10
Zyxin promotes colon cancer tumorigenesis in a mitotic phosphorylation-dependent manner and through CDK8-mediated YAP activation Jiuli Zhou a,b,c , Yongji Zeng a,b,c , Lian Cui d , Xingcheng Chen a,b,c , Seth Stauffer a,b,c , Zhan Wang a,b , Fang Yu e , Subodh M. Lele c , Geoffrey A. Talmon c , Adrian R. Black a,b , Yuanhong Chen a,b , and Jixin Dong a,b,1 a Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198; b Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198; c Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198; d Department of Dermatology, Shanghai Tenth Peoples Hospital, Tongji University School of Medicine, Shanghai 200072, China; and e Department of Biostatistics, College of Public Health, University of Nebraska Medical Center, Omaha, NE 68198 Edited by Kun-Liang Guan, University of California at San Diego, La Jolla, CA, and accepted by Editorial Board Member Rakesh K. Jain June 11, 2018 (received for review January 11, 2018) Zyxin is a member of the focal adhesion complex and plays a critical role in actin filament polymerization and cell motility. Several recent studies showed that Zyxin is a positive regulator of Yki/YAP (Yes-associated protein) signaling. However, little is known about the mechanisms by which Zyxin itself is regulated and how Zyxin affects HippoYAP activity. We first showed that Zyxin is phosphorylated by CDK1 during mitosis. Depletion of Zyxin resulted in significantly impaired colon cancer cell prolifera- tion, migration, anchorage-independent growth, and tumor for- mation in xenograft animal models. Mitotic phosphorylation is required for Zyxin activity in promoting growth. Zyxin regulates YAP activity through the colon cancer oncogene CDK8. CDK8 knockout phenocopied Zyxin knockdown in colon cancer cells, while ectopic expression of CDK8 substantially restored the tumor- igenic defects of Zyxin-depletion cells. Mechanistically, we showed that CDK8 directly phosphorylated YAP and promoted its activa- tion. Fully activated YAP is required to support the growth in CDK8-knockout colon cancer cells in vitro and in vivo. Together, these observations suggest that Zyxin promotes colon cancer tu- morigenesis in a mitotic-phosphorylation-dependent manner and through CDK8-mediated YAP activation. Zyxin | CDK8 | HippoYAP pathway | mitosis | colon cancer Z yxin is a LIM domain protein concentrated at sites of cellcell adhesion, where it is proposed to dock proteins involved in cytoskeletal dynamics and signaling (1). Zyxin has also been implicated in sensing the mechanotransduction signal (2, 3). Of interest, Zyxin contains a nuclear export signal and shuttles be- tween the nucleus and sites of cell adhesion (4), although the underlying mechanisms and function of its localization dynamics are unclear. Several recent studies showed that Zyxin is a posi- tive regulator of tissue growth through HippoYorkie/Yki sig- naling in Drosophila (58). Zyxin regulates Yki-mediated tissue growth by promoting F-actin polymerization independent of Hippo/Warts kinases (5). This mechanism is conserved in mammalian cancer cells (9, 10). Furthermore, in human breast cancer cells, Zyxin promotes growth and tumorigenesis by mod- ulating Hippoyes-associated protein (YAP) signaling activity (11), which plays an important role in the development and pro- gression of many human malignancies (1215). Despite the growth-promoting role of Zyxin, however, little is known about the mechanisms by which Zyxin itself is regulated and how Zyxin af- fects HippoYAP (and/or other signaling) activity in cancer cells. The protein kinase cyclin-dependent kinase 8 (CDK8) is a component of the mediator complex that functions as a bridge between basal transcription machinery and gene-specific tran- scriptional factors (16). CDK8 is amplified and overexpressed in colon cancer and exerts its oncogenic activity partially through regulating β-catenin activity (17). The precise mechanisms by which CDK8 regulates β-catenin remain obscure. CDK8 mRNA is up-regulated in malignant melanoma by loss of a transcrip- tional repressor called the histone variant macroH2A, which functions as a tumor suppressor in melanoma (18). In addition, CDK8 protein levels are also controlled by S-phase kinase as- sociated protein 2 (Skp2)-mediated degradation of macroH2A1 protein, and these three proteins work together to regulate G2/M transition and tumorigenesis in breast cancer (19). CDK8 exerts its oncogenic function mainly through phosphorylation of substrates. Several substrates for CDK8 have been identified, including the Notch intracellular domain, SMAD complexes, E2F1, STAT1, and the C-terminal domain of RNA polymerase II (16). These studies highlight an important oncogenic function of CDK8 kinase activity. A connection between CDK8 and YAP, the critical transcriptional coactivator of Hippo signaling, has not been established. Here, we report that Zyxin promotes colon cancer cell growth, and its oncogenic activity is partially controlled by mitotic phosphory- lation. We further showed that Zyxin regulates YAP activity through CDK8 in colon cancer cells. In addition, we identified YAP as a direct substrate of CDK8 and CDK8-mediated phos- phorylation that promotes YAP activity in vitro and in vivo. Significance Zyxin is a member of the cellcell adhesion complex and con- trols cell cytoskeleton and motility. Analysis from clinical samples revealed that Zyxin is highly expressed in colon cancer compared with normal tissue, suggesting an oncogenic role for Zyxin in cancer. Depletion of Zyxin resulted in significantly impaired colon cancer cell proliferation, migration, cellular transformation, and tumor formation in xenograft animal models. We also showed that Zyxin is phosphorylated by CDK1 during mitosis. Mitotic phosphorylation is required for Zyxin activity in promoting colon cancer growth. Zyxin regu- lates YAP activity through the colon cancer oncogene CDK8. We further showed that CDK8 directly phosphorylates YAP and promotes its activation. These observations identify the ZyxinCDK8YAP axis as a potential therapeutic target in cancer. Author contributions: J.Z. and J.D. designed research; J.Z., Y.Z., L.C., X.C., S.S., Z.W., and Y.C. performed research; G.A.T. and A.R.B. contributed new reagents/analytic tools; J.Z., Y.Z., L.C., F.Y., S.M.L., G.A.T., A.R.B., Y.C., and J.D. analyzed data; and J.Z. and J.D. wrote the paper. The authors declare no conflict of interest. This article is a PNAS Direct Submission. K.-L.G. is a guest editor invited by the Editorial Board. Published under the PNAS license. 1 To whom correspondence should be addressed. Email: [email protected]. This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10. 1073/pnas.1800621115/-/DCSupplemental. Published online July 2, 2018. E6760E6769 | PNAS | vol. 115 | no. 29 www.pnas.org/cgi/doi/10.1073/pnas.1800621115 Downloaded by guest on January 25, 2020

Zyxin promotes colon cancer tumorigenesis in a …Zyxin promotes colon cancer tumorigenesis in a mitotic phosphorylation-dependent manner and through CDK8-mediated YAP activation Jiuli

  • Upload
    others

  • View
    6

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Zyxin promotes colon cancer tumorigenesis in a …Zyxin promotes colon cancer tumorigenesis in a mitotic phosphorylation-dependent manner and through CDK8-mediated YAP activation Jiuli

Zyxin promotes colon cancer tumorigenesis in a mitoticphosphorylation-dependent manner and throughCDK8-mediated YAP activationJiuli Zhoua,b,c, Yongji Zenga,b,c, Lian Cuid, Xingcheng Chena,b,c, Seth Stauffera,b,c, Zhan Wanga,b, Fang Yue,Subodh M. Lelec, Geoffrey A. Talmonc, Adrian R. Blacka,b, Yuanhong Chena,b, and Jixin Donga,b,1

aEppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198; bFred and Pamela Buffett CancerCenter, University of Nebraska Medical Center, Omaha, NE 68198; cDepartment of Pathology and Microbiology, University of Nebraska Medical Center,Omaha, NE 68198; dDepartment of Dermatology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China;and eDepartment of Biostatistics, College of Public Health, University of Nebraska Medical Center, Omaha, NE 68198

Edited by Kun-Liang Guan, University of California at San Diego, La Jolla, CA, and accepted by Editorial Board Member Rakesh K. Jain June 11, 2018 (receivedfor review January 11, 2018)

Zyxin is a member of the focal adhesion complex and plays acritical role in actin filament polymerization and cell motility.Several recent studies showed that Zyxin is a positive regulator ofYki/YAP (Yes-associated protein) signaling. However, little isknown about the mechanisms by which Zyxin itself is regulatedand how Zyxin affects Hippo–YAP activity. We first showed thatZyxin is phosphorylated by CDK1 during mitosis. Depletion ofZyxin resulted in significantly impaired colon cancer cell prolifera-tion, migration, anchorage-independent growth, and tumor for-mation in xenograft animal models. Mitotic phosphorylation isrequired for Zyxin activity in promoting growth. Zyxin regulatesYAP activity through the colon cancer oncogene CDK8. CDK8knockout phenocopied Zyxin knockdown in colon cancer cells,while ectopic expression of CDK8 substantially restored the tumor-igenic defects of Zyxin-depletion cells. Mechanistically, we showedthat CDK8 directly phosphorylated YAP and promoted its activa-tion. Fully activated YAP is required to support the growth inCDK8-knockout colon cancer cells in vitro and in vivo. Together,these observations suggest that Zyxin promotes colon cancer tu-morigenesis in a mitotic-phosphorylation-dependent manner andthrough CDK8-mediated YAP activation.

