Regulation of cell proliferation and migration by p62 through ... - PNAS

1 downloads 0 Views 3MB Size Report
Jun 24, 2014 - 3E), whereas it had no effect on the stability of Twist2 (Fig. S2A), another member of the Twist subfamily of bHLH proteins regulating gene ex-.
Regulation of cell proliferation and migration by p62 through stabilization of Twist1 Lei Qianga, Baozhong Zhaoa, Mei Minga, Ning Wangb, Tong-Chuan Heb, Seungmin Hwangc, Andrew Thorburnd, and Yu-Ying Hea,1 Departments of aMedicine, Section of Dermatology, bOrthopaedic Surgery and Rehabilitation Medicine, and cPathology, University of Chicago, Chicago, IL 60637; and dDepartment of Pharmacology, University of Colorado at Denver and Health Sciences Center, Aurora, CO 80045 Edited by Beth Levine, University of Texas Southwestern, Dallas, TX, and approved May 14, 2014 (received for review December 9, 2013)

SQSTM1

| melanoma | skin cancer | ubiquitination | proteasome

M

acroautophagy (hereafter autophagy) is a catabolic process by which cellular proteins, cytoplasm, and organelles are captured and targeted for proteolytic degradation in lysosomes (1, 2). Autophagy dysfunction is associated with multiple human diseases, such as neurodegeneration, microbial infection, metabolic diseases, cardiovascular diseases, aging, and cancer (2–4). The multidomain protein p62/A170/SQSTM1 (hereafter referred to as “p62”) has been shown to be both a selective autophagy substrate and an autophagy adaptor protein that acts as a link between ubiquitination and autophagy (5, 6). Several studies have demonstrated the oncogenic role of p62 in tumor formation and/ or progression (7, 8) through regulating NF-kappaB (9, 10) and NRF2 (11-13). Furthermore, Ras induces p62 expression in tumorigenesis (10). However, much remains to be elucidated with regard to its function and interaction with other critical cellular pathways. The transcription factor Twist1 is a core regulator in both early embryonic morphogenesis and cancer development and metastasis (14–17). It induces loss of epithelial (E)-cadherin–mediated cell– cell adhesion and facilitates the epithelial–mesenchymal transition (EMT) (16), and it promotes cell proliferation (17). Twist1 is a basic helix–loop–helix (bHLH) protein and is structurally unrelated to other EMT factors including Slug and Snail. Recent studies have shown that Twist1 is a labile protein regulated by the ubiquitin–proteasome system through the F-box protein and E3 ubiquitin ligase Ppa (18). Despite these advances, the regulatory and functional role of Twist1 remains poorly understood. Here we demonstrate that p62 stabilizes Twist1 protein to increase cell proliferation and migration in vitro and in mice.

indicating a deficiency of autophagy (Fig. 1A). Expression of Ecadherin, a negative regulator of cell proliferation and migration, was decreased in all autophagy-deficient cells compared with WT cells (Fig. 1A). A Matrigel invasion assay indicated that Atg3, Atg5, Atg9, and Atg12 KO MEF cells exhibit much higher invasion activity than WT cells (Fig. 1B). A radius migration assay showed that autophagy deficiency promoted the migration ability of MEF cells (Fig. 1C). Furthermore, autophagy deficiency increased cell proliferation (Fig. 1D). Compared with normal human skin, in welldifferentiated human squamous cell carcinoma (SCC WD) and poorly differentiated SCC (SCC PD), p62 was up-regulated, whereas E-cadherin was down-regulated (Fig. 1E). Our data demonstrate that autophagy deficiency increases p62 levels, decreases E-cadherin expression, and promotes cell migration, invasion, and proliferation, and that the p62 levels are inversely associated with E-cadherin expression in human SCCs. Autophagy Deficiency Inhibits Twist1 Degradation by Both Autophagosome and Proteasome. To determine the mechanism

involved in the regulation of E-cadherin by autophagy, we first used the protein synthesis inhibitor cycloheximide (CHX) and the proteasome inhibitor (MG132) to determine the effect of autophagy inhibition on the protein stability of E-cadherin. Neither CHX nor MG132 affected E-cadherin levels in WT or Atg5 KO cells (Fig. S1A), suggesting that suppression of E-cadherin by autophagy deficiency occurs at the mRNA level. Compared with WT cells, the mRNA level of E-cadherin decreased in Atg5 KO MEF cells (Fig. S1B), and the transcriptional activity of the WT E-cadherin promoter but not the E-box mutant E-cadherin promoter was also significantly decreased in Atg5 KO MEF cells compared with WT MEF cells (Fig. 2A). These results indicate that autophagy suppression down-regulates E-cadherin at the transcriptional level. Significance The selective autophagy substrate p62 has been shown to regulate inflammatory and redox pathways in tumorigenesis following autophagy suppression. Here we have elucidated the critical role of p62 in promoting cell proliferation and migration in vitro and tumor growth and metastasis in vivo through its molecular interaction with the oncogenic transcription factor Twist1. Our findings suggest that targeting the interaction between p62 and Twist1 has potential for cancer prevention and therapy.

Results Autophagy Deficiency Decreases E-Cadherin Expression and Promotes Cell Migration, Invasion, and Proliferation. Two distinctive hallmarks

of autophagy are the conversion of light chain 3-I (LC3-I) to LC3-II and the selective degradation of p62 (19, 20). Multiple mammalian homologs of products of the autophagy-related genes (Atg) originally identified in yeast have been identified (3). Compared with wild-type (WT) mouse embryonic fibroblast (MEF) cells, cells with Atg3, Atg5, Atg9, and Atg12 knockout (KO) blocked the conversion of LC3-I to LC3-II and induced p62 accumulation, www.pnas.org/cgi/doi/10.1073/pnas.1322913111

Author contributions: L.Q. and Y.Y.H. designed research; L.Q. performed research; B.Z., M.M., N.W., T.-C.H., S.H., and A.T. contributed new reagents/analytic tools; L.Q. and Y.Y.H. analyzed data; and L.Q., S.H., and Y.Y.H. wrote the paper. The authors declare no conflict of interest. This article is a PNAS Direct Submission. 1

To whom correspondence should be addressed. E-mail: [email protected]. edu.

This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10. 1073/pnas.1322913111/-/DCSupplemental.

PNAS | June 24, 2014 | vol. 111 | no. 25 | 9241–9246

MEDICAL SCIENCES

The selective autophagy substrate p62 serves as a molecular link between autophagy and cancer. Suppression of autophagy causes p62 accumulation and thereby contributes to tumorigenesis. Here we demonstrate that autophagy deficiency promotes cell proliferation and migration through p62-dependent stabilization of the oncogenic transcription factor Twist1. p62 binds to Twist1 and inhibits degradation of Twist1. In mice, p62 up-regulation promotes tumor cell growth and metastasis in a Twist1-dependent manner. Our findings demonstrate that Twist1 is a key downstream effector of p62 in regulation of cell proliferation and migration and suggest that targeting p62-mediated Twist1 stabilization is a promising therapeutic strategy for prevention and treatment of cancer.

Atg3 Atg5 Atg9 Atg12 WT KO WT KO WT KO WT KO

E-cadherin p62 LC3-I LC3-II GAPDH

% Closure

C

% Cell Number

D

E

WT Atg3 KO

WT Atg3 KO

Normal Skin

WT Atg5 KO

WT Atg5 KO

B

# Migration cells/field

A

WT KO

600 500 400 300 200 100 0

Atg3 Atg5 Atg9 Atg12 WT Atg12 KO

WT Atg9 KO

WT Atg9 KO *

SCC WD

WT Atg12 KO

SCC PD

by autophagy (26), autophagy inhibition increased the protein stability of Twist1, but did not completely block Twist1 degradation (Fig. 2E), suggesting that Twist1 is degraded through autophagy and other pathways such as proteasome. Inhibition of proteasomes by MG132 increased Twist1 abundance in WT cells but not in autophagy-deficient cells (Fig. 2F and Fig. S1G). These results indicate that Twist1 can be degraded through both autophagy and proteasome pathways. To determine whether autophagy deficiency affects Twist1 levels in vivo in mouse epithelial tissue, we analyzed the protein levels of E-cadherin, p62, and Twist1 in the epidermis from mice with wild-type (WT, K14Cre;Atg7+/+) and skin-specific Atg7 conditional knockout (K14Cre;Atg7flox/flox, Atg7 cKO). Compared with WT epidermis, both p62 and Twist1 were up-regulated in the Atg7 cKO epidermis, in parallel with decreased Ecadherin expression (Fig. 2G and Fig. S1H). Reconstitution of Atg7 in Atg7 KO cells restored autophagy, increased E-cadherin expression, and decreased p62 and Twist1 abundance (Fig. 2H). p62 Regulates Twist1 Protein Levels in Autophagy-Proficient and -Deficient Cells. To determine the role of p62 in Twist1 up-regulation by

autophagy inhibition, we assessed the effect of p62 knockdown HE

Several E-cadherin transcriptional repressors have been characterized (Snail, Slug, ZEB1, and Twist1) and shown to act through an interaction with the proximal E-boxes of the E-cadherin promoter (21–23). Compared with WT cells, we found that only Twist1 was up-regulated, whereas Snail, Slug, and ZEB1 were unaltered in Atg5 KO cells (Fig. 2B). Autophagy deficiency increased the level of neural (N)-cadherin (Fig. 2B) and the activity of the transcription factor TCF/lymphoid enhancer factor (LEF) complex (Fig. S1C), the downstream pathways negatively regulated by E-cadherin (24). Compared with WT MEF cells, Twist1 was up-regulated in cells with autophagy deficiency, whereas it was down-regulated in p62 KO MEF cells compared with WT cells (Fig. S1D). However, autophagy inhibition had no effect on the mRNA level of Twist1 (Fig. S1E). In addition, adding Twist1 to WT cells suppressed the expression of E-cadherin (Fig. S1F), indicating that Twist1 is sufficient to suppress E-cadherin expression. In both WT and Atg5 KO cells, Twist1 and p62 colocalized in punctate structure with or without rapamycin treatment(Fig. 2C). The p62-positive puncta in Atg5 KO cells are likely early autophagic structures as demonstrated in recent studies (25). In contrast, Twist1 colocalized with LC3 in rapamycin-induced puncta only in WT cells, but not in Atg5 KO cells (Fig. 2D). Similar to the regulation of p53 protein stability 9242 | www.pnas.org/cgi/doi/10.1073/pnas.1322913111

