<<

Polo-like 1 in genomic instability

Xiaoqi Liu

Department of Toxicology and Cancer Biology

University of Kentucky The fate of a single parental throughout the eukaryotic cell cycle Cytokinesis Activation of cell division

Preparation for mitosis The stages of mitosis and cytokinesis in an animal cell

MT MT - + kinetochore Centrosome Spindle pole + (MTOC) Spindle pole Centrosome Spindle

Nucleus

G2 Prophase Prometaphase Metaphase

Central spindle Contractile ring Midbody

Anaphase Telophase Cytokinesis Control of mitotic entry and exit by Cdc2/ B and APC

Anaphase promoting complex (APC) Cdc2 Cyclin B

Cdc2 Anaphase inhibitor Cyclin B Cdc2

G2 Prophase Metaphase Anaphase Telophase G1

(Liu & Erikson, PNAS 2002; Liu & Erikson, PNAS 2003; Liu et al, MCB 2005; Liu et al, MCB 2006) The polo-like have a C-terminal polo-box domain M-phase kinase domain polo-box Plk1 603

Plx1 (Xl) 80% 598

polo (Dm) 65% 576

Plo1 (Sp) 50% 683

Cdc5 (Sc) 51% 705

Early phase of cell cycle

Plk2 52% 683

Plk3 52% 631 Plk1 localizes to mitotic structures G2/pro Prometa

Spindle poles Centrosomes kinetochores

Centrosomes Centrosomes kinetochores

Meta Telo

Spindle poles Midbodies kinetochores

Spindle poles Central spindles kinetochores Plk1 in cancer

---Plk1 overexpression is correlated with cell proliferation and carcinogenesis.

---Plk1 is a new diagnostic marker for cancer.

---Plk1 inhibitors are in various clinical trails.

---However, how Plk1 contributes to carcinogenesis is elusive. Does Plk1 elevation contribute to cancer progression?

Approach: generation of Plk1-Knock in (KI) mouse line Plk1 KI mice Phenotypes

Plk1 KI causes aneuploidy in MEFs No apparent phenotypes for Plk1-KI mice, but they are hyper-sensitive to IR

DDB

ATM

Chk2 gH2AX

Cell cycle arrest & DNA repair A liver B Ctrl CMV-Cre/Plk1-KI IR-treated, liver

CMV-Cre/Plk1-KI H&E 50µm

Plk1-KI mice have

Plk1 Plk1 -

increased incidence anti 50µm

of lymphoma or Ki67 Ki67

- Ctrl

severe fatty change anti 50µm

IR-treated, liver T cell lymphoma

upon IR C D anti-CD3

KI

-

/Plk1

Ctrl Cre Multifocal lymphoid hyperplasia - in perivascular location CMV 200µm

200µm 100µm 50µm

KI

KI -

Mild lymphoma -

/Plk1

/Plk1

Cre

Cre -

Multiple lymphocytic - CMV CMV 100µm

overgrowth in the parenchyma

KI

-

KI

-

/Plk1

/Plk1 Cre

Severe lymphoma -

Cre

- CMV

CMV 50µm

E Ctrl CMV-Cre/Plk1-KI CMV-Cre/Plk1-KI F Fatty liver

KI

-

Ki67

/Plk1

-

Cre

anti -

200µm 200µm 100µm 50µm CMV 50µm Plk1 KI inhibits expression of DNA damage repair

Dr. Zhiguo Li Dr. Jinghui Liu More specific mechanism?

DNA double strand breaks

MRN complex (Mre11/Rad50/Nbs1) Plk1

ATM

Chk2

p53

Cell cycle arrest DNA repair A hypothesis to be tested

Plk1 elevation leads to

1) premature termination of DNA damage checkpoint and

2) reduced DNA repair Plk1-associated activity antagonizes DNA damage checkpoint Plk1 phosphorylates Mre11 at S649 and S688 in vitro In vivo, Plk1 phosphorylates Mre11-S649 and CK2 targets Mre11-S688 Mre11-S649/S688 is required for G2 DNA damage checkpoint recovery

1st T block 8h rel. 2nd T block 7h rel. Dox for 1h caffeine for 3h p-H3 IF Phosphorylation of Mre11 at S649/S688 inhibits its binding to DNA

Xenopus egg extract + Purified Mre11 + Biotin-tagged dsDNA-bound avidin beads

Pellets

Mre11 IB Phosphorylation of Mre11 at S649/S688 abolishes its foci formation Phosphorylation of Mre11 at S649/S688 abolishes Nbs1 foci formation Phosphorylation of Mre11 at S649/S688 abolishes Rad50 foci formation Phosphorylation of Mre11 at S649/S688 inhibits DNA damage checkpoint Mre11-S649/S688 phosphorylation inhibits DNA repair

A B Conclusions

Plk1 phosphorylation of Mre11 leads to

1) premature termination of DNA damage checkpoint

2) reduced DNA repair

Publications: Li et al., Cancer Research, 2017, 77, 3169.

