the bead-conjugated antibodies overnight. Beads were

NATURE CELL BIOLOGY

2015 Macmillan Publishers Limited. All rights reserved

METHODS

DOI: 10.1038/ncb3227
then washed three times with lysis buffer and proteins were eluted as described
above and separated by SDSPAGE. Gels were silver-stained (Pierce SilverSnap kit),
according to the manufacturers protocol. Gel slices were selected and excised using
sterile scalpels and destained, reduced and alkylated before digestion with trypsin
(Promega). Samples were analysed using nanoHPLC-electrospray tandem mass
spectrometry (nanoLC-ESI-MS/MS) in a ThermoFinnigan LTQ Orbitrap hybrid
mass spectrometer (ThermoElectron Bremen). Mascot (Matrix Sciences London)
was used to search the Swissprot database to identify isolated peptides. Common
contaminants such as trypsin, casein, keratin and microbial-specific proteins were
removed from analysis. Results were visualized using Cytoscape 2.8.2 (ref. 58).

Total RNA isolation and qRTPCR. Cells were lysed using Trizol (Invitrogen) and
subjected to the Trizol Plus total transcriptome isolation protocol of the PureLink
RNA mini kit (AmbionLife Technologies) specified to isolate both mRNAs and
miRNAs. Final RNA concentrations were determined using a NanoDrop spectrophotometer. RNA was converted to complementary DNA using a high capacity
cDNA reverse transcriptase kit (Applied Biosystems). qPCR reactions were carried
out using the TaqMan FAST Universal PCR master mix (Applied Biosystems), in a
7900 HT or ViiA 7 thermocycler (Applied Biosystems). Data were analysed using RQ
Manager (Applied Biosystems). U6 was used as a control for miRNA expression normalization and GAPDH, -actin for mRNA and pri/pre-miRNA normalization. TaqMan assays used for miRNAs (Applied Biosystems, catalogue no. 4427975): hsa-miR24, 000402; hsa-miR-30a, 000417; hsa-miR-30b, 000602; hsa-let-7g, 002282; hsamiR-19a, 000395; U6, 001973. TaqMan assays for miRNA precursors: pri-miR-30b,
Applied Biosystems, catalogue no. 4427012, Hs03303066_pri; pre-miR-30b, Custom
Plus TaqMan RNA assay catalogue no. 4441114, assay ID AJN1E56, context sequence 50 -CTGTAATACATGGATTGGCTGGGAG-30 . TaqMan assays for mRNAs
(Applied Biosystems, catalogue no. 4331182): PLEKHA7, Hs00697762_m1; SNAI1,
Hs00195591_m1; MYC, Hs00153408_m1; CCND1, Hs00277039_m1; GAPDH,
Hs99999905_m1; actin (ACTB), Hs99999903_m1.
RNA-IPs. RNA-IPs were carried out as described above for standard IPs, but
with the addition of 100 U ml1 RNAse inhibitor (Promega) in the lysis buffer.
After the final wash, RNA was eluted from the beads using Trizol (Invitrogen)
and purified as described above. qRTPCRs were then carried out as above and
the results were analysed using the Sigma RIP-qRTPCR data analysis calculation
shell, associated with the Sigma Imprint RIP kit (http://www.sigmaaldrich.com/
life-science/epigenetics/imprint-rna.html and references therein).

Cell fractionation. To separate the nuclear fraction, cells were first lysed using a
buffer containing 25 mM Tris pH 7.4, 150 mM NaCl, 5 mM MgCl2 , 0.5% NP-40,
protease (Cocktail III, RPI), phosphatase (Pierce) and RNAse (100 U ml1 ; Promega)
inhibitors for 10 min on ice. The lysates were centrifuged at 300g for 5 min at 4 C
to pellet the nuclei and the supernatant was carefully transferred to a fresh tube.
The nuclear pellet was lysed using RIPA (see recipe above, immunoblotting section)
with protease, phosphatase and RNAse inhibitors for 10 min on ice, homogenized
through a 29 g needle and cleared by centrifugation for 5 min at 12,000g . Half of
each lysate was used for SDSPAGE, whereas the other half was mixed with Trizol
and used to extract total RNA and cary out qRTPCRs, as described above.