E-Cadherin-Luc

Snail

CHX

WT 3 6 12

Atg5 KO 3 6 12(h)

WT 3 6 12

Atg5 KO 3 6 12(h)

p53

Slug

GAPDH

ZEB1

F

Twist1 WT Atg5 KO Merge

Twist1

MG132

GAPDH p62

p53 DAPI

Twist1 GAPDH

WT

C

E Twist1

G

Atg5 KO

Fig. 1. Autophagy deficiency decreases E-cadherin expression and promotes cell migration, invasion, and proliferation. (A) Immunoblot analysis of E-cadherin, p62, LC3-I/II, and GAPDH in WT and Atg3, Atg5, Atg9, and Atg12 KO MEF cells. (B) Transwell assay of the invasion ability of WT and Atg3, Atg5, Atg9, and Atg12 KO MEF cells. Migrated cells on the underside of Transwell filters were stained and observed under microscopy. (C) Radius migration assay of the migration ability of WT and Atg3, Atg5, Atg9, and Atg12 KO MEF cells. (D) CellTiter 96 AQueous Non-Radioactive Cell Proliferation Assay (MTS) of WT and Atg3, Atg5, Atg9, and Atg12 KO MEF cells. The results were obtained from three independent experiments [mean ± SD (error bars), n = 3; *P < 0.05; **P < 0.01, compared with the WT group]. (E) Representative histological and immunohistochemical analysis of p62 and E-cadherin protein levels (brown) in normal skin (n = 14), well-differentiated squamous cell carcinoma (SCC WD) (n = 25), and poorly differentiated SCC (SCC PD) (n = 10). Black squares indicate the region shown in higher magnification. (Scale bar, 200 μm and 50 μm for Left and Right, respectively.)

B

Atg5 WT KO E-cad WT-Luc E-cad Ebox Mut-Luc N-cadherin

E-cadherin

WT Atg7 cKO #1 #2 #1 #2

Twist1 p62 LC3-I LC3-II

Rapamycin Atg5 KO WT

E-cadherin

A

D Rapamycin WT Atg5 KO

p62

GAPDH

H Merge

Twist1

LC3

WT Atg7 KO

Atg7 E-cadherin

DAPI

p62 Twist1 LC3 Atg7 GAPDH

Fig. 2. Autophagy deficiency delays Twist1 degradation via both autophagosome and proteasome pathways. (A) Luciferase reporter assay of the E-cadherin promoter with an intact (E-cad WT-Luc) or mutated (E-cad E-box Mut-Luc) E-box site in WT and Atg5 KO MEF cells. (B) Immunoblot analysis of N-cadherin, Snail, Slug, ZEB1, and Twist1 in WT and Atg5 KO MEF cells. (C and D) Immunofluorescence assay of the colocalization of p62 (C) or LC3 (D) with Twist1 in WT and Atg5 KO MEF cells following treatment with vehicle or rapamycin (500 nM) for 6 h. (Scale bar, 10 μm.) The blue is a DAPI nuclear counterstain. (E and F) Immunoblot analysis of p53, Twist1, and GAPDH in WT and Atg5 KO MEF cells treated with CHX (100 μg/mL, E) or MG132 (10 μM, F) for the indicated times. (G) Immunoblot analysis of p62, Twist1, and GAPDH in the skin from WT and Atg7 cKO mice. (H) Immunoblot analysis of E-cadherin, p62, Twist1, LC3-I/II, Atg7, and GAPDH in WT and Atg7 KO cells reconstituted with Atg7. These results were obtained from three independent experiments.

Qiang et al.

WT

Atg5 KO

siCon sip62 siTwist1 E-cadherin

B

WT

Atg5 KO

shCon shp62 shTwist1 E-cadherin

C WT Atg5 KO p62 Twist1

p62

p62

p62

Twist1

Twist1

GAPDH

GAPDH

GAPDH

D

WT

p62 Twist1 p62 GAPDH

p62 KO

E CHX HA

Myc-Twist1 3 6 12

HA-p62 Myc-Twist1 3 6 12(h)

Myc GAPDH

Fig. 3. Autophagy deficiency stabilizes Twist1 via p62 accumulation. (A) Immunoblot analysis of E-cadherin, p62, Twist1, and GAPDH in WT and Atg5 KO cells transfected with vector and siRNA targeting p62 and Twist1. (B) Immunoblot analysis of E-cadherin, p62, Twist1, and GAPDH in WT and Atg5 KO cells transfected with vector and shRNA targeting p62 and Twist1. (C) Immunoblot analysis of p62, Twist1, and GAPDH in WT and Atg5 KO cells transfected with vector and p62. (D) Immunoblot analysis of p62, Twist1, and GAPDH in WT, p62 KO cells, and p62 KO cells reconstituted with p62. (E) Immunoblot analysis of HA, Myc, and GAPDH in 293T cells transfected with Myc-Twist1 and HA-p62 for 48 h and then treated with CHX (100 μg/mL) for the indicated times. These results were obtained from three independent experiments.

Qiang et al.

Twist1-Ub

98

10% input GST GST-p62 wt ko wt ko wt ko HA-Ub 188

B

Twist1-Ub

188

IB:Twist1

IB:Twist1

98

GST-p62 IB:p62

28

p62

p62 IgG

IB:p62

5% input IgG IP:Twist1 wt ko wt ko wt ko HA-Ub

C

28

IB:Twist1

D WT 188 98

IB:Twist1

Atg5 KO

Myc-Flag-Rad23B Twist1 Rad23B

IgG

GAPDH 28 IB:p62

F

IB:Rad23B

E

Myc-Twist1

Myc NSEEE NLS1NLS2 1

b H L H WR

207

Myc-Twist1- WR

Myc NSEEE NLS1 NLS2 b H L H 1 PB1 1 PB1 1

TBS

LIR

KIR UBA

HA-p62- UBA ZZ TBS LIR KIR

HA

444 HA

IP:Myc

HA-Ub

188 IB:HA 98

180

HA-p62 ZZ

IgG

HA-p62 HA-p62- UBA Myc-Twist- WR Myc-Twist1

HA-p62 Input: HA-p62 Myc-Twist1

382

Fig. 4. p62 binds to Twist1 through its UBA domain. (A) Immunoblot analysis of Twist1 and p62 following immunoprecipitation using control speciesmatched IgG and anti-p62 antibody in WT and Atg5 KO MEF cells. (B) GST pulldown assay of the binding of GST-p62 with Twist1 in WT and Atg5 KO MEF cells. (C) Immunoblot analysis of Twist1, p62, and Rad23B following immunoprecipitation using control species-matched IgG, and anti-Twist1 antibody in WT and Atg5 KO MEF cells. (D) Immunoblot analysis of Twist1, Rad23B, and GAPDH in WT and Atg5 KO MEF cells transfected with vector control or MycFlag-Rad23B. (E) Schematic for p62 and Twist1 deletion constructs. (F) Immunoblot analysis of HA (HA-p62) and Myc (Myc-Twist1) in total cell lysates (input) or HA (HA-Ub, and HA-p62) following immunoprecipitation using control species-matched IgG and anti-Myc (Myc-Twist1) antibody in WT and Atg5 KO MEF cells. Immunoprecipitation assay of the binding of p62 or p62-ΔUBA with Twist1 or Twist1-ΔWR in 293T cells transfected with the combination of MycTwist1 and HA-p62, the combination of Myc-Twist1 and HA-p62-ΔUBA, and the combination of HA-p62 and Myc-Twist1-ΔWR for 48 h. The results were obtained from three independent experiments. Molecular weight in kilodaltons is marked in Fig. 4 A–C and F.

p62 has been demonstrated to bind with polyubiquitinated proteins through its UBA domain (30). To determine the role of the UBA domain of p62 in Twist1 binding, we generated a UBA deletion HA-p62-ΔUBA construct (Fig. 4E). Immunoprecipitation analysis showed that the UBA domain was required for binding between polyubiquitinated Twist1 and p62 (Fig. 4F). Taken together, our findings demonstrate that p62 binds with polyubiquitinated Twist1, and that the WR domain of Twist1 and the UBA domain of p62 are required for the interaction of Twist1 and p62. Twist1 Lysine 175 Is Critical for Twist1 Ubiquitination, Degradation, and Binding with p62. We have shown that the WR domain of Twist1

required for Twist1 polyubiquitination (18) is essential for Twist1 interaction with p62 (Fig. 4). However, there is no lysine site in the Twist1 WR domain (Fig. S3A). To determine the lysine site critical PNAS | June 24, 2014 | vol. 111 | no. 25 | 9243