Zyxin | CDK8 | Hippo–YAP pathway | mitosis | colon cancer

Zyxin is a LIM domain protein concentrated at sites of cell–cell adhesion, where it is proposed to dock proteins involved

in cytoskeletal dynamics and signaling (1). Zyxin has also beenimplicated in sensing the mechanotransduction signal (2, 3). Ofinterest, Zyxin contains a nuclear export signal and shuttles be-tween the nucleus and sites of cell adhesion (4), although theunderlying mechanisms and function of its localization dynamicsare unclear. Several recent studies showed that Zyxin is a posi-tive regulator of tissue growth through Hippo–Yorkie/Yki sig-naling in Drosophila (5–8). Zyxin regulates Yki-mediated tissuegrowth by promoting F-actin polymerization independent ofHippo/Warts kinases (5). This mechanism is conserved inmammalian cancer cells (9, 10). Furthermore, in human breastcancer cells, Zyxin promotes growth and tumorigenesis by mod-ulating Hippo–yes-associated protein (YAP) signaling activity(11), which plays an important role in the development and pro-gression of many human malignancies (12–15). Despite thegrowth-promoting role of Zyxin, however, little is known about themechanisms by which Zyxin itself is regulated and how Zyxin af-fects Hippo–YAP (and/or other signaling) activity in cancer cells.The protein kinase cyclin-dependent kinase 8 (CDK8) is a

component of the mediator complex that functions as a bridgebetween basal transcription machinery and gene-specific tran-scriptional factors (16). CDK8 is amplified and overexpressed incolon cancer and exerts its oncogenic activity partially throughregulating β-catenin activity (17). The precise mechanisms by

which CDK8 regulates β-catenin remain obscure. CDK8 mRNAis up-regulated in malignant melanoma by loss of a transcrip-tional repressor called the histone variant macroH2A, whichfunctions as a tumor suppressor in melanoma (18). In addition,CDK8 protein levels are also controlled by S-phase kinase as-sociated protein 2 (Skp2)-mediated degradation of macroH2A1protein, and these three proteins work together to regulate G2/Mtransition and tumorigenesis in breast cancer (19). CDK8 exerts itsoncogenic function mainly through phosphorylation of substrates.Several substrates for CDK8 have been identified, including theNotch intracellular domain, SMAD complexes, E2F1, STAT1, andthe C-terminal domain of RNA polymerase II (16). These studieshighlight an important oncogenic function of CDK8 kinase activity.A connection between CDK8 and YAP, the critical transcriptional

coactivator of Hippo signaling, has not been established. Here, wereport that Zyxin promotes colon cancer cell growth, and itsoncogenic activity is partially controlled by mitotic phosphory-lation. We further showed that Zyxin regulates YAP activitythrough CDK8 in colon cancer cells. In addition, we identifiedYAP as a direct substrate of CDK8 and CDK8-mediated phos-phorylation that promotes YAP activity in vitro and in vivo.

Significance

Zyxin is a member of the cell–cell adhesion complex and con-trols cell cytoskeleton and motility. Analysis from clinicalsamples revealed that Zyxin is highly expressed in colon cancercompared with normal tissue, suggesting an oncogenic role forZyxin in cancer. Depletion of Zyxin resulted in significantlyimpaired colon cancer cell proliferation, migration, cellulartransformation, and tumor formation in xenograft animalmodels. We also showed that Zyxin is phosphorylated byCDK1 during mitosis. Mitotic phosphorylation is required forZyxin activity in promoting colon cancer growth. Zyxin regu-lates YAP activity through the colon cancer oncogene CDK8.We further showed that CDK8 directly phosphorylates YAP andpromotes its activation. These observations identify the Zyxin–CDK8–YAP axis as a potential therapeutic target in cancer.

Author contributions: J.Z. and J.D. designed research; J.Z., Y.Z., L.C., X.C., S.S., Z.W., andY.C. performed research; G.A.T. and A.R.B. contributed new reagents/analytic tools; J.Z.,Y.Z., L.C., F.Y., S.M.L., G.A.T., A.R.B., Y.C., and J.D. analyzed data; and J.Z. and J.D. wrotethe paper.

The authors declare no conflict of interest.

This article is a PNAS Direct Submission. K.-L.G. is a guest editor invited by theEditorial Board.

Published under the PNAS license.1To whom correspondence should be addressed. Email: [email protected].

This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10.1073/pnas.1800621115/-/DCSupplemental.

Published online July 2, 2018.

E6760–E6769 | PNAS | vol. 115 | no. 29 www.pnas.org/cgi/doi/10.1073/pnas.1800621115

Dow

nloa

ded

by g

uest

on

Janu

ary

25, 2

020

Page 2: Zyxin promotes colon cancer tumorigenesis in a …Zyxin promotes colon cancer tumorigenesis in a mitotic phosphorylation-dependent manner and through CDK8-mediated YAP activation Jiuli

ResultsZyxin Is Phosphorylated by CDK1 in Vitro During Mitosis. Others andwe have shown that several Hippo–YAP components are regu-lated and implicated in mitosis (20–26). These studies suggestthat the Hippo–YAP pathway controls tumorigenesis throughdysregulation during mitosis. Given the connection betweenZyxin and Hippo–YAP signaling, we tested the possibility thatZyxin might contribute to tumorigenesis through regulating cell-cycle progression, especially mitosis.As shown in Fig. 1A, treatment with taxol or nocodazole—

both agents that bind to microtubules and arrest cells in mitosis—significantly increased mobility upshift of Zyxin on a Phos-tag gel(Fig. 1A). Lambda phosphatase treatment completely convertedall slow-migrating bands to fast-migrating bands, confirmingthat Zyxin is phosphorylated in mitosis induced by spindlepoisons (Fig. 1B).Next, we used various kinase inhibitors to identify the candi-

date kinase for Zyxin phosphorylation. Of interest, treatmentswith RO3306 (CDK1 inhibitor) or Purvalanol A (CDK1/2/5 in-hibitor) significantly inhibited mobility shift and phosphorylation(Fig. 1C, lanes 5 and 6). These data suggest that CDK1, a well-known master mitotic kinase, is likely the relevant kinase forZyxin phosphorylation. Fig. 1D shows that purified CDK1/cyclinB kinase complex (CDK2 and CDK5/p25 kinases to a lesser

extent) directly phosphorylated GST–Zyxin proteins in vitro(Fig. 1D). Zyxin is not a suitable substrate for MAPK-p38α,MEK1, ERK1, and JNK1/2 kinases in vitro (Fig. 1D), althoughthese kinases recognize the same phosphorylation sequenceconsensus as CDK1.

Zyxin Is Phosphorylated by CDK1 in Cells During Mitosis. CDK1phosphorylates an S/TP consensus sequence (27). Databaseanalysis identified three SPs (S281, S308, and S344) as possiblemain phosphorylation sites in Zyxin during mitosis (28). Wegenerated phospho-specific antibodies against these sites.Nocodazole or taxol treatment significantly increased phos-phorylation of S281, S308, and S344 of endogenous Zyxin (Fig.1E). Phosphopeptide, but not regular nonphosphopeptide, in-cubation completely blocked the phospho-signal, suggesting thatthis antibody detects the phosphorylated form of Zyxin (SI Ap-pendix, Fig. S1 A–C). Exogenous Zyxin was also phosphorylatedduring mitotic arrest, and mutating serines to nonphosphorylatablealanines abolished the phosphorylation (SI Appendix, Fig. S1D).The siRNA-mediated depletion of Zyxin significantly blockedthe phospho-signal, confirming the specificity of these phospho-antibodies (Fig. 1F). Phosphorylation of Zyxin occurred in dose-dependent and time course-dependent manners (SI Appendix,Fig. S1 E and F). Using kinase inhibitors, we demonstrated thatphosphorylation of Zyxin is CDK1 kinase-dependent (Fig. 1G,

DM

SO

VX68

0M

K510

8R

O33

06Pu

rval

anol

ABI

2536

U01

26M

K220

6SB

2035

80

DM

SO-100

-55-40

-70-70

-100

-70

-70

Zyxin

Cyclin B β-Actin

(Phos-tag)

DM

SO

Taxo

l

Noc

o

Zyxin

Zyxin (Phos-tag)

-70Zyxin

Zyxin (Phos-tag)

Nocodazole - + + + + + + + + +

Noco - - + - + +

ppaseλ

-55

-40

Cyclin B

β-Actin

Zyxin

Cyclin B β-Actin

-55-40

-70

∗ ∗

P-32

kinase CD

K1C

DK2

CD

K5p3

JNK1

JNK2

MEK

1ER

K1-

GST-Zyxin

GST

-130-100

-130-100

p-S

281

Zyxi

n

Zyxin

Cyclin B β-Actin

DM

SO

Taxo

l

Noc

o

p-S

344

Zyxi

n

Zyxin

Cyclin B

β-Actin

DM

SO

Taxo

l

Noc

o

p-S

308

Zyxi

n

Zyxin

Cyclin B

β-Actin

DM

SO

Taxo

l

Noc

o

Nocodazole - + + +

p-S281 Zyxin

Zyxin

p-S344 Zyxin

p-S308 Zyxin

Cyclin B

β-Actin

-70

-70

-70

-70

-55

-40

siZyxinsiControl

- - #1 #2 + + - -

Zyxin

DM

SO

VX68

0M

K510

8R

O33

06Pu

rval

anol

ABI

2536

U01

26M

K220

6SB

2035

80

DM

SO

Cyclin B

β-Actin

-70

-70

-70

-70

-55

-40

Noco - + + + + + + + + +p-S281

p-S344

p-S308

-170-130

-70-100

-55-40-30

-70

-55

-40-15P-32

Zyxin

MBP

----------------MBP + + + + + + + + +

p-S281

Zyxin

p-S344

p-S308

Cyclin B

β-Actin

siCyclin B1 - - +Taxol - + +

siControl + + -

-70

-55

-40

-70

-70

-70

A B C

D E

F G H

Fig. 1. CDK1 phosphorylation of Zyxin during mi-totic arrest. (A) Taxol (100 nM for 16 h) or nocoda-zole (100 ng/mL for 16 h; Noco) treatments in HeLacells caused mobility upshift of Zyxin on Phos-tagSDS-polyacrylamide gels. (B) HeLa cells were treatedwith nocodazole (Noco) as indicated and cell lysateswere further treated with (+) or without (−) λphosphatase (ppase). (C) HeLa cells were treatedwith taxol together with or without various kinaseinhibitors as indicated. Inhibitors were added (withMG132 to prevent cyclin B from degradation and cellsfrom exiting from mitosis) 1.5 h before harvesting thecells. (D) In vitro kinase assays with purified kinasesusing bacterially purified GST–Zyxin (Abnova) andMBP (myelin basic protein) (Sigma) proteins as sub-strates. MBP proteins were included as positive con-trols, confirming the kinases are active. (E) Zyxin isphosphorylated at multiple sites during mitotic arrest.HeLa cells were treated as in A and total cell lysateswere probed with the indicated antibodies. (F) Vali-dation of phospho-specific antibodies. HeLa cells weretransfected with 40 nM scramble (control) or 40 nMsiRNA against Zyxin for 48 h and were further treatedwith nocodazole as indicated. (G) HeLa cells weretreated as in C. Total cell lysates were subjected toWestern blotting with the indicated antibodies. (H)HeLa cells were transfectedwith 40 nM scramble (control)or siRNA against Cyclin B1 for 48 h and were furthertreated with taxol as indicated.