Proximity ligation. The Duolink In Situ Red Starter Kit Mouse/Rabbit (Sigma) was
used according to the manufacturers instructions.
In vitro pri-miRNA processing assay. Total cDNA from Caco2 cells was obtained
as described above (qRTPCR section) and was used to amplify the pri-miR30b transcript. The pri-miR-30b cDNA was then subcloned into the pBluescript
SK+ vector, downstream of the T7 promoter, between the EcoRI and XhoI sites.
Primers used: pri-30bF-XhoI, 50 -ATTAACTCGAGGTGAATGCTGTGCCTGTT
C-30 ; pri-30bR-EcoRI, 50 -ACGTTGAATTCGCCTCTGTATACTATTCTTGC-30 .
The cloned pri-miR-30b sequence is as follows (the pre-mRNA sequence is shown
in capital letters): 50 -gtgaatgctgtgcctgttctttttttcaacagagtcttacgtaaagaaccgtacaaactta
gtaaagagtttaagtcctgctttaaACCAAGTTTCAGTTCATGTAAACATCCTACACTC
AGCTGTAATACATGGATTGGCTGGGAGGTGGATGTTTACTTCAGCTGAC
TTGGAatgtcaaccaattaacattgataaaagatttggcaagaatagtatacagaggc-30 . The pri-miR30b construct was linearized using SmaI and in vitro transcription was carried out
using T7 polymerase (Roche) and fluorescein isothiocyanate (FITC) RNA labelling
mix (Roche), according to the manufacturers instructions. The FITC-labelled primiR-30b was gel-purified and stored at 20 C in the dark for no more than a
week. For the in vitro processing assay, IPs on Caco2 lysates were carried out as
described above using IgG (negative control), a PLEKHA7 (Sigma) or a DGCR8
(Sigma) (positive control) antibody. For the PLEKHA7 knockdown experiment,
control (NT) or PLEKHA7 shRNA cells were used and IPs were carried out using
an IgG (negative control), or a DGCR8 (Sigma) antibody. After the final wash, the
beads were mixed with a reaction mixture containing 20 mM Tris at pH 8.0, 100 mM

KCl, 7 mM MgCl2 , 20 mM creatine phosphate, 1 mM ATP, 0.2 mM phenylmethyl


sulphonyl fluoride, 5 mM dithiothreitol, 10% glycerol, 2 U l1 RNAse inhibitor and
3 g FITC-labelled pri-miR-30b. The labelled probe was denatured for 2 min at 95 C
and left to gradually cool down and refold before the assay. The processing assay
was carried out for 90 min at 37 C in the dark with constant gentle rocking. RNA
was eluted with RNA elution buffer (2% SDS, 0.3 M NaOAc pH 5.2) purified with
phenolchloroformIAA (25:24:1) pH 5.5 (Fisher), and precipitated overnight at
80 C using NaOAc 0.3 M pH 5.2, 1 l glycogen (Invitrogen) and 4:1 100% EtOH.
The RNA was recovered by full-speed centrifugation for 30 min at 4 C, washed with
75% EtOH, resuspended in 7.5 l DEPCH2 O, denatured in 1:1 Gel Loading Buffer
II (Life Technologies) for 5 min at 95 C, and loaded onto a 12% denaturing PAGE
RNA gel (SequaGel; National Diagnostics). The gel was scanned using a Typhoon
9400 (GE Healthcare) and quantification of bands was carried out using ImageJ.

ISH. For ISH of pri-miR-30b, Caco2 cells were grown on slides, fixed with 10%
formalin (Protocol; Fisher), dehydrated and rehydrated in a series of 5070
1007050% EtOH solutions, pretreated with protease and hybridized using the
RNAscope 2.0 HD Brown assay (Advanced Cell Diagnostics, ACD), according to
the manufacturers instructions. Probes used: human pri-miR-30b, ACD, 415331;
positive control probe Hs-PPIB, ACD, 313906; negative control probe DapB, ACD,
310048. After ISH, cells were rinsed twice with PBS and blocked with ProteinBlock reagent (Dako, X090930-2) for 10 min, and IF was carried out with an Ecad
antibody (BD) diluted in antibody diluent (Dako, S302281-2) for 30 min; Ecad was
the junctional marker used here because it resisted protease treatment and did not
produce background due to formalin fixation. Cells were then washed three times
with PBS and stained with a fluorescent-labelled secondary antibody and DAPI for
30 min. Cells were finally washed three times with PBS, mounted (Aqua Poly/Mount,
Polysciences) and imaged using a Leica DM5000B microscope under bright (ISH)
and fluorescent light (protein IF). All images were pseudocoloured and merged
images were created using the microscopes Leica suite software.