MEDICAL SCIENCES

A

10% input IgG IP:p62 wt ko wt ko wt ko

HA-Ub

HA-Ub

p62 Binds to Twist1 Through Its Ubiquitin-Associated Domain. To determine the role of p62 in Twist1 stabilization, we first assessed whether p62 binds with polyubiquitinated Twist1, which is important for selective degradation through the autophagosome–lysosome system. Using an immunoprecipitation (IP) assay, we found that endogenous p62 bound to ubiquitinated Twist1 (between 98 and 188 kDa) but not the nonubiquitinated Twist1 (28 kDa) in both WT and Atg5 KO MEF cells (Fig. 4A). A GST pulldown assay also showed that recombinant p62 bound to ubiquitinated Twist1 but not the nonubiquitinated Twist1 (Fig. 4B). Endogenous Twist1 bound to p62 (Fig. 4C). This interaction of p62 with Twist1 may be direct or indirect. Compared with WT cells, Twist1 bound more p62 in Atg5 KO cells, probably due to the increased availability of p62 in autophagydeficient cells, but bound less Rad23B (Fig. 4C), which is crucial in delivery of proteasome substrates to proteasomes through its ubiquitin-like (UBL) and ubiquitin-associated (UBA) domains (27–29). Addition of Rad23B reduced the Twist1 protein levels in both WT and Atg5 KO cells (Fig. 4D), suggesting that the stability of Twist1 is at least partially regulated competitively by its binding to either Rad23B or p62. To test the role of the Twist box (WR) domain of Twist1, which is required for Xenopus Twist1 ubiquitination and degradation (18), we generated a WR deletion Myc-Twist1-ΔWR construct (Fig. 4E). Using CHX and MG132, we found that WR deletion stabilized mammalian Twist1 (Fig. S2 B–D). Immunoprecipitation analysis showed that the WR domain was required for the polyubiquitination of Twist1 and for Twist1 binding with p62 (Fig. 4F).

A

Twist1-Ub

and addition. Knockdown of p62 decreased Twist1 protein levels in both WT and autophagy deficiency cells (Fig. 3 A and B). Addition of p62 to either WT or Atg5 KO cells increased the Twist1 levels (Fig. 3C), indicating the direct effect of p62 on the stability of Twist1. Reconstitution of p62 in p62 KO cells restored Twist1 abundance (Fig. 3D). p62 addition increased Twist1 protein stability in 293T cells (Fig. 3E), whereas it had no effect on the stability of Twist2 (Fig. S2A), another member of the Twist subfamily of bHLH proteins regulating gene expression in development and cancer (17). These findings demonstrated that p62 stabilizes Twist1 at least partially by preventing its proteasome degradation.

for Twist1 ubiquitination, we generated individual lysine mutation constructs (lysine to arginine, K→R) for all lysines (Fig. S3A). The K175R Twist1 mutant showed much higher protein levels than WT and other Twist1 mutants, similar to WR deletion (Fig. 5A). K175R mutation also increased Twist1 protein stability (Fig. 5B and Fig. S3B), indicating that lysine 175 is critical for Twist1 degradation. Immunoprecipitation analysis showed that the K175R mutation inhibits Twist1 polyubiquitination compared with wild-type Twist1 (Fig. 5C), indicating that the K175 site is critical for Twist1 polyubiquitination. Both the K175R mutation and WR deletion abolished the binding of Twist1 with p62 (Fig. 5C). K175R mutation and WR deletion also prevented the colocalization of Twist1 with p62 (Fig. 5D), because the polyubiquitination of Twist1 mutants and the binding with p62 was blocked (Fig. 5C). The Myc-Twist1K175R mutant showed increased protein abundance and enhanced the suppression of E-cadherin compared with Myc-

Twist1 (Fig. 5E). p62 increased Myc-Twist1 levels and enhanced E-cadherin suppression in the presence of Myc-Twist1, whereas it did not affect E-cadherin expression in the presence of MycTwist1-K175R (Fig. 5E). In contrast, whereas WR deletion increased abundance of the Twist1, it failed to affect the E-cadherin level (Fig. 5E). This failure may be due to the requirement of the WR domain for Twist1 function (31). These data indicate that K175 is critical for Twist1 ubiquitination, degradation, and interaction with p62 (Fig. 5F). Although K175 is critical for Twist1 polyubiquitination and stability, other lysine residues in Twist1 may also play a role. Therefore, we assessed the effect of other lysine mutations on the stability of Twist1. Mutation of all the K73, K76, and K77 sites or the K137 site together with the K175 site further inhibited Twist1 degradation, compared with K175 mutation alone, indicating that the K73, K76, K77, and K137 sites are also important for Twist1 degradation (Fig. S3C). Single mutation analysis showed that Twist1 stability was increased by mutations of the K76, K77, or K137 sites but not K73 alone (Fig. S3D).

A

p62 Promotes Tumor Cell Growth and Metastasis in Mice Through Regulating Twist1. To determine the functional significance of Myc GAPDH

B

Myc-Twist1 Myc-Twist- WR Myc-Twist-K175R 3 6 12 3 6 12 3 6 12(h)

CHX Myc GAPDH

C

IgG

D Merge

IP:Myc

HA-p62 Myc-Twist1 Myc-Twist- WR Myc-Twist1-K175R HA-Ub

98 HA-p62 Input: HA-p62

F Twist1

HA-p62 Myc-Twist1 Myc-Twist- WR Myc-Twist1-K175R E-cadherin

Myc-Twist1-K175R

Myc-Twist- WR

Myc-Twist1

E

DAPI

Myc-Twist1 188

HA-Ub

IB:HA

Rapamycin Myc p62

Ubiquitinated Twist1 K175

Proteasome

Autophagy Deficiency

HA

Rad23B? Autophagy Deficiency

p62

Autolysosome

Autophagosome

Myc GAPDH

LC3 Isolation Membrane

Fig. 5. Twist1 lysine 175 is critical for Twist1 ubiquitination, degradation, and binding with p62. (A) Immunoblot analysis of Myc and GAPDH in 293T cells transfected with vector (Con), Myc-Twist1 (K→R) mutation constructs. (B) Immunoblot analysis of Myc and GAPDH in 293T cells transfected with Myc-Twist1, Myc-Twist1-ΔWR, or Myc-Twist1-K175R for 48 h and then treated with CHX (100 μg/mL) for the indicated times. (C) Immunoblot analysis of ubiquitinated Twist1 (HA) and HA-p62 using an anti-HA antibody following immunoprecipitation (IP) with an anti-Myc antibody, and Myc in 293T cells transfected with HA-Ub together with a combination of Myc-Twist1, MycTwist1-ΔWR, or Myc-Twist1-K175R with HA-p62. (D) Immunofluorescence assay of the colocalization of p62 and Myc in WT MEF cells transfected with Myc-Twist1, Myc-Twist1-ΔWR, or Myc-Twist1-K175R, following rapamycin (500 nM) treatment for 6 h. (Scale bar, 10 μm.) The blue is a DAPI nuclear counterstain. (E) Immunoblot analysis of E-cadherin, HA, and Myc in WT MEF cells transfected with Myc-Twist1, a combination of Myc-Twist1 and HA-p62, Myc-Twist1-ΔWR, Myc-Twist1-K175R, or a combination of Myc-Twist1-K175R and HA-p62. (F) Schematic for the regulation of Twist1 by p62 through autophagy and proteasome. These results were obtained from three independent experiments.

9244 | www.pnas.org/cgi/doi/10.1073/pnas.1322913111

the p62/Twist1 axis in cancer, we assessed the role of p62 in the proliferation and migration of tumor cells in vitro and the growth and metastasis of a xenografted tumor in mice. In A431 SCC cells that do not express endogenous Twist1, p62 addition had no effect on the E-cadherin level, cell proliferation, or cell migration (Fig. 6 A–D). When exogenous Twist1 was added, p62 increased the level of Twist1, decreased the expression of E-cadherin, and the colocation of E-cadherin with β-catenin at the plasma membrane, a process important for E-cadherin lossinduced cell migration (32), and increased cell proliferation and migration (Fig. 6 A–D and Fig. S4A). Using xenograft mouse models of tumor growth and metastasis, we found that in the absence of Twist1, p62 had no effect on tumor growth (Fig. 6E) or metastasis (no metastasis detected for either control or p62added A431 cells). In the presence of Twist1, however, p62 increased tumor growth and metastasis (Fig. 6 E and F). Histological and immunohistochemical analysis showed that p62/Twist1 addition caused a loss of E-cadherin expression, increased the number of Ki67-positive cells, and caused a loss of tumor boundary compared with the control and Twist1-only groups, whereas p62 alone had no effect compared with the control group (Fig. S4B). In human A375 melanoma cells that express endogenous Twist1, adding p62 increased both the Twist1 level (Fig. 6G) and tumor growth in nude mice (Fig. 6H), whereas knockdown of p62 decreased the Twist1 level (Fig. 6I) and tumor growth in nude mice (Fig. 6J). Inhibition of autophagy in A375 melanoma cells by Atg7 knockdown increased the level of Twist1 and p62 and tumor growth in nude mice (Fig. 6 I and J). These results indicate that Twist1 regulation by p62 enhances tumor cell proliferation and migration in vitro and in vivo in mice. To investigate the role of p62-mediated stabilization of Twist1 in EMT, a process critical for morphogenesis and cancer progression, we treated the control and p62 knockdown human epithelial cell line HaCaT cells that express low-level endogenous Twist1 with EGF and TGF-β for 48 h as described previously (33). In control (Con) HaCaT cells, EGF/TGF-β induced EMT, decreased E-cadherin expression, and increased the protein levels of N-cadherin and the protein and mRNA levels of p62 and Twist1 (Fig. 6 K–M and Fig. S4C). It appeared that EGF/TGF-β induced p62 expression before Twist1 up-regulation (Fig. 6K). The mechanism by which loss of p62 protein up-regulation at 48 h (Fig. 6K) requires further investigation; one possible mechanism is the induction of autophagy by TGF-β as shown by previous studies (34). p62 induction is required for the upregulation of Twist1 protein and EMT, because knockdown of p62 Qiang et al.