Zhou et al. PNAS | vol. 115 | no. 29 | E6761

CELL

BIOLO

GY

Dow

nloa

ded

by g

uest

on

Janu

ary

25, 2

020

Page 3: Zyxin promotes colon cancer tumorigenesis in a …Zyxin promotes colon cancer tumorigenesis in a mitotic phosphorylation-dependent manner and through CDK8-mediated YAP activation Jiuli

lanes 5 and 6). Furthermore, knockdown of Cyclin B1 orCDK1 largely blocked Zyxin phosphorylation (Fig. 1H and SIAppendix, Fig. S1G). Enhanced expression of active CDK1/Cyclin B1 was sufficient to stimulate Zyxin phosphorylation (SIAppendix, Fig. S1H).Immunofluorescence staining revealed that a strong signal was

detected in nocodazole-arrested prometaphase cells for anti-bodies against Zyxin S281 and S344 (SI Appendix, Fig. S2 A and B,Upper, white arrows). Very low or no signal was detected in interphasecells (SI Appendix, Fig. S2 A and B, yellow arrows). Again, phos-phopeptide, but not nonphosphopeptide, incubation completelyblocked the signal, suggesting that these antibodies specifically detectphosphorylated Zyxin (SI Appendix, Fig. S2 A and B). The specificityof the antibodies was further confirmed by siRNA knockdown ofZyxin (SI Appendix, Fig. S2 C and D). Addition of RO3306 largelyabolished the signals detected by p-Zxyin S281 and S344 antibodiesin mitotic cells, further indicating that the phosphorylation isCDK1-dependent (SI Appendix, Fig. S2 A and B, Lower).To determine whether mitotic phosphorylation of Zyxin occurs

during unperturbed/normal mitosis, we performed immunofluo-rescence staining on cells collected from a double thymidine blockand release (29). Very weak signals were detected in interphase ortelophase/cytokinesis cells (Fig. 2 A and B, yellow arrows, and SIAppendix, Fig. S3 A and B). The phospho-signal increased inprometaphase and peaked in metaphase cells (Fig. 2 A and B, whitearrows, and SI Appendix, Fig. S3 A and B). After being releasedfrom double thymidine block, cells entered into mitosis at 10–12 h(21, 24) (revealed by increased p-S10 H3 levels), and p-ZyxinS281 and S344 signals also increased in these cells (Fig. 2C). Mi-totic phosphorylation of Zyxin occurred in freely cycling cells in aCDK1-dependent manner (SI Appendix, Fig. S4 A and B). Theseobservations indicate that Zyxin is phosphorylated by CDK1 duringmitosis.

Zyxin Expression Is Induced During Mitosis.During our experiments,we noticed that, in addition to mobility shift/phosphorylation,Zyxin protein levels were also increased during taxol ornocodazole-induced mitotic arrest (Fig. 2D) and in normal mi-tosis (SI Appendix, Fig. S4D). Quantitative RT-PCR (qRT-PCR)confirmed that Zyxin mRNA levels were induced by taxol ornocodazole (Fig. 2E) and in normal mitosis (Fig. 2F). YAPtargets survivin and Cyr61, but not YAP itself, were induced inmitosis (Fig. 2F and SI Appendix, Fig. S4 C–E). A previous studyshowed that Zyxin is a direct target of TAZ (a paralog of YAP)/TEAD (30), and we determined whether Zyxin induction inmitosis is YAP/TAZ-dependent. Indeed, addition of verteporfin[(VP) a YAP/TEAD inhibitor (31)] or knockdown of YAPlargely abolished Zyxin mRNA increase induced by taxol ornocodazole treatment (Fig. 2G and SI Appendix, Fig. S4F). Ofinterest, inhibition of CDK1 by Purvalanol A also blocked Zyxinexpression, suggesting that Zyxin expression in mitosis is inducedin a CDK1-dependent manner (Fig. 2G).

Zyxin Is Required for Colon Cancer Cell Growth in Vitro and in Vivo.We next explored the biological significance of Zyxin in cancercell growth. We analyzed the Oncomine and TCGA online da-tabases and found Zyxin to be highly expressed in colon cancer(Fig. 3A). We performed immunohistochemistry (IHC) stainingon tissue microarrays (TMAs) (n = 66 cancer/tumor, n = 35 normal)and confirmed that Zyxin protein levels were significantly increasedin colon cancer samples compared with normal tissue (Fig. 3 B andC). The specificity of the Zyxin IHC was validated by using Zyxin-knockdown tumor samples (Fig. 3D). These findings highlight acritical role of Zyxin in tumorigenesis in colon cancer.To directly determine the role of Zyxin in colon cancer, we

depleted Zyxin (with two independent shRNAs) in various coloncancer cells (Fig. 3 E–G). Knockdown of Zyxin greatly reduced

p-Zyxin S281 β-tubulin

DAPI Merged

20 um

p-Zyxin S344

DAPI

β-tubulin

Merged

20 um

Zyxin

β-Actin

p-S281 Zyxin

p-S344 Zyxin

-70

-70

-70

-40

p-S10 H3 -17

0 7.5 9.5 10.5 12 24Time (h)DT block and release

1 2 3 4 5 6

∗∗

∗∗

Rel

ativ

e Zy

x m

RN

A (fo

ld)

0.0

1.0

2.0

5.0

4.0

6.0

3.0

7.0

Control Noc Taxol

Cont

rol

Noco

dazo

le

Taxo

l

Cyclin B

β-Actin

Zyxin

Rel

ativ

e Zy

x m

RN

A (fo

ld)

0.0

1.0

2.0

5.0

4.0

3.0

∗∗∗∗

Taxol - + + +Inhibitors - - PA VP

-70

-40

-55

∗∗ ∗∗∗

G1/S G2/M

Rel

ativ

e m

RN

A (fo

ld)

G1/S G2/M0.0

1.0

2.0

1.5

0.5

0.0

1.0

2.0

2.5

1.5

0.5

3.0Zyxin Survivin

A B

C

D

F G

E Fig. 2. Zyxin is phosphorylated and induced duringunperturbed mitosis. (A) HeLa cells were synchro-nized by a double thymidine (DT) block and releasemethod. Cells were stained with antibodies againstp-Zyxin S281 or β-tubulin, or with DAPI. A 40× ob-jective lens was used to view various phases of thecells in a field. (B) Experiments were done similarly asin A with p-Zyxin S344 antibodies. White and yellowarrows (in A and B) mark the metaphase and in-terphase cells, respectively. (C) HeLa cells were syn-chronized by a DT block and release method. Totalcell lysates were harvested at the indicated timepoints and subjected to Western blotting analysis.(D) Zyxin protein levels increased during mitotic ar-rest. (E) Zyxin mRNA levels were induced during mi-totic arrest. (F) Zyxin mRNA levels were increasedduring normal mitosis. HeLa cells were treated as in Cand harvested at 10 h post release. Survivin serves asa positive control. (G) HeLa cells were treated withtaxol and inhibitors were added 2 h before harvest-ing the cells. qRT-PCR was performed to measureZyxin mRNA levels. Data were expressed as themean ± SEM of three independent experiments (E–G). **P < 0.01; ***P < 0.001 (t test).

E6762 | www.pnas.org/cgi/doi/10.1073/pnas.1800621115 Zhou et al.

Dow

nloa

ded

by g

uest

on

Janu

ary

25, 2

020

Page 4: Zyxin promotes colon cancer tumorigenesis in a …Zyxin promotes colon cancer tumorigenesis in a mitotic phosphorylation-dependent manner and through CDK8-mediated YAP activation Jiuli

migration (as shown by the wound healing assay) (Fig. 3 H–J andSI Appendix, Fig. S5 A–C), anchorage-independent growth (Fig.3 K and L and SI Appendix, Fig. S5 D and E), and proliferation(Fig. 3 M–O) in RKO, SW480, and HCT116 cells. Importantly,reexpression of shRNA-resistant wild-type Zyxin completelyrescued all these phenotypes in RKO cells (Fig. 4 A–C). How-ever, expression of nonphosphorylatable mutant Zyxin-3A(S281A/S308A/S344A) only partially restored these characteris-tics (Fig. 4 B and C), suggesting that mitotic phosphorylation isrequired for Zyxin oncogenic activity in colon cancer cells.Similar results were observed in SW480 cells (Fig. 4 D–F). Asexpected, Zyxin knockdown also greatly reduced tumor growthin animals, and cells bearing Zyxin-3A formed much smallertumors compared with cells expressing wild-type Zyxin (Fig. 4 Gand H). Knockdown efficiency was confirmed, and reexpressionlevels between wild-type and mutant Zyxin (Zyxin-3A) were similarin tumors (SI Appendix, Fig. S5F). These results suggest a criticalrole of Zyxin and its mitotic phosphorylation in colon cancer.

Zyxin Regulates YAP Activity in Colon Cancer Cells. Zyxin has beenshown to be a positive regulator of YAP/Yki activity (5–8, 11).We confirmed that Zyxin knockdown increased p-YAP S127 levelsand promoted YAP cytoplasmic localization (SI Appendix, Fig. S6 Aand B). Consistent with reduced YAP activity, expression of the targetgene CTGF was significantly reduced upon Zyxin knockdown in bothRKO and SW480 cells (SI Appendix, Fig. S6C). F-actin cytoskeletalremodeling was shown to control YAP/Yki activity in mammaliancells and in Drosophila (5–9, 32–34). F-actin polymerization was

inhibited by cofilin, for which activity is negatively controlled byphosphorylation at Serine3 (35–37). We found that phospho-S3 cofilinlevels were reduced and F-actin formation/cytoskeleton was stronglyinhibited upon Zyxin knockdown in colon cancer cells (SI Appendix,Fig. S6 D and E), suggesting that Zyxin might control YAP activityand subsequent cell migration and proliferation throughF-actin remodeling.To explore the functional interaction between Zyxin and YAP,

we reexpressed YAP–S127A in Zyxin knockdown cells (SIAppendix, Fig. S6 F and G). Enhanced expression of YAP–S127Acompletely rescued the effects (migration and proliferation) ofZyxin knockdown in both SW480 and HCT116 cells (SI Appendix,Fig. S6 H–K), suggesting that Zyxin promotes colon cancer cellgrowth through YAP.