NanoString miRNA analysis. Total RNA was isolated as above and global miRNA
analysis was carried out using the nCounter Human v2 miRNA expression assay
(NanoString) containing 735 miRNAs. Data were collected using the nCounter
Digital Analyzer (NanoString). miRNAs were first elongated by a sequence-specific
ligation and were then hybridized with specific, barcoded probes. The raw units
that result from the NanoString reaction reflect the number of times each probe is
counted in a sample of the total hybridization reaction and represent the frequency
of each miRNA, without amplification of starting material. The raw units were then
normalized for RNA input and background noise to internal positive and negative
controls according to the manufacturers instructions. miRNAs with low counts at
the level of the negative controls were excluded.
Northern blot. 1020 g of total RNA or the Low Range ssRNA Ladder (NEB)
were mixed in a 1:1 volume ratio with Gel Loading Buffer II (Life Technologies),
denatured at 95 C for 5 min and loaded onto 12% denaturing PAGE RNA
gels (SequaGel; National Diagnostics). RNAs were then transferred to Hybond
N+ membranes (Amersham, GE Healthcare) for 2 h at 250 mA using a semidry apparatus (Hoefer TE70) and cross linked using a Stratalinker (Stratagene).
Northern blot was carried out using the DIG Northern Starter Kit (Roche) and the
DIG Wash and Block Buffer Set (Roche) according to the manufacturers protocol.
Hybridization and stringent washes were carried out at 50 C; probes were used
at 5 nM final concentration. Probes used: hsa-miR-30b miRCURY LNA Detection
probe, DIG labelled, Exiqon, 18143-15; U6 hsa/mmu/rno control, miRCURY LNA
detection probe, DIG Labelled, Exiqon, 99002-01.
Statistics and reproducibility. For all quantitative experiments, averages and s.d.
were calculated and presented as error bars, whereas the number of independent
experiments carried out and the related statistics are indicated in each figure
legend. Students two-tailed t-test was employed for P-value calculations because
all comparisons were between two groups, control and experimental condition. For
all other experiments, at least three independent experiments were carried out and a
representative is shown in Figs 1ah, 2b,df, 3b (left panel), 3dg, 4bg, 5c,d, 6bk,
7ai and 8a,c,d,f; and Supplementary Figs 1aj, 2ac, 3bi, 4ce, 5bd,gi, 6ah and
7ad,fh,jm.

Accession numbers. The NanoString data generated for this manuscript were
submitted to the Gene Expression Omnibus database, accession number GSE61593.
57. Smith, A. L., Friedman, D. B., Yu, H., Carnahan, R. H. & Reynolds, A. B. ReCLIP
(reversible cross-link immuno-precipitation): an efficient method for interrogation of
labile protein complexes. PLoS ONE 6, e16206 (2011).
58. Smoot, M. E., Ono, K., Ruscheinski, J., Wang, P. L. & Ideker, T. Cytoscape 2.8:
new features for data integration and network visualization. Bioinformatics 27,
431432 (2011).

NATURE CELL BIOLOGY

2015 Macmillan Publishers Limited. All rights reserved

S U P P L E M E N TA R Y I N F O R M AT I O N
DOI: 10.1038/ncb3227

p120

apical

Ecad
z

basal

PLEKHA7

p120:Y96

PLEKHA7

p120:Y228
z
x

Caco2

y
MDCK

PLEKHA7

p120:T310

merged
y

basal

x
y

y
MDCK

PLEKHA7

RhoA

apical

Rac1

apical

basal
Caco2

PLEKHA7

basal

Rac1

apical

PLEKHA7

Caco2

basal

Caco2

Myosin
IIA

apical

Afadin

Actin

MDCK

PLEKHA7

apical

PLEKHA7

Caco2

apical

y
MDCK

basal

basal

merged

Ecad

PLEKHA7

basal

PLEKHA7

apical

Caco2

Supplementary Figure 1

Supplementary Figure 1 Distinct complexes exist at the junctions of


epithelial cells. Caco2 or PLEKHA7-GFP transfected MDCK cells were
grown to polarize, subjected to IF for PLEKHA7 or GFP respectively
and co-stained for (a) p120; (b,c) E-cadherin (Ecad); (d) Afadin,
Actin (phalloidin), Myosin IIA; (e) phosphorylated p120: Y228; (f)

phosphorylated p120: Y96; (g) phosphorylated p120: T310; (h,i) Rac1; (j)
RhoA. Stained cells were imaged by confocal microscopy and image stacks
were acquired, as in Figure 1. Representative x-y image stacks are shown
and/or the merged composite x-z images. Scale bars for x-y images: 20 M;
for x-z images: 5 M.