I shAtg7

A375

shp62 p62

Twist1 Atg7 GAPDH

J

Week Con p62

K

EGF/TGF- 0 E-cadherin

shCon 6 24 48 0

Twist1 p62/ Twist1

shp62 6 24 48h

N-cadherin Twist1 p62 Week shCon shAtg7 shp62

Week

GAPDH

L

shCon shp62

M

Con EGF/TGF-

Con EGF/ TGF-

Fig. 6. p62 promotes tumor cell growth and metastasis in mice through Twist1. (A) Immunoblot analysis of p62, Twist1, and GAPDH in A431 SCC cells transfected with vector, HA-p62, Twist1, or a combination of p62 and Twist1. (B) Luciferase reporter assay of the E-cadherin promoter with an intact (E-cad WT-Luc) or mutated (E-cad Mut-Luc) E-box site in A431 cells transfected with vector, p62, Twist1, or a combination of p62 and Twist1. (C) Wound healing assay of migration ability of A431 cells transfected with vector control (Con), Twist1, p62, or a combination of p62 and Twist1. (D) MTS assay of proliferation of A431 cells transfected with vector control (Con), p62, Twist1, or a combination of p62 and Twist1 as a function of days after transfection. (E) Average volume (mm3) of A431-Con, A431-p62, A431-Twist1, and A431-Twist1-p62 tumors at different weeks following s.c. injection. (F) Average number of lung tumor nodules per mouse in established A431-Twist1 and A431-Twist1-p62 lung metastasis tumors at 14 wk following injection. (G) Immunoblot analysis of HA, Twist1, and GAPDH in A375 melanoma cells transfected with vector or HA-p62. (H) Average volume (mm3) of A375-Con and A375-p62 tumors at different weeks following s.c. injection. (I) Immunoblot analysis of p62, Twist1, Atg7, and GAPDH in A375 melanoma cells transfected with vector or shRNA targeting Atg7 (shAtg7) or p62 (shp62). (J) Average volume (mm3) of A375-Con, A375-shp62, and A375-shAtg7 tumors at different weeks following s.c. injection. (K) Immunoblot analysis of E-cadherin, N-cadherin, Twist1, p62, and GAPDH in HaCaT human epithelial cells transfected with vector (shCon) or shp62, following treatment with EGF (10 ng/mL) together with TGF-β (10 ng/mL) (EGF/ TGF-β) for 0, 6, 24, or 48 h. (L) Real-time RT-PCR analysis of Twist1 in HaCaT human epithelial cells transfected with shCon or shp62, following treatment with vehicle or EGF/TGF-β for 48 h. (M ) Real-time RT-PCR analysis of p62 in HaCaT human epithelial cells following treatment with vehicle or EGF/TGF-β for 48 h. The results were obtained from three independent experiments [mean ± SD (error bars), n = 3; *P < 0.05; compared with WT Con cells (B); #P < 0.05; compared with the WT E-cadherin promoter (B); *P < 0.05; compared with the Con, A431-p62, and A431-Twist1 groups (E ); and **P < 0.01; compared with the A431-Twist1 group (F )].

inhibited Twist1 protein up-regulation and E-cadherin suppression and delayed EGF/TGF-β–induced EMT (Fig. 6K and Fig. S4C). However, p62 knockdown has no effect on Twist1 mRNA level (Fig. 6L). These data indicate that p62 is required for Twist1 protein up-regulation and E-cadherin down-regulation in EGF/ TGF-β–induced EMT. Qiang et al.

Discussion In this study, we demonstrate that p62 promotes loss of Ecadherin, cell proliferation and migration in vitro, and tumor growth and metastasis in vivo through stabilizing Twist1. In autophagy-competent cells, Twist1 can be degraded through both autophagy and proteasome. However, in autophagy-defective cells, Twist1 degradation by autophagosome was blocked, whereas accumulated p62 binds with ubiquitinated Twist1 and thus inhibits Twist1 degradation by proteasome (Fig. 5F). Accumulation of p62 inhibits Twist1 binding with Rad23B, which may act as a delivery protein for targeting Twist1 to proteasome for degradation (27–29) (Fig. 5F). Consequently, increased level of p62 leads to the loss of the cell–cell adhesion mediator E-cadherin in advanced human skin SCC. Our findings here elucidate the critical previously unidentified role of the selective autophagy substrate p62 in tumor promotion and progression. The posttranslational modifications of Twist1 and its impact on tumor growth and metastasis remain poorly understood. It was largely undetectable in healthy adult tissues but found to be up-regulated in multiple human cancers (35–39). A recent study has shown that the ectopic expression of death effector domaincontaining DNA-binding protein (DEDD) in metastatic breast cancer cells MDA-MB-231 induces autophagy and increases degradation of the two major oncogenic transcription factors Snail and Twist through the autophagy–lysosome degradation system (40). In addition, p62 has been shown to bind with ubiquitinated proteins to deliver these proteins such as the protein Tau (41) to proteasome for degradation (30). Conversely, our study demonstrates that p62 inhibits Twist1 protein degradation, without affecting Snail, through both autophagy and proteasome. In autophagy-deficient cells, the resulted up-regulation of p62 binds with ubiquitinated Twist1 to reduce Twist1 binding with Rad23B and to thus inhibit Twist1 degradation through proteasome. This discrepancy may be due to (i) the different cell types or autophagy modulation investigated and (ii) the dependence on protein substrates, including Twist1. Indeed, previous studies have shown that p62 binds with ubiquitinated p53, thus inhibiting its proteasome degradation at least partially through reducing p53 binding with a proteasome shuttle protein p97 (26). Future experiments will elucidate the distinct role of p62 in regulating stability of different proteins. Multiple mechanisms may be involved in the effect of p62 interaction with polyubiquitinated Twist1 on Twist1 transcriptional activity. Interaction of p62 with polyubiquitinated Twist1 may increase nonubiquitinated Twist1 abundance through an equilibrium between these two forms of Twist1. It is also possible that p62–Twist1 interaction regulates the transcriptional activity of ubiquitinated Twist1. Further investigation is needed to elucidate the precise mechanism of regulation of Twist1 activity by p62. In addition to K175, other lysines including K76, K77, and K137 also have a role in Twist1 degradation, suggesting that other regulatory pathways regulate Twist1 stability and function by promoting modifications of one or a combination of two or more of these lysines. Furthermore, we found that the UBA domain of p62 is required for binding with Twist1 and for the p62/Twist1 interaction. The potentially distinct consequences of Twist1 binding with p62 and Rad23B that contain a UBA domain may be due to the different structures of p62 and Rad23B and be substrate- or Twist1 specific. Recent reports have implied an oncogenic role of p62 in several human cancers (42–44). In human skin SCCs, we found that accumulation of p62 is inversely correlated with E-cadherin abundance (Fig. 1E). Previous reports have shown that p62 promotes the activation of the NF-kappaB and NRF2 pathways in the pathogenesis of cancer (9–13). In addition, inhibition of selective autophagy has been demonstrated to cause sequestration of RhoA in the autophagosomes and deregulation of multiple cellular PNAS | June 24, 2014 | vol. 111 | no. 25 | 9245

MEDICAL SCIENCES

GAPDH

Con Twist1 p62 Twist1/p62

p62 mRNA

Tumor Volume (mm3)

HA Twist1

Tumor Volume (mm3)

HA-p62

A375

F

Twist1

Tumor Volume (mm3)

Cell Number (%OD)

H

G

Con p62

Con p62

Twist1 p62/ Twist1

E

Con p62 Twist1 Twist1/p62

Twist1 mRNA

GAPDH

C

p62/ Twist1

E-Cadherin-Luc

p62 Twist1

D

E-cad WT-Luc E-cad Ebox Mut-Luc

% Wound closed

B

A431

Twist1 p62 E-cadherin

# Lung nodules/mouse

A

Experimental Procedures

processes, including cell motility (45, 46). Here our findings indicate that the oncogenic role of p62 in cell proliferation and migration in vitro and in vivo can be mediated via regulating Twist1. In addition to p62-mediated Twist1 stabilization following autophagy inhibition, p62 is required for the up-regulation of Twist1 protein induced by EGF/TGF-β. The p62/Twist1 axis may also play an important role in Ras-driven cancers, including lung and pancreatic cancers, because Ras induces p62 expression (10). Future experiments will demonstrate the importance of the p62/ Twist1 interaction in cancer progression as well as morphogenesis. In addition to Twist1, recent studies have shown that selective autophagy also regulates RhoA signaling involving p62 (45, 46). Further investigations are under way in our laboratory to demonstrate the significance of autophagy/p62/Twist1 in tumorigenesis and tumor progression using clinically relevant genetic mouse models. In summary, we have demonstrated that the autophagy adaptor p62 promotes cell proliferation and migration in vitro and increases tumor growth and metastasis in mice through stabilizing the oncogenic protein Twist1. Our findings suggest that intervention of Twist1 stabilization through its interaction with p62 may be a promising therapeutic strategy for the prevention and treatment of cancer.