Zyxin Regulates CDK8 to Promote Migration and Proliferation inColon Cancer Cells. To elucidate the underlying mechanisms inwhich Zyxin regulates YAP activity and controls colon cancercell growth, we performed an array analysis involving 84 cell-cycle regulators in control and Zyxin-knockdown cells. Five geneswere down-regulated and one gene up-regulated (CDKN2B) inZyxin-depleted cells compared with control cells (Fig. 5A). Most(except for NBN) were confirmed by qRT-PCR in an independentZyxin-knockdown cell line (SI Appendix, Fig. S7A). Among thesegenes, CDK8 (cyclin-dependent kinase 8) attracted us, since itfunctions as an oncogene in colon cancer (17) and in melanoma(18). We confirmed that CDK8 protein levels were also reduced inZyxin-knockdown RKO and SW480 cells (Fig. 5B) and restored by

Rel

. Zyx

in e

xpre

ssio

n (lo

g2)

0

1

2

3

Normal (N=35)

Tumor (N=66)

Normal (+)

Tumor (+) Tumor (++)

Tumor (+++) Control-tumor

shZyxin-tumor

-4% +

17%

++34%

+++45%

-48%

+36%

++13%

+++3%

Tumor Normal

N=483 N=41p=4.8e-06

β-actin

Zyxin

RKO

-70

-40

0.70

0.60

0.90

0.85

0.80

Rel

ativ

e w

ound

wid

th

shRNA Ctrl Zyx-1 Zyx-2

0.75

0.65

∗∗∗ ∗∗∗

shC

ontro

lsh

Zyxi

n-1

shZy

xin-

2

SW480

β-actin

Zyxin -70

-40

0.65

0.55

0.95

0.85

0.75

Rel

ativ

e w

ound

wid

th

∗∗∗∗∗∗

shRNA Ctrl Zyx-1 Zyx-2

β-actin

Zyxin -70

-40

shC

ontro

lsh

Zyxi

n-1

shZy

xin-

2HCT116

Rel

ativ

e w

ound

wid

th

shRNA Ctrl Zyx-1 Zyx-2

0.7

0.6

0.8

0.9

∗∗ ∗∗

∗∗∗∗∗∗

∗∗∗

∗∗∗

Days 0 2 4 60

5

10

15

20

Cel

l num

ber (

x10

)5 HCT116-shControlHCT116-shZyxin-1HCT116-shZyxin-2

∗∗∗

∗∗∗

∗∗

0 1 2 3 4 5 605

1015202530

RKO-shControlRKO-shZyxin-1RKO-shZyxin-2

Days

Cel

l num

ber (

X10

5 )

0 1 2 3 4 5 6 70

5

10

15SW480-shControlSW480-shZyxin-1SW480-shZyxin-2

Days

Cel

l num

ber (

X10

5 )

∗∗∗∗∗∗

∗∗∗∗∗∗

Col

ony

num

ber

HCT116

0

500

1000

∗∗∗ ∗∗∗

shRNA Ctrl Zyx-1 Zyx-2

shRNA Ctrl Zyx-1 Zyx-2

Col

ony

num

ber

0

500

1000

∗∗∗ ∗∗∗

SW480

shC

ontro

lsh

Zyxi

n-1

shZy

xin-

2

A B C D

E F G K

H I JL

M N O

Fig. 3. Zyxin levels are increased in colon cancerpatients and knockdown of Zyxin inhibits coloncancer cell growth. (A) Zyxin mRNA levels are in-creased in colon cancer samples. RNA-sequencingdata from The Cancer Genome Atlas (TCGA) weredownloaded from NCBI’s Gene Expression Omnibus(GEO) with the accession number of GSE62944 (theStudent t test was used for statistical evaluation). (Band C) IHC staining analysis of Zyxin on colon cancerTMAs (P < 0.001, Wilcoxon rank sum test). Represen-tative staining images were shown (C). Staining andscoring were done as we described (63). −: negative; +:low expression; ++: moderate expression; +++: strongexpression. (D) Validation of the Zyxin antibody in IHCspecificity using control and Zyxin-knockdown tumors.(E–G) Establishment of colon cancer cell lines withstable knockdown of Zyxin. (H–J) Cell migration(wound healing) assays of cell lines established in E–G.(K and L) Anchorage-independent growth (colony as-says on soft agar) of HCT116 and SW480 cell lines withknockdown of Zyxin (F and G). (M–O) Cell proliferationassays in cell lines established in E–G. Data wereexpressed as the mean ± SEM of three independentexperiments (H–O). **P < 0.01; ***P < 0.001 (t test).

Zhou et al. PNAS | vol. 115 | no. 29 | E6763

CELL

BIOLO

GY

Dow

nloa

ded

by g

uest

on

Janu

ary

25, 2

020

Page 5: Zyxin promotes colon cancer tumorigenesis in a …Zyxin promotes colon cancer tumorigenesis in a mitotic phosphorylation-dependent manner and through CDK8-mediated YAP activation Jiuli

reexpression of exogenous Zyxin (SI Appendix, Fig. S7B). TheZyxin-3A mutant had similar activity to wild-type Zyxin in rescuingCDK8 levels, suggesting that mitotic phosphorylation of Zyxin isdispensable in controlling CDK8 expression (SI Appendix, Fig.S7B). Importantly, ectopic expression of CDK8, but not the kinase-dead form (CDK8-KD), substantially restored the migratory(assayed by wound healing) and proliferating ability of Zyxin-depleted RKO and SW480 cells (Fig. 5 C–F). Moreover,siRNA-mediated transient knockdown (two independent siRNAs)or CRISPR/double nickase-mediated knockout (KO; two inde-pendent gRNAs) (38) of CDK8 significantly impaired cell mi-gration, proliferation, and anchorage-independent growth inSW480 cells (Fig. 5 G–L). CDK8 KO also reduced proliferationin DLD-1 cells (Fig. 5 M and N). Of note, both mRNA andprotein levels of CDK8 were up-regulated in colon tumorscompared with normal tissue (SI Appendix, Fig. S7 C and D). Wefound that CDK8 and Zyxin protein levels were positively cor-related in metastatic colon cancer samples (SI Appendix, Fig. S7E and F). Together, these data suggest that Zyxin promotescolon cancer cell growth via CDK8.In line with the above functional studies, reexpression of

CDK8 largely restored the YAP phosphorylation and target gene(CTGF) expression in Zyxin knockdown cells (Fig. 6 A and B),suggesting that Zyxin regulates YAP activity through CDK8.

CDK8 Requires YAP to Promote Cell Growth in Colon Cancer. Basedon our observations that Zyxin regulates YAP through CDK8(Figs. 5 and 6 A and B), we reasoned that CDK8 also controlsYAP activity. Indeed, we found that S127 phosphorylation ofYAP was increased in CDK8-KO cells (Fig. 6C). Consistent withdecreased activity of YAP in CDK8-KO cells, the expression ofYAP target gene CTGF was inhibited upon CDK8 deletion (Fig.6D). Furthermore, CRISPR-mediated KO (two independentgRNAs) of YAP strongly inhibited migration and proliferation

in RKO and SW480 cells in which Zyxin was depleted andCDK8 reexpressed (Fig. 6 E–H), suggesting that YAP is essentialfor CDK8-driven tumorigenesis in colon cancer. Thus, we identifiedCDK8 as a positive regulator of YAP activity.

CDK8 Phosphorylates and Promotes YAP Activity. CDK8 is a mem-ber of the mediator complex, which couples transcriptionalregulators to control basal transcription (16). Since CDK8 is akinase and regulates YAP transcriptional activity, we testedwhether CDK8 directly phosphorylates YAP. In vitro kinaseassays showed that purified CDK8/Cyclin C complex readilyphosphorylated GST- or His-tagged YAP proteins (Fig. 6 I and J).CDK8 could also undergo autophosphorylation under these con-ditions (SI Appendix, Fig. S8). CDKs phosphorylate at S/T-P mini-mal consensus sequences. YAP contains seven conserved S/TPs(T119, S128, S138, S217, S289, S367, and T412; the T143, T154, andS340 sites do not exist in mouse and were excluded from furtheranalysis). Of interest, mutating four amino acid positions (T119,S128, S289, and S367) to alanines (YAP-4A) completely blockedphosphorylation (Fig. 6J), suggesting that CDK8 phosphorylatesthese sites in YAP in vitro.Given that both Zyxin (Figs. 1 and 2) and YAP are phos-

phorylated during mitosis (21, 39), we next explored whether CDK8phosphorylates YAP in mitosis. Interestingly, S128 phosphorylationwas significantly increased in nocodazole-arrested mitotic cells (Fig.6K). Consistent with our previous study (21), phosphorylation ofT119 and S289 was also increased in mitotic cells (Fig. 6K). Im-portantly, CDK8 deletion largely blocked the phosphorylation of allthree sites induced by mitotic arrest, suggesting that CDK8 is arelevant YAP kinase in vivo, specifically in mitosis (Fig. 6K). Usingreporter assays, we next examined whether phosphorylation of YAPaffects its activity. Mutating these four sites to alanines greatlysuppressed YAP–S127A activity (Fig. 6L). These observations suggest

RKO

β-actin

Zyxin -70

-40

0.65

0.55

0.85

0.75

shZyxin - + + +Res-Zyxin - - WT 3A

∗∗∗

Rel

ativ

e w

ound

wid

th

Res-Zyxin - - WT 3A

shC

ontro

lsh

Zyxi

n-1

shZy

xin-

1sh

Zyxi

n-1

∗∗∗

∗∗

RKO

0 2 4 60

5

10

15

20

25 shControlshZyxinshZyxin+Res-Zyxin-WTshZyxin+Res-Zyxin-3A

Days

Cel

l num

ber (

X10

5 )

shControl (N=5)shZyxin (N=5)shZyxin+Res-Zyxin-WT (N=5)shZyxin+Res-Zyxin-3A (N=5)

Tum

or v

olum

e (x

100m

m )3

0

2

4

6

8

Days 10 14 17 21 25 29post injection

∗∗∗∗∗∗∗∗

shControl

shZyxin

shZyxin+Res-Zyxin-WT

shZyxin+Res-Zyxin-3A

SW480

β-actin

Zyxin -70

-40

Res-Zyxin - - WT 3A

shC

ontro

lsh

Zyxi

n-1

shZy

xin-

1sh

Zyxi

n-1

Rel

ativ

e w

ound

wid

th

0.65

0.55

0.85

0.75

shZyxin - + + +Res-Zyxin - - WT 3A

∗∗

∗∗

∗∗SW480

0 2 4 60

10

20

30

Days

Cel

l num

ber (

X10

5 )

shControlshZyxinshZyxin+Res-Zyxin-WTshZyxin+Res-Zyxin-3A

A B

D

C

E F

G H

Fig. 4. Mitotic phosphorylation of Zyxin is requiredfor colon cancer tumorigenesis. (A) Establishment ofZyxin-knockdown cell lines expressing shRNA-resistant(Res) Zyxin or Zyxin-3A in RKO colon cancer cells. 3A:S281A/S308A/S344A. (B and C) Cell migration (woundhealing) and proliferation assays in cell lines estab-lished in A. (D) Establishment of Zyxin-knockdown celllines expressing shRNA-resistant (Res) Zyxin or Zyxin-3A inSW480 colon cancer cells. 3A, S281A/S308A/S344A. (E andF) Cell migration (wound healing) and proliferation as-says in cell lines established in D. Data were expressed asthemean ± SEM of three independent experiments (B, C,E, and F). **P < 0.01; ***P < 0.001 (Res-Zyxin-WT vs. Res-Zyxin-3A) (t test). (G and H) Zyxin-3A failed to rescue tu-mor growth in Zyxin-knockdown RKO cells. RKO cell linesfromAwere s.c. inoculated into athymic nudemice (bothleft and right sides) and the representative tumors in eachgroup were excised and photographed at the endpoint(H). Yellow asterisks mark the comparisons between Res-Zyxin-WT and Res-Zyxin-3A. **P < 0.01; ***P < 0.001(t test).