WWW.NATURE.COM/NATURECELLBIOLOGY

1
2015 Macmillan Publishers Limited. All rights reserved

S U P P L E M E N TA R Y I N F O R M AT I O N

WBs

Ecad

Ecad
-catenin

PLEKHA7

merged

NT

-catenin

shEcad
IPs

c
p120

PLEKHA7

merged

NT

shp120

sh120+1A

sh120+4A

Supplementary Figure 2 PLEKHA7 localization to the junctions is


E-cadherin- and p120-dependent. (a) Western blot of the lysates from the
separated apical and basolateral fractions shown in Figure 2 for E-cadherin
(Ecad), -catenin, and -catenin. (b) Caco2 control (NT) or E-cadherin
knockdown (shEcad) cells, stained by IF for E-cadherin and PLEKHA7.

Supplementary Figure 2

(c) p120 and PLEKHA7 IF stainings of Caco2 control (NT) cells, p120
knockdown (shp120) cells, and of p120 knockdown cells transfected either
with the full length murine mp120-1A isoform (shp120+1A) or the murine
mp120-4A isoform that lacks the N-terminal PLEKHA7-binding domain
(shp120+4A). All scale bars: 20 M.

2

WWW.NATURE.COM/NATURECELLBIOLOGY
2015 Macmillan Publishers Limited. All rights reserved

S U P P L E M E N TA R Y I N F O R M AT I O N

a
1.2

fold change

PLEKHA7
mRNA

shPLEKHA7

NT

soft agar colonies

PLEKHA7

0.8
0.6

0.4
0.2
0

NT

#8

#8
#10
shPLEKHA7

NT

p120

#10

shPLEKHA7

PLEKHA7

p120

merged

p-p120:
Y228

e
soft agar
colonies

PLEKHA7

NEZHA

Src
p-Src:
Y416

NEZHA

SNAI1

p120

adGFP

p-p120:
Y228

adSNAI1

p-Src:
Y416

MDCK

NT

Actin

Src

shNEZHA

Cad11

SNAI1

i
mp120

MYC

PLEKHA7

CCND1

p120

**

Src

1A

Actin

PLEKHA7

p-Src:Y416

NT #72 #73
shNEZHA

SNAI1
ARM1

MYC
CCND1
Actin

Supplementary Figure 3

Supplementary Figure 3 PLEKHA7 loss from the junctions results in


increased anchorage-independent growth and related signalling. (a)
Demonstration of the PLEKHA7 mRNA knockdown by qRT-PCR after
infection of Caco2 cells with two PLEKHA7 shRNAs (shPLEKHA7 #8 and
#10) or non-target control shRNA (NT) (mean s.d. from n=3 independent
experiments; *P<0.0001, Students two-tailed t-test). Source data are
provided in Supplementary Table 2. (b) Caco2 control (NT) and PLEKHA7
shRNA knockdown (shPLEKHA7) cells were stained and imaged for
PLEKHA7 and p120. (c) Caco2 control (NT) or PLEKHA7 knockdown cells
(shPLEKHA7 #8, #10) grown on soft agar and imaged for colony formation
(images are in 2x magnification; see Fig. 3c for quantitation). (d) MDCK cells
were infected with either control (NT) or PLEKHA7 shRNA (shPLEKHA7#8)
and subjected to western blot for the markers shown. Phosphorylation
sites are denoted by p-. Actin is the loading control. (e) Soft agar assay

of Caco2 cells infected with either vector control (adGFP) or a SNAI1expressing construct (adSNAI1) (images are in 2x magnification; see Fig.
3h for quantitation). (f) Western blot of control (NT) or NEZHA knockdown
(shNEZHA#72 and #73 shRNAs) Caco2 cells for the markers shown; Actin is
the loading control. (g) IF of control (NT) or NEZHA knockdown (shNEZHA)
Caco2 cells for PLEKHA7 and NEZHA. (h) IF of Caco2 cells transfected
with either wild type murine mp120-1A (1A) or the murine mp120-ARM1
(ARM1) construct that cannot bind E-cadherin, stained with the murinespecific p120 antibody 8D11 (mp120) and co-stained with PLEKHA7.
(i) Western blot of pcDNA (vector control), mp120-ARM1, or mp120-1A
transfected cells, for the markers shown; Actin is the loading control. Single
and double stars on the p120 blot indicate the 1A and ARM1 bands,
respectively, right above the endogenous p120 bands. Scale bars for x-y
images: 20 M; for x-z images: 2.5 M; for panels c and e: 2 mm.