ACKNOWLEDGMENTS. We are grateful to Drs. Deborah Lang and Kay Macleod for their helpful suggestions, Dr. Noboru Mizushima for kindly providing WT and Atg5 KO MEFs, Dr. Tony Firuli for providing Myc-Twist1 pcDNA3.1 and Flag-Twist1 pcDNA3.1, Dr. Kimmelman for providing pLKO.1 shAtg7 (human), Dr. Masaaki Komatsu for providing the Atg7flox/flox mice, Terri Li for immunohistochemistry, and Dr. Ann Motten for a critical reading of the manuscript. This work was supported by the National Institutes of Health (NIH)/National Institute on Environmental Health Sciences Grant ES016936 (to Y.Y.H.), the American Cancer Society Grant RSG-13-078-01 (to Y.Y.H.), the University of Chicago Cancer Research Center (P30 CA014599), the CTSA (NIH UL1RR024999), and the University of Chicago Friends of Dermatology Endowment Fund.

1. Klionsky DJ (2007) Autophagy: From phenomenology to molecular understanding in less than a decade. Nat Rev Mol Cell Biol 8(11):931–937. 2. Mizushima N, Levine B, Cuervo AM, Klionsky DJ (2008) Autophagy fights disease through cellular self-digestion. Nature 451(7182):1069–1075. 3. Choi AM, Ryter SW, Levine B (2013) Autophagy in human health and disease. N Engl J Med 368(19):1845–1846. 4. White E (2012) Deconvoluting the context-dependent role for autophagy in cancer. Nat Rev Cancer 12(6):401–410. 5. Bjørkøy G, et al. (2005) p62/SQSTM1 forms protein aggregates degraded by autophagy and has a protective effect on huntingtin-induced cell death. J Cell Biol 171(4):603–614. 6. Pankiv S, et al. (2007) p62/SQSTM1 binds directly to Atg8/LC3 to facilitate degradation of ubiquitinated protein aggregates by autophagy. J Biol Chem 282(33):24131–24145. 7. Moscat J, Diaz-Meco MT (2009) p62 at the crossroads of autophagy, apoptosis, and cancer. Cell 137(6):1001–1004. 8. Moscat J, Diaz-Meco MT (2012) p62: A versatile multitasker takes on cancer. Trends Biochem Sci 37(6):230–236. 9. Mathew R, et al. (2009) Autophagy suppresses tumorigenesis through elimination of p62. Cell 137(6):1062–1075. 10. Duran A, et al. (2008) The signaling adaptor p62 is an important NF-kappaB mediator in tumorigenesis. Cancer Cell 13(4):343–354. 11. Inami Y, et al. (2011) Persistent activation of Nrf2 through p62 in hepatocellular carcinoma cells. J Cell Biol 193(2):275–284. 12. Komatsu M, et al. (2010) The selective autophagy substrate p62 activates the stress responsive transcription factor Nrf2 through inactivation of Keap1. Nat Cell Biol 12(3): 213–223. 13. Lau A, et al. (2010) A noncanonical mechanism of Nrf2 activation by autophagy deficiency: Direct interaction between Keap1 and p62. Mol Cell Biol 30(13):3275–3285. 14. Yang J, Weinberg RA (2008) Epithelial-mesenchymal transition: At the crossroads of development and tumor metastasis. Dev Cell 14(6):818–829. 15. Eckert MA, et al. (2011) Twist1-induced invadopodia formation promotes tumor metastasis. Cancer Cell 19(3):372–386. 16. Yang J, et al. (2004) Twist, a master regulator of morphogenesis, plays an essential role in tumor metastasis. Cell 117(7):927–939. 17. Franco HL, Casasnovas J, Rodríguez-Medina JR, Cadilla CL (2011) Redundant or separate entities?—roles of Twist1 and Twist2 as molecular switches during gene transcription. Nucleic Acids Res 39(4):1177–1186. 18. Lander R, Nordin K, LaBonne C (2011) The F-box protein Ppa is a common regulator of core EMT factors Twist, Snail, Slug, and Sip1. J Cell Biol 194(1):17–25. 19. Kabeya Y, et al. (2000) LC3, a mammalian homologue of yeast Apg8p, is localized in autophagosome membranes after processing. EMBO J 19(21):5720–5728. 20. Klionsky DJ, et al. (2008) Guidelines for the use and interpretation of assays for monitoring autophagy in higher eukaryotes. Autophagy 4(2):151–175. 21. Bolós V, et al. (2003) The transcription factor Slug represses E-cadherin expression and induces epithelial to mesenchymal transitions: A comparison with Snail and E47 repressors. J Cell Sci 116(Pt 3):499–511. 22. Batlle E, et al. (2000) The transcription factor snail is a repressor of E-cadherin gene expression in epithelial tumour cells. Nat Cell Biol 2(2):84–89. 23. Vesuna F, van Diest P, Chen JH, Raman V (2008) Twist is a transcriptional repressor of Ecadherin gene expression in breast cancer. Biochem Biophys Res Commun 367(2):235–241. 24. Gravdal K, Halvorsen OJ, Haukaas SA, Akslen LA (2007) A switch from E-cadherin to N-cadherin expression indicates epithelial to mesenchymal transition and is of strong and independent importance for the progress of prostate cancer. Clin Cancer Res 13(23):7003–7011.

25. Itakura E, Mizushima N (2011) p62 Targeting to the autophagosome formation site requires self-oligomerization but not LC3 binding. J Cell Biol 192(1):17–27. 26. Korolchuk VI, Mansilla A, Menzies FM, Rubinsztein DC (2009) Autophagy inhibition compromises degradation of ubiquitin-proteasome pathway substrates. Mol Cell 33(4):517–527. 27. Hiyama H, et al. (1999) Interaction of hHR23 with S5a. The ubiquitin-like domain of hHR23 mediates interaction with S5a subunit of 26 S proteasome. J Biol Chem 274(39):28019–28025. 28. Chen L, Madura K (2002) Rad23 promotes the targeting of proteolytic substrates to the proteasome. Mol Cell Biol 22(13):4902–4913. 29. Farmer LM, et al. (2010) The RAD23 family provides an essential connection between the 26S proteasome and ubiquitylated proteins in Arabidopsis. Plant Cell 22(1):124–142. 30. Seibenhener ML, et al. (2004) Sequestosome 1/p62 is a polyubiquitin chain binding protein involved in ubiquitin proteasome degradation. Mol Cell Biol 24(18):8055–8068. 31. Laursen KB, Mielke E, Iannaccone P, Füchtbauer EM (2007) Mechanism of transcriptional activation by the proto-oncogene Twist1. J Biol Chem 282(48):34623–34633. 32. Onder TT, et al. (2008) Loss of E-cadherin promotes metastasis via multiple downstream transcriptional pathways. Cancer Res 68(10):3645–3654. 33. Kao YC, et al. (2010) Downregulation of thrombomodulin, a novel target of Snail, induces tumorigenesis through epithelial-mesenchymal transition. Mol Cell Biol 30(20):4767–4785. 34. Kiyono K, et al. (2009) Autophagy is activated by TGF-beta and potentiates TGF-betamediated growth inhibition in human hepatocellular carcinoma cells. Cancer Res 69(23):8844–8852. 35. Puisieux A, Valsesia-Wittmann S, Ansieau S (2006) A twist for survival and cancer progression. Br J Cancer 94(1):13–17. 36. Ansieau S, et al. (2008) Induction of EMT by twist proteins as a collateral effect of tumor-promoting inactivation of premature senescence. Cancer Cell 14(1):79–89. 37.  Sošic D, Richardson JA, Yu K, Ornitz DM, Olson EN (2003) Twist regulates cytokine gene expression through a negative feedback loop that represses NF-kappaB activity. Cell 112(2):169–180. 38. Cheng GZ, et al. (2008) Twist is transcriptionally induced by activation of STAT3 and mediates STAT3 oncogenic function. J Biol Chem 283(21):14665–14673. 39. Rodrigues CO, Nerlick ST, White EL, Cleveland JL, King ML (2008) A Myc-Slug (Snail2)/ Twist regulatory circuit directs vascular development. Development 135(11):1903–1911. 40. Lv Q, et al. (2012) DEDD interacts with PI3KC3 to activate autophagy and attenuate epithelial-mesenchymal transition in human breast cancer. Cancer Res 72(13): 3238–3250. 41. Babu JR, Geetha T, Wooten MW (2005) Sequestosome 1/p62 shuttles polyubiquitinated tau for proteasomal degradation. J Neurochem 94(1):192–203. 42. Inoue D, et al. (2012) Accumulation of p62/SQSTM1 is associated with poor prognosis in patients with lung adenocarcinoma. Cancer Sci 103(4):760–766. 43. Thompson HG, Harris JW, Wold BJ, Lin F, Brody JP (2003) p62 overexpression in breast tumors and regulation by prostate-derived Ets factor in breast cancer cells. Oncogene 22(15):2322–2333. 44. Rolland P, et al. (2007) The ubiquitin-binding protein p62 is expressed in breast cancers showing features of aggressive disease. Endocr Relat Cancer 14(1):73–80. 45. Belaid A, et al. (2013) Autophagy plays a critical role in the degradation of active RHOA, the control of cell cytokinesis, and genomic stability. Cancer Res 73(14): 4311–4322. 46. Belaid A, et al. (2014) Autophagy and SQSTM1 on the RHOA(d) again: Emerging roles of autophagy in the degradation of signaling proteins. Autophagy 10(2):201–208.