E6764 | www.pnas.org/cgi/doi/10.1073/pnas.1800621115 Zhou et al.

Dow

nloa

ded

by g

uest

on

Janu

ary

25, 2

020

Page 6: Zyxin promotes colon cancer tumorigenesis in a …Zyxin promotes colon cancer tumorigenesis in a mitotic phosphorylation-dependent manner and through CDK8-mediated YAP activation Jiuli

that CDK8 directly phosphorylates and promotes YAP transcriptionalactivity, identifying YAP as a direct substrate for CDK8.

YAP Phosphorylation Is Required for CDK8-Driven Migration andProliferation. To further explore the functional relationship be-tween CDK8 and YAP, we performed complementary experi-ments. Ectopic expression of the constitutively active form ofYAP (YAP–S127A) had little impact on proliferation and mi-gration in SW480 and DLD-1 CDK8-KO cells (SI Appendix, Fig.S10 A and B). Expression of phospho-mimetic mutant YAP-4Dfailed to rescue proliferation and migration in CDK8-KO cells(Fig. 7 A and B). However, expression of YAP-S127A/4D(highest activity) substantially (but not completely) restoredCDK8-KO–mediated migration, proliferation, and anchorage-independent growth, suggesting that CDK8-mediated phos-phorylation is essential for full activation of YAP (Fig. 7 B–E andSI Appendix, Fig. S10 C–E), which is consistent with our earlierobservations (Fig. 6). CDK8 KO significantly inhibited tumorgrowth in vivo in both SW480 and DLD-1 cells (Fig. 7 F–I).Ectopic expression of YAP–S127A/4D also largely restored tu-mor growth of CDK8-KO cells in animals (Fig. 7 F–I).A previous report showed that CDK8 functions as a colon

oncogene through regulating β-catenin (17). Consistent with thisstudy, the inhibitory phosphorylation of β-catenin was increased,and target gene (LEF1) expression was inhibited in CDK8-KOcells (SI Appendix, Fig. S9 A and B). Interestingly, KO of β-cateninalso greatly impaired migration and proliferation in RKO cells withZyxin depletion and CDK8 reexpression (SI Appendix, Fig. S9 C–E).These findings suggest that CDK8 controls YAP and β-catenin ac-tivity, both of which are critical regulators in colon cancer.

Like YAP–S127A or YAP-4D, enhanced expression ofβ-catenin–S33Y (active form) had little effect on CDK8-KO cellgrowth (Fig. 7B and SI Appendix, Fig. S10 A, B, and D), sug-gesting that active β-catenin alone is not sufficient to rescueCDK8-KO–mediated cell growth. Coexpression of β-catenin-S33Y with YAP–S127A/4D did not further increase growthcompared with YAP–S127A/4D alone in CDK8-KO cells (Fig. 7B–I and SI Appendix, Fig. S10 D and E). Our data suggest thatCDK8 promotes YAP activation through a twofold mechanism:canonical S127 phosphorylation and direct phosphorylation.Both YAP and β-catenin are required for CDK8-driven tu-morigenesis in colon cancer. Altogether, our findings indicate thatZyxin promotes colon tumorigenesis in a mitotic phosphorylation-dependent manner and through CDK8-mediated YAP activation.

DiscussionOur study revealed that the cell-adhesion protein Zyxin pro-motes colon cancer cell growth by controlling CDK8 expression.It is currently not known how Zyxin regulates CDK8 expression.CDK8 mRNA expression is negatively regulated by the histonevariant macroH2A (18). A recent study demonstrated that theE3 ligase, Skp2, promotes macroH2A ubiquitination and deg-radation (19). Interestingly, in our array analysis, Skp2 is one ofthe five genes down-regulated by Zyxin depletion (Fig. 5 and SIAppendix, Fig. S7A). Therefore, it is possible that Zyxin controlsCDK8 expression through the Skp2–macroH2A axis. Since bothZyxin and CDK8 levels are increased in colon cancer patients(Fig. 3 and SI Appendix, Fig. S7 C and D) and possess oncogenicroles in colon cancer development (17), it will be interesting toexplore the biological significance of Skp2 and macroH2A incolon cancer and their functional interactions with Zyxin/CDK8.

CDK8

β-actin

RKOSW480

-40

-55

Zyxin -70

Magnitude of log2 (F. C.)

-1.934 0 1.934

CCND2 -3.82

CDK8 -2.94CDKN2B 2.08MAD2L1 -2.03NBN -2.27SKP2 -2.15

Gene F.C.

β-actin

Zyxin

Flag

0.50

0.60

0.70

0.80

0.90

Rel

ativ

e w

ound

wid

th

∗∗∗

RKO

shRNA Ctrl Zyx Zyx Zyx CDK8 - - WT KD

-40

-55

-70

shZyxin - + + + - + + +F-CDK8 - - WT KD - - WT KD

∗∗∗

∗∗∗_

_

SW480

_ _∗∗

∗∗∗

RKO ∗∗∗

SW480

0 2 4 60

10

20

30

40shControlshZyxinshZyxin+CDK8shZyxin+CDK8-KD

Days

Cel

l num

ber (

X10

5 )

RKO

0 2 4 60

5

10

15

20

25

30shControlshZyxinshZyxin+CDK8shZyxin+CDK8-KD

Days

Cel

l num

ber (

X10

5 )

Cel

l num

ber (

x10

)5

-55

-40

CDK8

β-actin

SW480

Con

trol

CD

K8-K

O-1

CD

K8-K

O-2

0

10

20

30

∗∗∗

∗∗∗

CDK8

β-actin

-55

-40DLD-1

Con

trol

CD

K8-K

O-1

CD

K8-K

O-2

shC

ontro

lsh

Zyxi

n-1

shZy

xin-

2sh

Con

trol

shZy

xin-

1sh

Zyxi

n-2

∗∗∗

∗∗∗

∗∗

β-actin -40

SW480

-55CDK8

siC

ontro

lsi

CD

K8-1

siC

DK8

-2

∗∗∗∗∗

0 2 4 6

Cel

l num

ber

( x1

0 )5

Days

∗∗∗

∗∗∗

∗∗∗

0.600.650.70

0.75

0.800.85

0.90

Rel

ativ

e w

ound

wid

th

siRNA Ctrl CDK8-1CDK8-2

0.650.70

0.75

0.80

0.85

0.90

Rel

ativ

e w

ound

wid

th

∗∗ ∗∗

Ctrl KO-1 KO-2

SW480

CDK8

Cel

l num

ber (

x10

)5

0 2 4 6Days 0 2 4 6Days

SW480 DLD-1

siControlsiCDK8-1siCDK8-2

0

5

10

15

05

1015202530Control

CDK8-KO-1CDK8-KO-2

ControlCDK8-KO-1CDK8-KO-2

A B C

D E F

G H I M

J K L N

Fig. 5. Zyxin regulates migration and proliferationthrough CDK8 in colon cancer cells. (A) Expressionprofiling of cell cycle regulators identified multiplegenes including CDK8 that are regulated by Zyxin.The RT2 cell cycle arrays (Qiagen) were used and ex-periments were performed following manufacturer’sinstructions. F.C., fold change. (B) CDK8 protein levelswere decreased in Zyxin-knockdown cells. (C) Estab-lishment of Zyxin-knockdown cells expressing Flag-tagged CDK8 or CDK8-KD (kinase dead, D173A).(D–F) Migration (wound healing) and proliferationassays in cell lines from C. Ectopic expression ofCDK8, but not CDK8-KD, rescued growth defects ofZyxin-knockdown cells. (G–I) Transient knockdown(two independent siRNAs) of CDK8 inhibited SW480cell migration (wound healing) and proliferation. (J)Establishment of CDK8 KO cell lines. Two indepen-dent gRNAs were used. (K and L) Cell migration(wound healing) assays and proliferation curves ofcell lines from J. (M and N) CDK8 KO inhibited pro-liferation in DLD-1 cells. Data were expressed as themean ± SEM of three independent experiments (D, F,H, I, K, L, and N). **P < 0.01; ***P < 0.001 (t test).

Zhou et al. PNAS | vol. 115 | no. 29 | E6765

CELL

BIOLO

GY

Dow

nloa

ded

by g

uest

on

Janu

ary

25, 2

020

Page 7: Zyxin promotes colon cancer tumorigenesis in a …Zyxin promotes colon cancer tumorigenesis in a mitotic phosphorylation-dependent manner and through CDK8-mediated YAP activation Jiuli

CDK8 positively regulates β-catenin transcriptional activity incolon cancer cells (17), although the precise mechanism remainsobscure. It may involve the phosphorylation of E2F1 and relieveE2F1-mediated repression on β-catenin/TCF signaling activity(17, 40, 41). The kinase activity of CDK8 is necessary forβ-catenin–driven transcription (17). Furthermore, a dominant-negative TCF construct, previously shown to inhibit β-catenin–induced cellular transformation, also only partially abrogatedCDK8 activity (17). This notion is supported by the observationsthat several profiling experiments failed to identify the broadderegulation of β-catenin signaling in CDK8-disturbed cells (16).These results strongly indicate that CDK8 controls additionalfactor(s) also outside the β-catenin signaling pathway to promotecolon cancer cell growth (i.e., other factors in addition toβ-catenin exist downstream of CDK8). Although our data (SIAppendix, Fig. S9 D and E) and previous studies (17) showed thatβ-catenin–KO suppressed the CDK8-driven cell migration andproliferation, restoration of β-catenin in CDK8-KO cells was notsufficient to support colon cancer cell growth (SI Appendix, Fig.S10), further suggesting that β-catenin signaling is not the main(or only) effector of CDK8. Our study highlights the importanceof YAP in mediating CDK8-driven tumorigenesis in colon can-cer (Figs. 6 and 7). CDK8 regulates YAP activity through atwofold mechanism: phosphorylation on S127/localization anddirect phosphorylation at T119/S128/S289/S367 (Figs. 6 and 7).CDK8-mediated direct phosphorylation promotes YAP further

activation and is required for CDK8-KO cell growth (Fig. 7).These observations not only added additional layers of YAPregulation, but also raised complex questions. For example,S128 has been shown to be phosphorylated by Nemo-like kinase(42, 43), and T119/S289/S367 are phosphorylated by CDK1 (21).How these kinases and phosphorylation are coordinated in cellsrequires further investigation. Another question is: How doesCDK8 regulate YAP S127 phosphorylation and localization?YAP S127 is largely mediated by Lats1/2 kinases in the Hippopathway; however, we did not observe significant changes ofLats1/2 kinase activity (revealed by phosphorylation levels oftheir activation loop) in CDK8-KO cells and -intact cells, sug-gesting a Hippo-independent regulation of YAP activity.Our study showed that the active β-catenin mutant (β-catenin–