WWW.NATURE.COM/NATURECELLBIOLOGY

3
2015 Macmillan Publishers Limited. All rights reserved

S U P P L E M E N TA R Y I N F O R M AT I O N

BC

normal

Triple neg.

PLEKHA7

DCIS

absent

100%

mislocalized

normal

p120

% cases

80%
60%
40%
20%

Ecad

0%

PLEKHA7

RCC

stage 2

Ecad

stage 3
absent

PLEKHA7

normal

p120

mislocalized

normal

p120

% cases

100%
80%
60%
40%
20%

Ecad

0%

PLEKHA7

PLEKHA7
p120
shPLEKHA7
shp120

+
-

+
+

Ecad

soft agar colonies

p120

Ecad

p120

p120:Y228

control

-Tubulin

shPLEKHA7 - - + +
shp120 - + - +

PP2

Supplementary Figure 4
Supplementary Figure 4 PLEKHA7 is mis-localized or lost in breast and
renal tumour tissues. Representative immunohistochemistry images of
(a) breast and (b) kidney (renal), normal and cancer tissues stained for
PLEKHA7, p120 and E-cadherin (Ecad) (left panels) and the percentage
of tissues that exhibit presence, absence, or mis-localization of the three
markers examined (right panels). BC: breast cancer; RCC: renal cell
carcinoma. Scale bars: 20 M. Number of tissues examined per cancer
type/stage; BC TMA: Benign, n=8; DCIS, n=12; ILC ER+ Her-, n=10; IDC
ER+, Her-, n=16; IDC Her+, n=16; IDC Triple Negative, n=13; RCC TMA:

Matched normal, n=119; stage 1, n=71; stage 2, n=20; stage 3, n=22;


stage 4, n=6. (c) Caco2 control (NT), PLEKHA7 knockdown (shPLEKHA7),
and PLEKHA7, p120 (shPLEKHA7, shp120) double knockdown cells were
grown on soft agar for colony formation assay (images are shown in 2x
magnification; see Fig. 4a for quantitation) and (d) examined by western
blot for E-cadherin (Ecad) levels; -Tubulin is the loading control. (e) Caco2
cells treated with either vehicle (DMSO) or the Src inhibitor PP2 (10 M)
were stained for p120 and phosphorylated p120: Y228. Scale bars 20 M;
for panel c: 2 mm.

4

WWW.NATURE.COM/NATURECELLBIOLOGY
2015 Macmillan Publishers Limited. All rights reserved