9246 | www.pnas.org/cgi/doi/10.1073/pnas.1322913111

All human specimens were studied after approval by the University of Chicago Institutional Review Board. All animal procedures have been approved by the University of Chicago Institutional Animal Care and Use Committee. WT, Atg5 KO MEF cells [obtained from Noboru Mizushima (the University of Tokyo, Tokyo)], Atg3 KO, Atg7 KO, Atg9 KO, Atg12 KO, p62 KO, control, and Atg14 cKO MEF cells, doxycycline-inducible Atg12 knockdown (KD) 4T1 cells (murine mammary epithelial tumor cells), HeLa (human cervical cancer cells), HEK-293T (human embryonic kidney cell), A431 (human squamous carcinoma cells), HaCaT (human keratinocytes and epithelial cells, kindly provided by Norbert Fusenig [German Cancer Research Center (DKFZ), Heidelberg, Germany]), and A375 (human amelanotic melanoma cells) were used. Detailed descriptions of all methods are provided in SI Experimental Procedures.

Qiang et al.

Supporting Information Qiang et al. 10.1073/pnas.1322913111 SI Experimental Procedures Human Skin Tumor Samples. All human specimens were studied after approval by the University of Chicago Institutional Review Board. Formalin-fixed, paraffin-embedded tissue blocks were obtained from the archives in the tissue bank of the Department of Medicine, Section of Dermatology, University of Chicago. Non–sun-exposed nonlesional normal epidermis, actinic keratosis and squamous cell carcinoma tissues were used for immunohistochemical analysis of epithelial (E)-cadherin and p62 protein levels. Cell Culture. WT, Atg5 KO mouse embryonic fibroblast (MEF) cells (obtained from Dr. Mizushima), Atg3 KO, Atg7 KO, Atg9 KO, Atg12 KO, p62 KO, control, and Atg14 cKO MEF cells, doxycyclineinducible Atg12 knockdown (KD) 4T1 cells, HaCaT (human keratinocytes, kindly provided by Nobert Fusenig), HEK-293T (human embryonic kidney cell), A431 (human squamous carcinoma cells), and A375 (human amelanotic melanoma cells) were maintained in a monolayer culture in 95% air/5% CO2 (vol/vol) at 37 °C in DMEM supplemented with 10% FBS, 100 units/mL penicillin, 100 μg/mL streptomycin (Invitrogen), and 1% nonessential amino acids (Invitrogen). For the epithelial–mesenchymal transition experiment, cells were washed with PBS twice and incubated with DMEM supplemented with EGF (10 ng/mL, R&D) and TGF-β (10 ng/mL, R&D) for 48 h. siRNA and Plasmid Transfection. MEF cells were transfected with negative control (NC) or siRNA (ON-TARGETplus SMARTpool; Dharmacon) targeting Twist1 or p62, using DharmaFECT 4 transfection reagent (Dharmacon) according to the manufacturer’s instructions as described previously (1). HEK293T, and MEF cells (except for p62 KO MEF cells) were transfected with vector, Myc-Twist1, Flag-Twist1, HA-p62, HA-ubiquitin, p62, or Twist1 mutants, E-cad (108) WT-Luc, and E-cad (108) E-box Mut-Luc using X-tremeGENE 9 (Roche) as described previously (2–4). A431 and A375 cells were transfected using Amaxa Nucleofector according to the manufacturer’s instructions as described previously (2, 3). Reconstitution of Atg7 and p62 and knockdown by shRNA used lentiviral vectors, with HEK293T cells used as packaging cells for the lentiviral system. pLKO.1 vectors and packaging mix (psPAX2 and pMG2) were transfected into HEK293T cells. Two days after transfection, the supernatants were collected and diluted with the same volume of fresh complete DMEM, and Polybrene was added to the infection medium at 8 μg/mL. The infection mixtures were added to cell cultures seeded on a six-well plate, incubated overnight, and then replaced with fresh complete DMEM containing 8 μg/mL puromycin. The puromycin-resistant cells were isolated and propagated for use in the experiments. Plasmids. Myc-Twist1 pcDNA3.1 and Flag-Twist1 pcDNA3.1 were kindly provided by Tony Firuli. We cloned the Myc-Twist1 (HindIII/ XbaI) from pcDNA3.1 vector to pMPB3 vector (modified piggyBac vector). HA-p62 pcDNA4 was obtained from Qing Zhong (University of California, Berkeley, Berkeley, CA) (Addgene plasmid 28027) (5). HA-Ubiquitin pcDNA3 was obtained from Edward T. H. Yeh (The University of Texas-Houston Health Science Center, Houston) (Addgene plasmid 18712) (6). PGL2 E-cad (108) WTLuc and PGL2 E-cad (108) E-box Mut-Luc were obtained from Eric R. Fearon (University of Michigan Medical School, Ann Arbor, MI) (Addgene plasmids 19291 and 19290) (7). The transcription factor TCF/lymphoid enhancer factor luciferase reporter Qiang et al. www.pnas.org/cgi/content/short/1322913111

containing TCF binding sites TOPFLASH (TOP) and its negative control plasmid containing inactive TCF binding sites FOPFLASH (FOP) plasmids were from Tong-Chuan He. MycFlag-Rad23b-pCMV, Flag-Twist2, and pCMV6 vector were obtained from OriGene Technologies. pLKO.1 shp62 (human) was obtained from Sigma. pLKO.1 shp62 (mouse) and pLKO.1 shTwist1 (mouse) were obtained from Dharmacon. pENTER vector, pHAGE con, and pHAGE Atg7 (mouse) were obtained from Seungmin Hwang. pLenti CMV Dest vector was obtained from Addgene. pLKO.1 shAtg7 (human) was obtained from Dr. Kimmelman. DNA Constructs. Twist1 deletion mutants (Myc-Twist1-WR) were

generated using the following primers: N-terminal sense 5′TACGACTCACTATAGGGAGACCC-3′ and C-terminal sense 5′-GTTCTAGACTAGCAGCTTGCCATCTTGGAGTCC-3′ from Myc-Twist1 pcDNA3.1. They were then subcloned (HindIII/ XbaI) into pMPB3 vector. p62 deletion mutants (primers A–F) were generated using the primers C-terminal sense 5′GGAATTCTATGGTGCACCCCAATGTGATCTG-3′ and Nterminal primer A 5′-GGAATTCTATGGTGCACCCCAATGTGATCTG-3′, primer B 5′-GGAATTCTATGGGTCCACCAGGAAACTGGA-3′ and primer C 5′-GGAATTCTATGGAGTCGGATAACTGTTCAGG-3′; N-terminal sense 5′-GGAATTCTATGGCGTCGCTCACCGTGAA-3′ and C-terminal primer D 5′-GAGTGCGGCCGCACGGGTCCACTTCTTTTGAAG3′, primer E 5′-GAGTGCGGCCGCAGCTTGGCCCTTCGGATTCT-3′ and primer F 5′-GAGTGCGGCCGCGGCTTCTTTTCCCTCCGTGCT-3′ and subcloned (EcoR1/Not1) back into the HA-pcDNA4 vector. HA-p62 WT was generated using the primers N-terminal sense 5′-GTGGTACCGCTATGGCGTCGCTCACCGTGAA-3′ and C-terminal sense 5′-GAGGCGCGCCCTAAGCGTAATCTGGAACATCGT-3′ from HA-p62 pcDNA4 and subcloned (Kpn1/Asc1) into pMPB3 vector and pENTER vector or Myc-Twist1-pMPB3. Establishment of a stable transfected cell line using the piggyBac transposon system (pMPB3) was performed as described previously (8). pENTER p62 was recombined into pLenti CMV Dest vector using Gateway LR Clonase Enzyme Mix kit following the manufacturer’s instructions. All constructs were confirmed by sequencing. Site-Directed Mutagenesis. Mutations of lysine to arginine (K→R) at positions 33, 38, 73, 76, 77, 137, 146, 149, 154, and 175 of wildtype Myc-Twist1 pMPB3 plasmid were carried out using the QuikChange XL Site-Directed Mutagenesis kit following the manufacturer’s instructions (Stratagene). The amino acid changes were introduced into the plasmid by using the following primers: K33R 5′-CCGGCGAGCGGCAGGCGCGGG-3′ and K33R antisense 5′-CCCGCGCCTGCCGCTCGCCGG-3′; K38R 5′GCGGGGCTCGCAGGAGACGCAGCAG-3′ and K38R antisense 5′-CTGCTGCGTCTCCTGCGAGCCCCGC-3′; K73R 5′CCGGCCCAGGGCAGGCGCGGC-3′ and K73R antisense 5′GCCGCGCCTGCCCTGGGCCGG-3′; K76R 5′-GGCAAGCGCGGCAGGAAATCTGCGGGC-3′ and K76R antisense 5′GCCCGCAGATTTCCTGCCGCGCTTGCC-3′; K77R 5′-AGCGCGGCAAGAGATCTGCGGGCGG-3′ and K77R antisense 5′-CCGCCCGCAGATCTCTTGCCGCGCT-3′; K137R 5′-CCGCCCTGCGCAGGATCATCCCCAC-3′ and K137R antisense 5′-GTGGGGATGATCCTGCGCAGGGCGG-3′; K146R 5′CACGCTGCCCTCGGACAGGCTGAGCAAG-3′ and K146R antisense 5′-CTTGCTCAGCCTGTCCGAGGGCAGCGTG-3′; 1 of 6