S33Y) had little effect on rescuing the phenotypes of CDK8 KOin colon cancer cells (SI Appendix, Fig. S10). This implies thatadditional target(s) exists or, alternatively, that β-catenin–S33Y(like YAP–S127A) is not fully active in CDK8-deleted cells. Thesimilarity between YAP and β-catenin prompted us to reasonthat β-catenin is a direct substrate for CDK8. Indeed, β-cateninsequence analysis revealed three S/TP phospho-consensus (S191/S246/S605) as potential sites for CDK8 phosphorylation, andthese sites have been shown to be important for β-catenin cel-lular localization and activity (44).The global burden of colon cancer is rising steadily. Although

much progress has been made in therapeutic approaches, for

-100

-100GST-YAP-

P-YAP-

GST-CDK8-

P-CDK8-

CDK8 - + +GST-YAP + + +

Autoradiography

GST-Cyc C- -55

-70

3232

SW480 RKO ∗∗

∗∗∗∗∗∗

∗∗

Rel

ativ

e C

TGF

mR

NA 1.0

0.8

0.6

0.4

0.2

0.0

1.0

0.5

0.0

1.51.2

shZyxin - + + - + +CDK8 - - + - - +

YAP

p-S127 YAP

Flag

shZyxin - + +F-CDK8 - - +

RKO

β-actin

Zyxin

YAP

β-actin

-70

-40

_shZyxin+CDK8YAP-KO - #1 #2

RKO

Flag -55

YAP

CDK8

β-actin

_shZyxin+CDK8YAP-KO - #1 #2

SW480

-55

-70

-40

Cel

l num

ber (

x10

)5

10

20

0

5

15

DLD-1

CDK8

p-S127 YAP

β-actin

SW480

YAP

CDK8-KO - #1 #2 - #1 #2

SW480

∗∗∗

∗∗∗

Rel

ativ

e C

TGF

mR

NA

1.0

0.8

0.6

0.4

0.2

0.0CDK8-KO - #1 #2

1.2DLD-1

∗∗∗∗∗∗

- #1 #2

1.0

0.80.6

0.4

0.2

0.0

1.2

-70

-55

-70

-70

-40

-55

-70

-70

-40

shZyxin+CDK8shZyxin+CDK8+YAP-KO-1shZyxin+CDK8+YAP-KO-2

∗∗∗

∗∗∗

Days 0 2 4 6

RKO

Rel

. luc

ifera

se a

ctiv

ity

0

50

100

150

Vector S127A S127A/4A

∗∗∗

His-YAP-

GST-CDK8-

-70-100

-70-100

-70

-100

CDK8 - + +His-YAP WT WT 4A

P-YAP-P-CDK8-

Autoradiography

-55GST-Cyc C-

3232

p-S289 YAP

p-S128 YAP-SE

p-S128 YAP-LE

p-T119 YAP

IP: Y

AP

CDK8-KO - - + +

YAP

CDK8

-70

-55

-70

-70

-70

-70

-70

YAP

Nocodazole - + - +

1.0 0.31

-40β-Actin

-55

-40

1.0 0.32

CDK8-KO - - + +Nocodazole - + - +

CDK8

β-Actin

IP: Y

AP

Lysa

te

Lysa

te

1.0 0.26

Cel

l num

ber (

x10

)5

Days 0 2 4 6

8

16

0

4

12

SW480

shZyxin+CDK8shZyxin+CDK8+YAP-KO-1shZyxin+CDK8+YAP-KO-2

∗∗∗

∗∗∗G

A B C

D E F

H I J

K

L

Fig. 6. CDK8 phosphorylates YAP and promotes itsactivation. (A and B) Zyxin regulates YAP activitythrough CDK8. Ectopic expression of CDK8 inhibitedp-YAP S127 (A) and rescued CTGF (YAP target gene)expression (B) in Zyxin-knockdown cells. (C and D)CDK8 regulates YAP activity. CDK8 KO increased YAPphosphorylation (S127) (C) and inhibited CTGF ex-pression (D). (E–H) YAP is required for CDK8-drivenproliferation in colon cancer cells. (E and G) Estab-lishment of YAP KO cell lines. CRISPR/double nickase-mediated (two independent gRNAs were used) KO ofYAP in RKO and SW480 cells where Zyxin was de-pleted and CDK8 reexpressed. (F and H) Cell pro-liferation assays for cell lines from E and G. (I) In vitrokinase assays using GST-YAP as substrates. Twobatches of CDK8 kinases (SignalChem) were used. (J)In vitro kinase assays with His-YAP-WT and -YAP-4A(T119A/S128A/S289A/S367A) proteins as substrates.(K) CDK8 phosphorylates YAP in mitosis. SW480control and CDK8-KO cells were treated with DMSOor nocodazole for 16 h. Endogenous YAP proteinswere immunoprecipitated and probed withphospho-antibodies against YAP T119, S128,S289 and total YAP antibodies. Relative phosphory-lation (fold changes in signal intensity) was quanti-fied by Image J and shown below the blots fromthree independent experiments. LE, long exposure;SE, short exposure. (L) CDK8-mediated phosphoryla-tion stimulates YAP transcriptional activity. Lucifer-ase reporter assays were done as we described (60,64). Data were expressed as the mean ± SEM of threebiological repeats (B, D, F, H, and L). **P < 0.01;***P < 0.001 (t test).

E6766 | www.pnas.org/cgi/doi/10.1073/pnas.1800621115 Zhou et al.

Dow

nloa

ded

by g

uest

on

Janu

ary

25, 2

020

Page 8: Zyxin promotes colon cancer tumorigenesis in a …Zyxin promotes colon cancer tumorigenesis in a mitotic phosphorylation-dependent manner and through CDK8-mediated YAP activation Jiuli

patients with locally advanced primary or recurrent colon cancer,often unresectable for cure, survival remains poor (45). Further-more, available treatment options for colon cancer are very limiteddue to lack of validated targets. Our study has demonstrated thecritical role and regulatory mechanisms of the Zyxin–CDK8–YAP/β-catenin signaling in colon cancer. These studies suggest that thisaxis is a potential therapeutic target and provide several options fortargeted therapy using small-molecule inhibitors in colon cancer.For example, CDK8 function requires its kinase activity, and several

selective inhibitors have been developed (16, 46, 47). Furthermore,animal studies confirmed the potential efficacy of CDK8 inhibitors(48), although interpretation of CDK8 inhibitor data requires cau-tion since CDK8 may also have a tumor-suppressive function duringearly stages of colon tumorigenesis (49). VP was recently identifiedas an effective compound for preventing YAP activity (31). In-terestingly, VP (without light activation) significantly inhibited YAPactivity in vitro and suppressed YAP-driven tumorigenesis in themouse liver (31). Subsequent studies further confirmed that VP is

∗∗∗

∗∗∗

Rel

ativ

e w

ound

wid

th0.60

0.65

0.70

0.75

0.80

0.85

CDK8-KO - + + + + + +

∗∗∗ SW480

Control

CDK8-KO-2

CDK8-KO-2+YAP-S127A/4D

CDK8-KO-2+YAP-S127A/4D+β-Cat-CA6 9 12 15 18 21 24

0

2

4

6

8

10

Days post injection (DLD-1)

Tum

or v

olum

e (x

100

mm

3)

ControlCDK8-KO-1

CDK8-KO-2+YAP-S127A/4DCDK8-KO-2+YAP-S127A/4D+β-Cat-CA

CDK8-KO-2

∗∗

n.s.

∗∗∗∗

∗∗

∗∗∗

∗∗∗

∗∗∗∗∗

Con

trol

YAP

-4D

YAP

-S12

7A/4

D

YAP

-S12

7A

β-C

aten

in-C

A

Vect

or

∗∗

YAP

-S12

7A/4

D+β

-Cat

-CA

Cel

l num

ber

( X

105 )

10 20 30 40 500

2

4

6

8 ControlCDK8-KO-2CDK8-KO-2+YAP-S127A/4DCDK8-KO-2+YAP-S127A/4D+ β -Cat-CA

Days post injection (SW480)

Tum

or v

olum

e(x1

00 m

m3)

Control

CDK8-KO-2

CDK8-KO-2+YAP-S127A/4D

CDK8-KO-2+YAP-S127A/4D+β-Cat-CA

Col

ony

num

ber

0

100

200

300

CDK8-KO - + + +

Con

trol

Vect

or

YAP

-S12

7A/4

D

YAP

-S12

7A/4

D+β

-Cat

-CA

∗∗∗n.s.

CDK8-KO - + + +

Col

ony

num

ber (

x100

)

0

5

10

15

Con

trol

Vect

or

YAP

-S12

7A/4

D

YAP

-S12

7A/4

D+β

-Cat

-CA

∗∗∗∗

SW480

YAP

CDK8

Flag (β-Cat-CA)

β-actin -40

-55

-100

-70

Con

trol

Vect

orS1

27A

127A

/4D

YAP-

4Dβ-

Cat

-CA

S127

A/4D

+β-C

at-C

A

CDK8-KO

Days

SW480

DLD-1

SW480

ControlCDK8-KO-2CDK8-KO-2+S127A/4DCDK8-KO-2+S127A/4D+ β -Cat-CA

30

40

20

10

00 2 4 6

∗∗

∗∗

∗∗

A B C

D F G

E H I

Fig. 7. YAP phosphorylation is required for CDK8-driven tumorigenesis in colon cancer cells. (A) Establishment of cell lines expressing various YAPmutants and/orβ-catenin in CDK8 KO SW480 cells. CA, constitutive active form (S33Y mutant). (B–D) Functional assays of cell lines established in A. Cell migration (wound healing)assays in various cell lines as indicated (B). Cell proliferation curves of cell lines with various gene expression status (C). Anchorage-independent growth (colonyassays in soft agar) in cell lines as indicated (D). Data (B–D) are from three to four independent experiments and expressed as the mean ± SEM. *P < 0.05; **P <0.01; ***P < 0.001 (t test). (E) Anchorage-independent growth in various DLD-1 cells lines as indicated. Data are from three independent experiments andexpressed as the mean ± SEM. ***P < 0.001. n.s., not significant. (F and G) Tumor growth curves and images of all tumors (at the endpoint) from SW480 cell lines.2.5 × 106 cells (1:1 mixed with Matrigel) were s.c. inoculated into athymic nude mice on both flanks. n = 5 mice and 10 injections per group. Only three tumorswere formed at the last two time points in the CDK8-KO group and the other three groups formed seven to eight tumors (G). Red asterisks mark the comparisonsbetween control and CDK8-KO-2; Blue asterisks mark the comparisons between YAP-S127A/4D and CDK8-KO-2. *P < 0.05; **P < 0.01 (t test). (H and I) Tumorgrowth curves and representative images of tumors from DLD-1 cell lines. 1.0 × 106 cells (1:1 mixed with Matrigel) were s.c. inoculated into athymic nude mice onboth flanks (5 mice and 10 injections/group, 10 tumors were averaged for each group at each time point). Red asterisks mark the comparisons between controland CDK8-KO-2; Blue asterisks mark the comparisons between YAP-S127A/4D and CDK8-KO-2. *P < 0.05; **P < 0.01; ***P < 0.001 (t test). n.s., not significant.