S U P P L E M E N TA R Y I N F O R M AT I O N

fold change

2.5

NT
shPLEKHA7

NT
CHX (min):
PLEKHA7

1.5

SNAI1

MYC

0.5

CCND1

SNAI1 CCND1 c-Myc

Actin

mRNAs

NT

shPLEKHA7

MG-132 (min): 0 15 30 60 120 240 0 15 30 60 120 240


PLEKHA7

PLEKHA7
p-GSK3

SNAI1

GSK3

Actin

miR-19a

fold change

3.5
2.5
2
1.5
1
0.5
0

- + +
- - +
NT

shPLEKHA7

0.6

0
shPLEKHA7
LZRS-PLEKHA7

- + +
- - +
NT

shPLEKHA7

shPLEKHA7+LZRS-PLEKHA7

p-p120:
Y228

p120

0.8

0.2

p-p120:
Y228

shPLEKHA7
LZRS-PLEKHA7

0.4
NT #8 #10
shPLEKHA7

CCND1

miR-30b

1.2

Actin

MYC

shPLEKHA7

0 15 30 60 120 240 0 15 30 60 120 240

fold change

NT shPLEKHA7
p-p120:
Y228

p120

p-Src:
Y416

p-Src:
Y416

Src

Src

p120
Actin

shPLEKHA7+LZRS-PLEKHA7

anti-miRs

5but not postSupplementarySupplementary


Figure 5 PLEKHA7 actsFigure
via miRNAs
translational modification mechanisms. (a) qRT-PCR analysis for the
indicated mRNAs of Caco2 control (NT) or PLEKHA7 knockdown cells
(shPLEKHA7) (mean s.d. from n=3 independent experiments). (b)
Western blot for the markers shown (Actin: loading control) of control (NT)
or PLEKHA7 knockdown (shPLEKHA7) Caco2 cells treated with 20 g/ml
cycloheximide (CHX) or (c) 10 M MG-132, for the indicated time points.
(d) Western blot of control (NT) or PLEKHA7 knockdown (shPLEKHA7)
Caco2 cells for the markers shown; Actin is the loading control. (e,f) miR30b and miR-19a qRT-PCR analysis of PLEKHA7-knockdown (shPLEKHA7)

miR-mimics

Caco2 cells after ectopic re-expression of PLEKHA7 (LZRS-PLEKHA7)


(mean s.d. from n=3 independent experiments; *P<0.05, Students
two-tailed t-test) (g) Caco2 cells transfected with the indicated anti-miRs
(a-miR) were subjected to western blot for the markers shown. (h) Caco2
control (NT) and PLEKHA7 knockdown (shPLEKHA7) cells were transfected
with either control or miR-30b mimic and blotted for the markers shown.
(i) Caco2 cells infected with either vector control (adGFP) or a SNAI1expressing construct (adSNAI1) were subjected to western blot for the
markers shown; Actin is the loading control. Source data for panels a, e, f
are provided in Supplementary Table 2.

WWW.NATURE.COM/NATURECELLBIOLOGY

5
2015 Macmillan Publishers Limited. All rights reserved

S U P P L E M E N TA R Y I N F O R M AT I O N

nt:

*2nd ab

*2nd ab

80

p120
apical
basal

merged

apical

21

DROSHA*

p120

basal

premiR-30b

50

merged

mature
miR-30b

U6

Caco2

Caco2

DROSHA

merged

p120

merged

DGCR8

basal

apical

apical

p120

basal

DGCR8*

MDCK

p120

DROSHA

NT

kDa

202
113
73
37

MDCK

p120

DGCR8

DROSHA

kDa
202
113
73

NT

47

Actin

37

siDROSHA

DGCR8
Actin

siDGCR8

WBs
PLEKHA7
p120
Ecad

Supplementary Figure 6

IPs

Supplementary Figure 6 The microprocessor complex localizes at the


ZA. (a) Northern blot analysis of control (NT) or PLEKHA7 knockdown
(shPLEKHA7) Caco2 cells using a miR-30b probe, indicating the premiR-30b and the mature miR-30b. U6 is the loading control. (b,c) IF of
polarized Caco2 cells for p120 and DROSHA or DGCR8. Antibodies used
here: DROSHA: Sigma and DGCR8: Sigma. The indication *2nd ab refers to
the use here of a different antibody for DROSHA and DGCR8, compared to
the antibodies used in Fig. 6d,e (for antibody details, see Supplementary
Table 3). Enlarged details in boxes are shown on top of apical fields. The
x-z composite image of panel b is shown in Fig. 6f and of c in Fig. 6g. (d,e)

IF of polarized MDCK cells for p120 and DROSHA (antibody: Abcam) or


DGCR8 (antibody: Abcam). (f,g) siRNA-mediated knockdown of DROSHA
(siDROSHA) and DGCR8 (siDGCR8) in Caco2 cells, shown both by IF (left
side of each panel), and western blot (right side of each panel). Non-target
(NT) siRNA is the control. p120 was used as a co-stain for the IF; Actin is
the loading control for the blots; molecular weights (kDa) are indicated on
the left side of each blot. (h) Western blots of PLEKHA7, p120, DROSHA,
and DGCR8 IPs for PLEKHA7, p120 and E-cadherin (Ecad). IgG is the
negative IP control. Scale bars: 20 M; for enlarged parts of panels b and
c: 3 M.