K149R 5′-CTCGGACAAGCTGAGCAGGATTCAGACCCTCAAAC-3′ and K149R antisense 5′-GTTTGAGGGTCTGAATCCTGCTCAGCTTGTCCGAG-3′; K154R 5′-AGCAAGATTCAGACCCTCAGACTGGCGGCC-3′ and K154R antisense 5′-GGCCGCCAGTCTGAGGGTCTGAATCTTGCT-3′; K175R 5′-CGAGCTGGACTCCAGGATGGCAAGCTGCA-3′ and k175R antisense 5′-TGCAGCTTGCCATCCTGGAGTCCAGCTCG3′; all mutants were confirmed by sequencing. Immunofluorescence. Immunofluorescence was done as described previously (1, 9). Briefly, the cells attached to the coverslips were washed three times with PBS and fixed with 4% paraformaldehyde in PBS solution for 25 min. The cells were then permeabilized with cold 0.5% Triton X-100 in PBS for 20 min at 4 °C and washed with cold PBS. PBS supplemented with 5% normal goat serum (Invitrogen) was used as a blocking solution (30 min, 37 °C). After removal of the blocking solution, the cells were incubated at 4 °C overnight in primary rabbit anti–E-cadherin (1:200; Cell Signaling Biotechnology), β-catenin (1:200 BD Biosciences), HA (1:200 Cell Signaling Biotechnology), Myc (1:200 Cell Signaling Biotechnology), LC3 (1:200 Cell Signaling Biotechnology), Twist1 (1:200 Abcam), and p62 (1:200 Progene Biotechnik) monoclonal antibody. After washing with PBS, the cells were incubated at 37 °C for 1 h with Alexa Fluor 488 F (ab′) two fragments of goat anti-mouse IgG antibodies and Alexa Fluor 568 of goat anti-rabbit IgG antibodies. The cells were then fixed in Prolong Gold Antifade with DAPI (Invitrogen) to visualize the cell nuclei, and observed under a fluorescence microscope (Olympus IX71) with a peak excitation wavelength of 340 nm. Immunoprecipitation and GST-Pulldown Assay. Immunoprecipitation was performed as described previously by using anti–HA-Tag and anti–Myc-Tag antibodies (Cell Signaling Technology), an anti-p62 antibody (Sigma), and anti-Twist1 antibody (Abcam) (1). For ubiquitin analysis, cells were treated with the proteasome inhibitor MG132 (10 μM) for 6 h before collection and Nethylmaleimide (NEM, 5 mM) was added to all of the buffers. GST-P62 recombinant protein was obtained from Enzo Life Sciences, and GST (control, Con) recombinant protein was obtained from Sigma. The GST-pulldown assay was performed using Pierce GST Protein Interaction Pull-Down kit following the manufacturer’s instructions. WT and Atg5 KO cells were treated with MG132 (10 μM) for 6 h before collection and NEM (5 mM) was added to all of the buffers. Western Blotting. Western blotting was performed as described

previously (10). Antibodies used were as follows: p53, Rad23b, GAPDH, and HA-Tag (mouse) (Santa Cruz), E-cadherin, LC3I/II, HA-Tag (rabbit), Myc-Tag (mouse), Myc-Tag (rabbit), Snail, Slug, Atg7, Flag-Tag, N-cadherin, ZEB1 (Cell Signaling Technology), p62 (Progene Biotechnik), and Twist1 (Abcam). Protein levels were quantified using ImageJ software and normalized so that the protein level at t = 0 was 100%. The log10 of the percentage of the protein level was plotted versus time, and the t1∕2 was calculated from the log10 of 50%. Luciferase Reporter Assays. PGL2 E-cad WT-Luc, PGL2 E-cad E-box Mut-Luc (obtained from Eric R. Fearon, Addgene plasmids 19291 and 19290), and 0.025 μg of pRL-TK (Promega, used as a transfection efficiency control) were transfected with X-treme 9 Transfection Reagent (Roche) or Amaxa Nucleofector according to the manufacturer’s protocols. The luciferase activity was measured as described previously (2). Immunohistochemical Analysis. E-cadherin and Ki67 levels were determined using immunohistochemical analysis by the Immunohistochemistry core facility (University of Chicago). E-cadherin Qiang et al. www.pnas.org/cgi/content/short/1322913111

(Cell Signaling) and Ki67 (Cell Signaling) antibodies were used, with the protein levels visualized with the diaminobenzidine (DAB) method (brown color). To exclude the contribution of endogenous brown pigmentation due to melanin, we also performed hematoxylin and eosin (H&E) staining. Real-Time PCR. Quantitative real-time PCR assays were performed using ABI7300 (Applied Biosystems) in 96-well plates with the SYBR Green PCR Master Mix (Applied Biosystems) as in our previous studies (11). The threshold cycle number (CT) for each sample was determined in triplicate. The CT for values for E-cadherin and Twist1 were normalized against GAPDH as described previously (2, 3, 11). Amplification primers were 5′CAGGTCTCCTCATGGCTTTGC-3′ (forward), 5′-CTTCCGAAAAGAAGGCTGTCC-3′ (reverse) for the mouse E-cadherin gene; 5′-AGGTCGGTGTGAACGGATTTG-3′ (forward) and 5′TGTAGACCATGTAGTTGAGGTCA-3′ (reverse) for GAPDH; 5′-CAGAGAAGCCCATGGACAG-3′ (forward) and 5′-AGCTGCCTTGTACCCACATC-3′ (reverse) for the human p62 gene; 5′-CGGGAGTCCGCAGTCTTA-3′ (forward) and 5′-GCTTGAGGGTCTGAATCTTG-3′ (reverse) for the human Twist1 gene; and 5′-GCAGGACGTGTCCAGCTC-3′ (forward) and 5′-CTGGCTCTTCCTCGCTGTT-3′ (reverse) for the mouse Twist1 gene. The CT for each sample was determined in triplicate. The CT values for E-cadherin and Twist1 were normalized against GAPDH as described previously (2, 3, 11). Animal Treatments. All animal procedures have been approved by the University of Chicago Institutional Animal Care and Use Committee. K14-Cre mice were obtained from The Jackson Laboratories and Atg7flox/flox mice were a gift from Masaaki Komatsu (Tokyo Metropolitan Institute of Medical Science, Tokyo). Analysis of Tumor Growth and Metastasis in Nude Mice. All animal procedures have been approved by the University of Chicago Institutional Animal Care and Use Committee. Athymic nude mice were obtained from Harlan Sprague–Dawley. A total of 1 × 106 A431-Con, A431-p62, A431-Twist1 and A431-Twist1p62, and A375-Con and A375-p62 cells were injected s.c. into the right flank of 6-wk-old female nude mice. Tumors were monitored weekly and tumor volume (TV) was calculated using the following formula: TV (mm3) = d2 × D/2, where d and D are the shortest and the longest diameters, respectively. For metastasis analysis, cells were injected via tail vein as described previously (12). Lungs and other tissues with metastasis were harvested for metastasis analysis. Tumors were fixed in formalin for histological analysis or immunohistochemical analysis for E-cadherin and Ki67-positive cells (Immunohistochemistry core facility). Migration and Invasion Assay. For 2D migration assays, cells were plated onto Radius 24-Well Cell Migration Assay plates according to the manufacturer’s instructions (Cell Biolabs). To analyze the invasion activity of WT, Atg3 KO, Atg5 KO, Atg9 KO, and Atg12 KO MEF cells, the upper surface of the Transwell filters (8-μm pore size Millipore MultiScreen plates) was coated with Matrigel as described previously (13). For 2D migration assays, cells were plated into a Radius 24-Well Cell Migration Assay plate (Cell Biolabs). Each well contains a circular 680-μm diameter “gel spot” to which cells do not attach. Before the experiment, cells were confluent and quiescent. At the start of the experiment, the gel spot was removed within 5 min, and cells then populated the circular void space. Digital images of the gap closure were taken with an inverted microscope (Olympus IX71) with a 10× phase lens. Images were analyzed with CellProfiler cell image analysis software. To analyze the invasion activity of WT, Atg3 KO, Atg5 KO, Atg9 KO, and Atg12 KO MEF cells, the upper surface of the Transwell filters 2 of 6

(8-μm pore size Millipore MultiScreen-MIC plates) was coated with Matrigel. Cells suspended in 60 μL serum-free media (5 × 104) were then added to the chamber, and the chamber was placed in a 96-well plate containing complete medium. After a 24-h incubation at 37 °C, the filters were gently taken out and Matrigel on the upper surface of the filters was removed by cotton swabs. Cells on the undersides of Transwell filters were fixed with 4% paraformaldehyde for 30 min, stained with 0.1% crystal violet for 10 min, and then photographed. The extent of cell migration was quantified as the number of migrated cells per field as described (13).

In Vitro Cell Proliferation Assay. Cell proliferation of MEF cells was analyzed using the CellTiter 96 AQueous Non-Radioactive Cell Proliferation Assay (MTS) (Promega) according to the manufacturer’s instructions as in our recent studies (4).