Zhou et al. PNAS | vol. 115 | no. 29 | E6767

CELL

BIOLO

GY

Dow

nloa

ded

by g

uest

on

Janu

ary

25, 2

020

Page 9: Zyxin promotes colon cancer tumorigenesis in a …Zyxin promotes colon cancer tumorigenesis in a mitotic phosphorylation-dependent manner and through CDK8-mediated YAP activation Jiuli

also effective in inhibiting tumor growth in prostate cancer (50), uvealmelanoma (51, 52), and colon cancer in vivo (53), suggesting thatinhibiting YAP is a viable strategy in cancer treatment. Cyclin D1 isfrequently deregulated in colon cancer and is a known target ofβ-catenin signaling (17, 54). Interestingly, Cyclin D1 was also inducedby YAP activation in the intestine (55) and was identified as a directtarget of YAP/TEAD in mesothelioma cells (56). Therefore, CyclinD1 is a common target of YAP and β-catenin. Cyclin D1 is up-regulated (probably through YAP/β-catenin activation) in theApcMin/+ mice, and deletion of Cyclin D1 blocks/reduces ApcMin/+-driven adenoma formation (57). Acute deletion of Cyclin D1 in adultmice does not affect the animal’s health (58). These observationssuggest that targeting Cyclin D1 (e.g., with Palbociclib/PD-0332991,which is a Food and Drug Administration-approved selective inhibitorof Cyclin D1–CDK4/6) would be a viable strategy in colon cancer,especially in CDK8 and YAP/β-catenin–activated subtypes.

Materials and MethodsCRISPR/Double Nickase-Mediated KO. To construct the all-in-one CRISPR/Cas9n(double nickase) plasmid targeting β-catenin, the sense and antisense oli-gonucleotides from Table 1 were synthesized, annealed, and Golden Gateassembled into the pX330A_D10A-1 × 2-EGFP (38) and pX330S-2 vectors asdescribed (59). After the two vectors were generated, a final Golden Gateassembly was performed to generate the all-in-one vector as described (59).The resulting pX330A_D10A-1 × 2-EGFP–β-catenin-1 or -2 construct wastransfected into cells, and GFP-positive clones were selected by flowcytometry-based cell sorting. CDK8 and YAP CRISPR/double nickase KOplasmids were purchased from Santa Cruz Biotechnology, and transfectionand clone selection were done as instructed by the manufacturer.

Phos-Tag and Western Blot Analysis. Phos-tag was obtained from Wako PureChemical Industries, Ltd. (catalog no. 304-93521) and used at a concentrationof 20 μM (with 100 μM MnCl2) in 8% SDS-acrylamide gels. Before trans-ferring, the gels were equilibrated in transfer buffer containing 10 mMEDTA two times, each for 10 min. The gels were then soaked in transferbuffer (without EDTA) for another 10 min. Western blotting and lambdaphosphatase treatment assays were done as described (20).

Immunofluorescence Staining and Confocal Microscopy. Cell fixation, per-meabilization, fluorescence staining, and microscopy were done as described(29). The stained cells were mounted with Fluoromount (Vector Laborato-ries) and visualized on an LSM710 Zeiss fluorescence microscope (Carl Zeiss).The Slidebook 4.2 software (Intelligent Imaging Innovations) was used foranalyzing and processing all immunofluorescence images. F-actin wasstained by using rhodamine-labeled phalloidin (Cytoskeleton). YAP cellularlocalization was visualized by YAP immunofluorescence staining with an AlexaFluor 647-conjugated YAP antibody (Cell Signaling Technology). For peptideblocking, the phospho-Zyxin antibodies were first neutralized by an excess of

immunizing (phosphorylated) peptides (1 μg/mL for 1 h at room temperature).The antibody (containing the phospho-peptide) was then used for staining inparallel with staining using antibodies with no peptide or non-phospho-peptide.

Cell Proliferation, Cell Migration (Wound Healing), and Colony FormationAssays. Colony formation assays in soft agar were performed as described(60). Cells (5,000 per well) were seeded in six-well plates, and colonies werecounted by ImageJ online. For cell proliferation assays, cells (SW480 andHCT116: 100,000 per well; RKO and DLD-1: 50,000 per well) were seeded in asix-well plate in triplicate. Cells were counted by the automated cell counter(Invitrogen) at indicated time points and proliferation curves were madebased on the cell number in each well from at least three independent ex-periments. For wound healing (migration) assays, the cell monolayer wasscratched by a P200 pipet tip when cells were confluent. Cells were washed threetimes with PBS and incubated with 2 mL of serum-free medium for an additional24 h. The wound width was measured by QCapture Software (QImaging).

qRT-PCR. Total RNA isolation, RNA reverse transcription, and qRT-PCR weredone as we have described (61).

IHC Staining and Scoring. The TMA slides consisted of 66 colon adenocarci-nomas and 35 normal adjacent tissues collected from colon cancer patients atthe University of Nebraska Medical Center treated during 2008–2009. Slidedeparaffinization, antigen retrieving, and blocking were performed as wehave described (60). Tumors from RKO xenografts (control and Zyxin knock-down) were also used for Zyxin antibody validation. The sections were thenstained with anti-Zyxin antibody (at 1:100 dilutions; Cell Signaling Technology)by using a Histostain-Plus IHC kit following the manufacturer’s instructions(Invitrogen). The TMAs were also stained with anti-CDK8 antibody (IHC-00704, at1:300 dilutions; Bethyl Laboratory). Cell nuclei were stained with hematoxylin.Ventana iScan HT (Roche) was used for slide scanning with a 20× objective lens.The staining results were independently evaluated by three researchers, in-cluding one pathologist (S.M.L.). The staining intensity (a scale of 0–3was used: 0,negative; 1, weak; 2, moderate; and 3, strong) was scored (62).

Animal Studies. For in vivo xenograft studies, RKO cells (shControl, shZyxin,shZyxin with Res-Zyxin-WT, or Res-Zyxin-3A) (1.0 × 106 cells each injection,mixed with Matrigel at 1:1 ratio) were s.c. injected into the left or right flankof 6-wk-old male athymic nude mice (Nude-Foxn1nu; Envigo). Five animals(10 injections) were used per group. Similar approaches were used for DLD-1(1.0 × 106 cells per injection) and SW480 (2.5 × 106 cells per injection) cells inFig. 7. Tumor sizes were measured at the indicated time points. Tumorvolume (V) was calculated by the formula: V = 0.5 × length × width2 (60).Mice were euthanized at the end of the experiments, and the tumors wereexcised for subsequent analysis. The animals were housed in pathogen-freefacilities. All animal experiments were approved by the University ofNebraska Medical Center Institutional Animal Care and Use Committee.

Statistical Analysis. Statistical significance was evaluated by using a two-tailed, unpaired Student’s t test. The Wilcoxon rank sum test was used tocompare the IHC staining data between groups. A P value of <0.05 wasconsidered as indicating statistical significance.

ACKNOWLEDGMENTS. We thank Dr. Shian Wu (Nankai University, China)for the Zyxin shRNA constructs, Dr. Eek-hoon Jho (University of Seoul, Korea)for the p-S128 YAP antibody, and Xiaolong Yang (Queen’s University, Can-ada) for the TetOn-shCDK1 cell line. We thank Dr. Wayne Xu (University ofManitoba, Canada) for analyzing the mRNA expression levels of Zyxin andCDK8 from TCGA. We also thank Dr. Joyce Solheim for critical reading andcomments on the manuscript. All fluorescence images were acquired by ZeissLSM 710 or LSM 800 confocal microscopes at the Advanced Microscopy Coreat the University of Nebraska Medical Center. The core is supported in partby National Institutes of Health (NIH) Grant P30 GM106397. Research in theJ.D. laboratory is supported by Fred and Pamela Buffett Cancer Center Sup-port Grant P30 CA036727; NIH Grants P30 GM106397 and R01 GM109066;and Department of Defense Health Program Grant W81XWH-14-1-0150.

1. Beckerle MC (1997) Zyxin: Zinc fingers at sites of cell adhesion. BioEssays 19:949–957.2. Hirata H, Tatsumi H, Sokabe M (2008) Zyxin emerges as a key player in the mecha-

notransduction at cell adhesive structures. Commun Integr Biol 1:192–195.3. Hirata H, Tatsumi H, Sokabe M (2008) Mechanical forces facilitate actin polymeriza-

tion at focal adhesions in a zyxin-dependent manner. J Cell Sci 121:2795–2804.4. Nix DA, Beckerle MC (1997) Nuclear-cytoplasmic shuttling of the focal contact protein,

zyxin: A potential mechanism for communication between sites of cell adhesion and

the nucleus. J Cell Biol 138:1139–1147.

5. Gaspar P, HolderMV, Aerne BL, Janody F, TaponN (2015) Zyxin antagonizes the FERMprotein

expanded to couple F-actin and Yorkie-dependent organ growth. Curr Biol 25:679–689.6. Fernández BG, et al. (2011) Actin-capping protein and the Hippo pathway regulate

F-actin and tissue growth in Drosophila. Development 138:2337–2346.7. Rauskolb C, Pan G, Reddy BV, Oh H, Irvine KD (2011) Zyxin links fat signaling to the

hippo pathway. PLoS Biol 9:e1000624.8. Sansores-Garcia L, et al. (2011) Modulating F-actin organization induces organ growth

by affecting the Hippo pathway. EMBO J 30:2325–2335.

Table 1. β-catenin guide sequences

Oligo name Sequence

β-catenin-1A-Fwd CACCGGACGGTCGGACTCCCGCGG

β-catenin-1A-Rev aaacCCGCGGGAGTCCGACCGTCC

β-catenin-1B-Fwd CACCGCGACCGTCCTCGACCTGCGG

β-catenin-1B-Rev aaacCCGCAGGTCGAGGACGGTCGC

β-catenin-2A-Fwd CACCGCGAGGACGGTCGGACTCCCG

β-catenin-2A-Rev aaacCGGGAGTCCGACCGTCCTCGC

β-catenin-2B-Fwd CACCGACCTGCGGTGGCGGCTCGG

β-catenin-2B-Rev aaacCCGAGCCGCCACCGCAGGTC

Fwd, forward; Rev, reverse.