6

WWW.NATURE.COM/NATURECELLBIOLOGY
2015 Macmillan Publishers Limited. All rights reserved

S U P P L E M E N TA R Y I N F O R M AT I O N

PLEKHA7

DROSHA

mp120

DGCR8

mp120

1A

1A

1A

ARM1

ARM1

ARM1

DROSHA

p120

fold change

mp120

NT

NT

1.2
1.0
0.8

0.6

shp120

PLEKHA7

DROSHA

DGCR8

control

0.4
0.2
0.0

Nocodazole

shp120

NEZHA

DROSHA

NEZHA

NT

NT

shNEZHA

shNEZHA

j
p120

DGCR8

k
DROSHA

p120

DGCR8

l
Cad11

DROSHA

Cad11

DGCR8

NT

NT

NT

NT

shPLEKHA7

shPLEKHA7

shPLEKHA7

shPLEKHA7

shPLEKHA7 + shp120

shPLEKHA7 + shp120

shPLEKHA7 + shCad11

shPLEKHA7 + shCad11

Supplementary Figure 7
Supplementary Figure 7 Regulation of the microprocessor at the ZA is
PLEKHA7-depended. (a-c) IF of Caco2 cells transfected with either the wild
type murine mp120-1A (1A) construct or the murine mp120-ARM1 (ARM1)
construct that cannot bind E-cadherin, both stained with the murine-specific
p120 antibody 8D11 (mp120) and co-stained with PLEKHA7, DROSHA, or
DGCR8. Arrows indicate affected junctional staining of the indicated markers
in transfected cells. (d) IF of control (NT) or p120 knockdown (shp120)
Caco2 cells for DROSHA and p120. (e) qRT-PCR of control (NT) and p120
knockdown (shp120) Caco2 cells for the miRNAs shown (mean s.d. from
n=3 independent experiments; *P<0.05, Students two-tailed t-test). (f) IF of
control (DMSO) or Nocodazole (10 for 8h) treated Caco2 cells for PLEKHA7,
DROSHA, or DGCR8. (g,h) IF of control (NT) or NEZHA knockdown (shNEZHA)
Caco2 cells for DROSHA and DGCR8, co-stained with NEZHA. (i) qRT-PCR

of control (NT), PLEKHA7 knockdown (shPLEKHA7), PLEKHA7+p120


(shPLEKHA7+shp120), and PLEKHA7+Cadherin-11 (shPLEKHA7+shCad11)
double knockdown Caco2 cells for the indicated miRNAs (individual data
points and mean from n=2 independent experiments are shown). (j,k) IF of
control (NT), PLEKHA7 knockdown (shPLEKHA7), and PLEKHA7+p120
double knockdown (shPLEKHA7+shp120) Caco2 cells for DROSHA
and DGCR8, co-stained with p120. (l,m) IF of control (NT), PLEKHA7
knockdown (shPLEKHA7), and PLEKHA7+Cadherin-11 double knockdown
(shPLEKHA7+Cad11) Caco2 cells for DROSHA and DGCR8, co-stained with
Cadherin 11 (Cad11). Non-specific cytoplasmic or nuclear background appears
respectively for the two Cadherin-11 antibodies used in the IF (see Methods
for antibody details). All scale bars: 20 M. Source data for panels e and i are
provided in Supplementary Table 2.

WWW.NATURE.COM/NATURECELLBIOLOGY

7
2015 Macmillan Publishers Limited. All rights reserved

S U P P L E M E N TA R Y I N F O R M AT I O N

kDa
202

114
202
114

Fig. 2d
PLEKHA7

p120

73
53

TRIM21

36
28
202

IQGAP1

114

Actin

206
118
97
54
37
29

202

114
73

p-p120:Y228

28
114

EWS

73

CCAR1

114
73

kDa

SNAI1

Cad11

117
77

CCND1

28

78
53
36
28

short
exposure
-Tubulin
SNAI1

p-Src:
Y416

54

Actin

77
52
36
28

203
117
77

p120

47

Actin

33
27
202
114
72

Cad11
shPLEKHA7 - + +
LZRS-PLEKHA7-myc - - +

202
114
72

VIM

52

PLEKHA7

202
114
72

MYC (~62kDa)

77

Fig. 3g

202
114
72

PLEKHA7

52

Fig. 3h

206
118
97

NT #8 #10
shPLEKHA7

Fig. 3f

36

IPs

p-p130CAS:
Y165

NT #8 #10
shPLEKHA7

203

202

97
54
37

37
29
17

p-p120:Y96

kDa
203
117
52
36
28

HNRNP
A2B1

36

Src

Actin

ACTN1

53

p-p120:
Y228

47
33
27

FAK

SNAI1

72
47

Actin

MYC

47
33
27

202

52
36
28

PLEKHA7

p130CAS

54
37

p120

206
118
97
54

Fig. 3e

206
118
97

NT #8 #10
shPLEKHA7

206
118
97

73

kDa
206
118
97

PLEKHA7

37

pp120:Y228

114

206
118
97
97
54

73
202

Fig. 3d

kDa

114
72

CCND1

p-p120:
Y96

NT #8 #10
shPLEKHA7

Supplementary Figure 8
Supplementary Figure 8 Full western blot scans. Multiple experiments
were routinely run on every gel and the membranes were cut and blotted for
multiple antibodies for each experiment. The cut blots for each antibody

are shown here per case. Molecular weights are indicated on the left; the
cropped images shown in the respected Figure panels are indicated in yellow
boxes.