1. Qiang L, Wu C, Ming M, Viollet B, He YY (2013) Autophagy controls p38 activation to promote cell survival under genotoxic stress. J Biol Chem 288(3):1603–1611. 2. Ming M, et al. (2010) Regulation of global genome nucleotide excision repair by SIRT1 through xeroderma pigmentosum C. Proc Natl Acad Sci USA 107(52):22623–22628. 3. Han W, et al. (2012) Nrf1 CNC-bZIP protein promotes cell survival and nucleotide excision repair through maintaining glutathione homeostasis. J Biol Chem 287(22): 18788–18795. 4. Wu CL, et al. (2013) Role of AMPK in UVB-induced DNA damage repair and growth control. Oncogene 32(21):2682–2689. 5. Fan W, et al. (2010) Keap1 facilitates p62-mediated ubiquitin aggregate clearance via autophagy. Autophagy 6(5):614–621. 6. Kamitani T, Kito K, Nguyen HP, Yeh ET (1997) Characterization of NEDD8, a developmentally down-regulated ubiquitin-like protein. J Biol Chem 272(45):28557–28562. 7. Hajra KM, Chen DY, Fearon ER (2002) The SLUG zinc-finger protein represses E-cadherin in breast cancer. Cancer Res 62(6):1613–1618.

8. Ding S, et al. (2005) Efficient transposition of the piggyBac (PB) transposon in mammalian cells and mice. Cell 122(3):473–483. 9. Qiang L, et al. (2012) HIF-1α is critical for hypoxia-mediated maintenance of glioblastoma stem cells by activating Notch signaling pathway. Cell Death Differ 19(2):284–294. 10. Qiang L, et al. (2009) Isolation and characterization of cancer stem like cells in human glioblastoma cell lines. Cancer Lett 279(1):13–21. 11. Han W, Ming M, He TC, He YY (2010) Immunosuppressive cyclosporin A activates AKT in keratinocytes through PTEN suppression: Implications in skin carcinogenesis. J Biol Chem 285(15):11369–11377. 12. Zhao J, et al. (2008) Inhibition of alpha(4) integrin mediated adhesion was involved in the reduction of B16-F10 melanoma cells lung colonization in C57BL/6 mice treated with gambogic acid. Eur J Pharmacol 589(1-3):127–131. 13. Tang Y, et al. (2009) TGF-beta1-induced migration of bone mesenchymal stem cells couples bone resorption with formation. Nat Med 15(7):757–765.

Qiang et al. www.pnas.org/cgi/content/short/1322913111

Statistical Analyses. Statistical analyses were performed using Prism 5 (GraphPad). Data were expressed as the mean of at least three independent experiments and analyzed by Student t test. Error bars indicate the SDs of the means. P < 0.05 was considered statistically significant.

3 of 6

Atg5 KO

WT 3

CHX

6

12

6

12

3

6 12(h)

E-cadherin p27 GAPDH Atg5 KO

WT 3

MG132 E-cadherin

3

6 12(h)

WT Atg5 KO

p27 GAPDH Atg3

Atg9

Atg12

Atg14

Atg12

p62

WT KO WT KO WT KO Con cKO Con KD WT KO Twist1 p62 LC3-I LC3-II GAPDH

p62

E-cadherin

WT

Twist1

Con E-cadherin Twist1

Atg7 cKO

WT Atg5 KO

GAPDH

Fig. S1. (Related to Fig. 2.) Autophagy regulates Twist1 protein degradation involving both autophagosome and proteasome. (A) Immunoblot analysis of E-cadherin and GAPDH in WT and Atg5 KO MEF cells treated with cycloheximide (CHX, 100 μg/mL) or MG132 (10 μM) for the indicated times as in Fig. 2 E and F. (B) Real-time RT-PCR analysis of E-cadherin mRNA levels in WT and Atg5 KO MEF cells. (C) Luciferase reporter assay of the TCF/lymphoid enhancer factor luciferase reporter (TOP) and its negative control (FOP) in WT and Atg5 KO MEF cells. The results were obtained from three independent experiments [mean ± SD (error bars), n = 3; *P < 0.05; **P < 0.01, compared with the Con group). (D) Immunoblot analysis of Twist1, p62, LC3-I/II, and GAPDH in WT and Atg3, Atg9, Atg12, and p62 KO MEF cells; Con and Atg14 cKO MEF cells; and Con and Dox-inducible Atg12 knockdown (KD) 4T1 cells. (E) Real-time RT-PCR analysis of Twist1 in WT and Atg5 KO MEF cells. (F) Immunoblot analysis of E-cadherin, Twist1, and GAPDH in WT MEF cells transfected with vector control (Con), FlagTwist1, or Myc-Twist1. (G) Quantification of the Twist1 levels in Fig. 2E using ImageJ. H, Representative immunohistochemical analysis of Twist1, p62 and E-cadherin protein (brown) in the skin of WT and Atg7 cKO mice. Scale bar, 50 μm. The results were obtained from three independent experiments (mean ± SD, n = 3; *P < 0.05 compared with the WT MEF; #P < 0.05 compared with the WT E-cadherin promoter).

Qiang et al. www.pnas.org/cgi/content/short/1322913111

4 of 6

Flag-Twist2 0 3 6 24 0

CHX

Flag-Twist2 HA-p62 3 6 24 (h)

Flag HA GAPDH

HA-p62 Myc-Twist1 3 6 12

Myc-Twist1 3 6 12

CHX

Myc-Twist- WR 3 6 12 (h)

HA Myc GAPDH

HA-p62 Myc-Twist1

Myc-Twist1 3

MG132

6

12

3

6

Myc-Twist- WR 12

3

6

12 (h)

HA Myc GAPDH

Fig. S2. (Related to Fig. 4.) p62 binds to Twist1 through its ubiquitin-associated domain. (A) Immunoblot analysis of HA, Flag, and GAPDH in 293T cells transfected with Flag-Twist2 alone or in combination with HA-p62 for 48 h, and then treated with CHX (100 μg/mL) for the indicated times. The results were obtained from three independent experiments. (B and C) Immunoblot analysis of HA and Myc in 293T cells transfected with Myc-Twist1, a combination of MycTwist1 and HA-p62, or Myc-Twist1-ΔWR for 48 h, and then treated with CHX (100 μg/mL) (B) or MG132 (10 μM) (C) for the indicated times. (D) Quantification of B using ImageJ.

Mut1, K73-76-77-175R; Mut2, K137-175R; Mut3, K146-149-175R; Mut4, K154-175R; Mut5, K137-154-175R; Mut6, K146-154-175R; Mut7, K137-146-175R

Myc-Twist1 CHX 0

6

12

Mut1 0

Mut2

6

12

0

12

0

6

Mut4

Mut3 12

0

12

0

6

12

0

6

Mut5 12

0

6

Mut7

Mut6 12 0

6

12

0

6 12(h)

Myc GAPDH

6

12

K76R

K73R

Myc-Twist1 CHX 0

0

6

6

K73-76-77-175R K137R

K77R 6

12

0

6

12

0

6

K175R 12 0

6

12

K137-175R 0

6 12(h)

Myc GAPDH

Fig. S3. (Related to Fig. 5.) Twist1 lysine 175 is critical for Twist1 ubiquitination, degradation, and binding with p62. (A) Schematic for comparison of amino acid sequences among human (Homo), mouse (Mus), rat (Rattus), and Xenopus. Lysine sites are marked by rectangles. “*” indicates the identical residues in all sequences in the alignment; “:” indicates conserved substitutions; “.” indicates semiconserved substitutions; and “-“ indicates gaps in the alignment. (B) Quantification of Fig. 5B using ImageJ. (C) Immunoblot analysis of Myc and GAPDH in 293T cells transfected with Myc-Twist1 (K→R) mutation constructs (Mut1, K73-76-77-175R; Mut2, K137-175R; Mut3, K146-149-175R; Mut4, K154-175R; Mut5, K137-154-175R; Mut6, K146-154-175R; and Mut7, K137-146-175R), and then treated with CHX (100 μg/mL) for the indicated times. K73-76-77-175R, Twist1 mutant with K→R mutations for all the K73, K76, K77, and K175 sites. We used a similar format to denote other Twist1 mutants with K-R mutations for more than one site. (D) Immunoblot analysis of Myc and GAPDH in 293T cells transfected with Myc-Twist1 or constructs with mutations of K73R, K76R, K77R, K73-76-77-175R, K137R, K175R, or K137-175R for 48 h, and then treated with CHX (100 μg/mL) for the indicated times. The results were obtained from three independent experiments.

Qiang et al. www.pnas.org/cgi/content/short/1322913111

5 of 6

-Catenin

DAPI

24h

48h

EGF+TGF-

Twist1-p62

A431-p62

A431-Twist1

A431-Twist1-p62

HE

E-Cadherin

Ki67

A431-Con

shp62

Twist1

shGFP

p62

Control

0h shGFP

E-cadherin

Control

Merge

Fig. S4. (Related to Fig. 6.) p62 promotes tumor cell growth and metastasis in vivo through regulating Twist1. (A) Immunofluorescence assay of β-catenin in WT and Atg5 KO MEF cells. (Scale bar, 50 μm.) (B) Histological H&E staining and immunohistochemical analysis of E-cadherin and Ki67 (brown) in A431-Con, A431-p62, A431-Twist1, and A431-Twist1-p62 tumors. (Scale bar, 200 μm and 50 μm for Left and Right, respectively.) (C) Morphology of HaCaT human epithelial cells transfected with vector or shRNA target p62 (shp62), following treatment with control vehicle or the combination of EGF (10 ng/mL) and TGF-β (10 ng/mL) for 48 h. (Scale bar, 50 μm.)

Qiang et al. www.pnas.org/cgi/content/short/1322913111

6 of 6