E6768 | www.pnas.org/cgi/doi/10.1073/pnas.1800621115 Zhou et al.

Dow

nloa

ded

by g

uest

on

Janu

ary

25, 2

020

Page 10: Zyxin promotes colon cancer tumorigenesis in a …Zyxin promotes colon cancer tumorigenesis in a mitotic phosphorylation-dependent manner and through CDK8-mediated YAP activation Jiuli

9. Aragona M, et al. (2013) A mechanical checkpoint controls multicellular growththrough YAP/TAZ regulation by actin-processing factors. Cell 154:1047–1059.

10. Feng X, et al. (2014) Hippo-independent activation of YAP by the GNAQ uvealmelanoma oncogene through a trio-regulated rho GTPase signaling circuitry. CancerCell 25:831–845.

11. Ma B, et al. (2016) Zyxin-Siah2-Lats2 axis mediates cooperation between Hippo andTGF-β signalling pathways. Nat Commun 7:11123.

12. Pan D (2010) The hippo signaling pathway in development and cancer. Dev Cell 19:491–505.

13. Yu FX, Zhao B, Guan KL (2015) Hippo pathway in organ size control, tissue homeostasis,and cancer. Cell 163:811–828.

14. Hong AW, Meng Z, Guan KL (2016) The Hippo pathway in intestinal regeneration anddisease. Nat Rev Gastroenterol Hepatol 13:324–337.

15. Meng Z, Moroishi T, Guan KL (2016) Mechanisms of Hippo pathway regulation. GenesDev 30:1–17.

16. Rzymski T, Mikula M, Wiklik K, Brzózka K (2015) CDK8 kinase—An emerging target intargeted cancer therapy. Biochim Biophys Acta 1854:1617–1629.

17. Firestein R, et al. (2008) CDK8 is a colorectal cancer oncogene that regulates beta-catenin activity. Nature 455:547–551.

18. Kapoor A, et al. (2010) The histone variant macroH2A suppresses melanoma pro-gression through regulation of CDK8. Nature 468:1105–1109.

19. Xu D, et al. (2015) Skp2-macroH2A1-CDK8 axis orchestrates G2/M transition andtumorigenesis. Nat Commun 6:6641.

20. Xiao L, et al. (2011) KIBRA protein phosphorylation is regulated by mitotic kinaseaurora and protein phosphatase 1. J Biol Chem 286:36304–36315.

21. Yang S, et al. (2013) CDK1 phosphorylation of YAP promotes mitotic defects and cellmotility and is essential for neoplastic transformation. Cancer Res 73:6722–6733.

22. Zhang L, et al. (2015) CDK1 phosphorylation of TAZ in mitosis inhibits its oncogenicactivity. Oncotarget 6:31399–31412.

23. Chen X, Chen Y, Dong J (2016) MST2 phosphorylation at serine 385 in mitosis inhibitsits tumor suppressing activity. Cell Signal 28:1826–1832.

24. Chen X, Stauffer S, Chen Y, Dong J (2016) Ajuba phosphorylation by CDK1 promotescell proliferation and tumorigenesis. J Biol Chem 291:14761–14772.

25. Hergovich A, Hemmings BA (2012) Hippo signalling in the G2/M cell cycle phase:Lessons learned from the yeast MEN and SIN pathways. Semin Cell Dev Biol 23:794–802.

26. Zeng Y, et al. (2017) Cyclin-dependent kinase 1 (CDK1)-mediated mitotic phosphor-ylation of the transcriptional co-repressor Vgll4 inhibits its tumor-suppressing activity.J Biol Chem 292:15028–15038.

27. Nigg EA (1993) Cellular substrates of p34(cdc2) and its companion cyclin-dependentkinases. Trends Cell Biol 3:296–301.

28. Hornbeck PV, et al. (2015) PhosphoSitePlus, 2014: Mutations, PTMs and recalibrations.Nucleic Acids Res 43:D512–D520.

29. Zhang L, et al. (2012) KIBRA regulates aurora kinase activity and is required for precisechromosome alignment during mitosis. J Biol Chem 287:34069–34077.

30. Diepenbruck M, et al. (2014) Tead2 expression levels control the subcellular distri-bution of Yap and Taz, zyxin expression and epithelial-mesenchymal transition. J CellSci 127:1523–1536.

31. Liu-Chittenden Y, et al. (2012) Genetic and pharmacological disruption of the TEAD-YAP complex suppresses the oncogenic activity of YAP. Genes Dev 26:1300–1305.

32. Dai X, et al. (2013) Phosphorylation of angiomotin by Lats1/2 kinases inhibits F-actinbinding, cell migration, and angiogenesis. J Biol Chem 288:34041–34051.

33. Zhao B, et al. (2012) Cell detachment activates the Hippo pathway via cytoskeletonreorganization to induce anoikis. Genes Dev 26:54–68.

34. Mana-Capelli S, ParamasivamM, Dutta S, McCollum D (2014) Angiomotins link F-actinarchitecture to Hippo pathway signaling. Mol Biol Cell 25:1676–1685.

35. Ostrowska Z, Moraczewska J (2017) Cofilin—A protein controlling dynamics of actinfilaments. Postepy Hig Med Dosw 71:339–351.

36. Mizuno K (2013) Signaling mechanisms and functional roles of cofilin phosphoryla-tion and dephosphorylation. Cell Signal 25:457–469.

37. Prunier C, Prudent R, Kapur R, Sadoul K, Lafanechère L (2017) LIM kinases: Cofilin andbeyond. Oncotarget 8:41749–41763.

38. Stauffer S, et al. (2017) CDK1-mediated mitotic phosphorylation of PBK is involved incytokinesis and inhibits its oncogenic activity. Cell Signal 39:74–83.

39. Bui DA, et al. (2016) Cytokinesis involves a nontranscriptional function of the Hippopathway effector YAP. Sci Signal 9:ra23.

40. Zhao J, Ramos R, Demma M (2013) CDK8 regulates E2F1 transcriptional activitythrough S375 phosphorylation. Oncogene 32:3520–3530.

41. Morris EJ, et al. (2008) E2F1 represses beta-catenin transcription and is antagonizedby both pRB and CDK8. Nature 455:552–556.

42. Hong AW, et al. (2017) Osmotic stress-induced phosphorylation by NLK atSer128 activates YAP. EMBO Rep 18:72–86.

43. Moon S, et al. (2017) Phosphorylation by NLK inhibits YAP-14-3-3-interactions andinduces its nuclear localization. EMBO Rep 18:61–71.

44. Wu X, et al. (2008) Rac1 activation controls nuclear localization of beta-catenin duringcanonical Wnt signaling. Cell 133:340–353.

45. Haddock MG (2017) Intraoperative radiation therapy for colon and rectal cancers: Aclinical review. Radiat Oncol 12:11.

46. Koehler MF, et al. (2016) Development of a potent, specific CDK8 kinase inhibitorwhich phenocopies CDK8/19 knockout cells. ACS Med Chem Lett 7:223–228.

47. Mallinger A, et al. (2016) Discovery of potent, selective, and orally bioavailable small-molecule modulators of the mediator complex-associated kinases CDK8 and CDK19.J Med Chem 59:1078–1101.

48. Clarke PA, et al. (2016) Assessing the mechanism and therapeutic potential of mod-ulators of the human mediator complex-associated protein kinases. eLife 5:e20722.

49. McCleland ML, et al. (2015) Cdk8 deletion in the Apc(Min) murine tumour modelrepresses EZH2 activity and accelerates tumourigenesis. J Pathol 237:508–519.

50. Nguyen LT, et al. (2015) ERG activates the YAP1 transcriptional program and inducesthe development of age-related prostate tumors. Cancer Cell 27:797–808.

51. Lyubasyuk V, Ouyang H, Yu FX, Guan KL, Zhang K (2014) YAP inhibition blocks uvealmelanogenesis driven by GNAQ or GNA11 mutations. Mol Cell Oncol 2:e970957.

52. Yu FX, et al. (2014) Mutant Gq/11 promote uveal melanoma tumorigenesis by acti-vating YAP. Cancer Cell 25:822–830.

53. Zhang H, et al. (2015) Tumor-selective proteotoxicity of verteporfin inhibits coloncancer progression independently of YAP1. Sci Signal 8:ra98.

54. Tetsu O, McCormick F (1999) Beta-catenin regulates expression of cyclin D1 in coloncarcinoma cells. Nature 398:422–426.

55. Camargo FD, et al. (2007) YAP1 increases organ size and expands undifferentiatedprogenitor cells. Curr Biol 17:2054–2060.

56. Mizuno T, et al. (2012) YAP induces malignant mesothelioma cell proliferation byupregulating transcription of cell cycle-promoting genes. Oncogene 31:5117–5122.

57. Hulit J, et al. (2004) Cyclin D1 genetic heterozygosity regulates colonic epithelial celldifferentiation and tumor number in ApcMin mice. Mol Cell Biol 24:7598–7611.

58. Choi YJ, et al. (2012) The requirement for cyclin D function in tumor maintenance.Cancer Cell 22:438–451.

59. Sakuma T, Nishikawa A, Kume S, Chayama K, Yamamoto T (2014) Multiplex genomeengineering in human cells using all-in-one CRISPR/Cas9 vector system. Sci Rep 4:5400.

60. Dong J, et al. (2007) Elucidation of a universal size-control mechanism in Drosophilaand mammals. Cell 130:1120–1133.

61. Xiao L, Chen Y, Ji M, Dong J (2011) KIBRA regulates Hippo signaling activity via in-teractions with large tumor suppressor kinases. J Biol Chem 286:7788–7796.

62. Song S, et al. (2014) Hippo coactivator YAP1 upregulates SOX9 and endows esoph-ageal cancer cells with stem-like properties. Cancer Res 74:4170–4182.

63. Zhang L, et al. (2015) The hippo pathway effector YAP regulates motility, invasion,and castration-resistant growth of prostate cancer cells. Mol Cell Biol 35:1350–1362.

64. Yang S, et al. (2015) Active YAP promotes pancreatic cancer cell motility, invasion andtumorigenesis in a mitotic phosphorylation-dependent manner through LPAR3.Oncotarget 6:36019–36031.

Zhou et al. PNAS | vol. 115 | no. 29 | E6769

CELL

BIOLO

GY

Dow

nloa

ded

by g

uest

on

Janu

ary

25, 2

020