8

WWW.NATURE.COM/NATURECELLBIOLOGY
2015 Macmillan Publishers Limited. All rights reserved

S U P P L E M E N TA R Y I N F O R M AT I O N

Fig. 4b

kDa

Fig. 4c

PLEKHA7

211
118
78
211
118
78

205
113
79

p120

211
118
78

p130CAS

211
118
78

p-p130CAS:
Y165

36
28

SNAI1

PLEKHA7

205
113
79

CCND1

Cad11
MYC

53

Actin

36

p120 202
114
72

202
114
72

33
27

33
27

34

27

47
33

17

27

34

MYC

CCND1

27

202
114
72

79

MYC
(~62kDa)

p130CAS

kDa

211
118
78

- + shp120

205
113
79

p-p130CAS: Y165

205
113
79

Fig. 4e

37

211
118
78
53
36

SNAI1

211
118
78

p-p120:
Y228

28

211
118
78

53
36

p120

Cad11
Actin

Supplementary Figure 8
(part 2)

Actin
-

+
-

+
+

shPLEKHA7
shEcad

PLEKHA7

kDa
50
37
25
75
50

Fig. 4f
kDa

202
114
72

p-p130CAS:
Y165

50
37

Cad11

211
118
78

25

CCND1

53
36
28
17

shCad11 -

MYC

- +

+ -

- - + +
- +

- +

Fig. 5c
SNAI1

MYC
(~62kDa)

CCND1

Actin

SNAI1

78
53
36
shPLEKHA7 -

Actin

shPLEKHA7
PP2

p130CAS

47

p120

Cad11

202
114
72

47
33
27

37

- + + shPLEKHA7
-

CCND1

SNAI1

72

SNAI1

47

211
118
78
78
53

PLEKHA7
p-p120:Y228

Ecad

17

36
28

Fig. 4d

kDa
202
114
72

53
36
28
+
+

antimiRs:

Actin

- - + +
- +

- +

Supplementary Figure 8 continued

WWW.NATURE.COM/NATURECELLBIOLOGY

9
2015 Macmillan Publishers Limited. All rights reserved

S U P P L E M E N TA R Y I N F O R M AT I O N

kDa

Fig. 5d

201
114
74
47
34

Fig. 6b

kDa
202
114

PLEKHA7

SNAI1

27
73

47

MYC

34

CCND1

27
47

Fig. 6h

114
73

73
47

DROSHA

kDa
202

DGCR8

114

Lamin A/C

17
6

114
77

Histone H3
47
33
27

GAPDH

47
33

Actin

p120

202
114
77
202
114
77

Actin

34
27

PLEKHA7

DROSHA

DGCR8

IPs

miR-mimics

total

non- nuclear
nuclear

NT shPLEKHA7

kDa

Fig. 6i

Fig. 6j

202
114

PLEKHA7

202
114
77

p120

202
114
114
77

DROSHA

kDa
202
114

kDa
PLEKHA7

202
114

DROSHA

114
73

DGCR8

DGCR8
IPs
IPs

202
114
73
202
114
73

Fig. 8a
DROSHA
DGCR8
short exposure:
DROSHA
DGCR8

73
47

Lamin A/C

17
6
47
33
27

Histone H3
GAPDH

Supplementary Figure 8 continued

Supplementary Figure 8 (part 3)

10 

WWW.NATURE.COM/NATURECELLBIOLOGY
2015 Macmillan Publishers Limited. All rights reserved

S U P P L E M E N TA R Y I N F O R M AT I O N

Supplementary Table Legends


Supplementary Table 1 The proteins identified by proteomics of junctional fractions.
Supplementary Table 2 The source data file.
Supplementary Table 3 The antibodies used in the study.

WWW.NATURE.COM/NATURECELLBIOLOGY

11
2015 Macmillan Publishers Limited. All rights reserved

You might also like