Li et al., JBC, 2017, 292, 17461. Dr. Zhiguo Li MDC1 • MEDIATOR OF DNA DAMAGE CHECKPOINT

DNA damage MRN (Mre11/Rad50/Nbs1)

Initiation of damage response Plk1

MDC1 Amplification of damage response

Bekker-Jensen and Mailand, DNA repair, 2010, 9, 1219 MDC1 KO phenotype

MDC1 heterozygous mice are cancer prone by a DDR-independent pathway MDC1 reduction causes aneuploidy A

Foci formation in regular dish

MDC1 heterozygosity sensitizes transformation of immortalized MEFs

Soft agar assay

To immortalize, MEFs were transfected with SV40 large T antigen MDC1 is required for mitotic progression Double Thymidine Block--- Release

G1

S

G2

M

G1 MDC1 is required for timely progression of mitosis

HeLa expressing GFP-H2B

HeLa expressing GFP-a-tubulin MDC1 is phosphorylated in mitosis Plk1 phosphorylates MDC1 at T4 p-T4-MDC1 localization in prophase and prometaphase

Nuclear envelope Centrosomes

Kinetochores Centrosomes p-T4-MDC1 localization in mitosis

Kinetochores

Kinetochores

Microtubule (+) ends

Midbody p-T4-MDC1 localization in mitosis

Nuclear envelope

Kinetochores

Kinetochores

Kinetochores

Midzone

Midbody Plk1 phosphorylation of MDC1-T4 is required for mitotic progression Double Thymidine Block --- release

G1 Noc release

S

G2

M

G1 Plk1 phosphorylationPlk1 phosphorylatesof MDC1-T4 is required MDC1 at for T4 mitotic progression Plk1 phosphorylatesSummary MDC1 at T4

--MDC1 has a function independent of DNA damage response

--MDC1 is required for mitosis

--MDC1 is phosphorylated by Plk1 at T4

--Plk1 phosphorylation of MDC1-T4 is required for timely progression through mitosis

Publications: Li et al., Mol Cell Biol, 2017, 37, e00595.

Dr. Zhiguo Li PThelk1 phosphorylates Numb/p53 MDC1pathway at T4

Numb

MDM2 p53

-- Numb is an inhibitor of Notch signaling

-- Numb is involved in the cell-fate decisions of a number of cell lineages

-- Numb, MDM2 and p53 form a trimeric complex

-- Numb stabilizes p53 by inhibiting the E3 activity of MDM2

Colaluca, Nature 2008, 451, 76-80. Plk1 phosphorylates Numb at S265 and S284 in vitro

GST-Numb (aa) GST-Numb (aa)

GST-Numb (aa) Numb-S265 is phosphorylated in vivo in a Plk1 dependent manner Numb degradation is enhanced by Plk1-associated kinase activity Plk1 phosphorylation of Numb results in its proteasome degradation Plk1 negatively regulates the Numb/p53 pathway

**

* Plk1-KI MEFs * Tamoxifen - + * DOX + + 24 * Plk1 22 20 * p53 * 18 p21 16 g-H2AX 14 β-actin 12 c 10 8 A higher binding affinity between p53 and Numb-AA p53 reporter RLU 6 Flag-Numb WT AA DD 4 2 Flag 0

p53

β-actin p53/actin 1.0 3.3 1.2

Flag IP:Flag GFPGFP--Plk1 p53 FlagFLAG β-actin U2OS cells β-actin Plk1 phosphorylation of Numb contributes to p53 degradation

Numb WT AA DD

180KD IP:p53 IB:Ub

90KD

p53

FLAG p53 β-actin

p53/β-actin 1 2.7 1.6 Cells expressing Numb-S265A/S284A are more sensitive to

** Numb WT AA DD ** DOX - + - + - + *** *

) 20 Flag 5 15

p53 10 b-actin 5

Cell # (1 x 10 x (1 # Cell 0 p53/b-actin 1.0 5.1 3.8 9.1 1.6 4.1 MCF cells

* Numb WT AA DD * 60 * DOX - + - + - + * **

) **

Flag 4 50 10 × 40 p53 30

Cleaved PARP Number (1 20

b-actin Cell 10

p53/b-actin 1.0 8.5 2.6 11.3 1.3 6.1 0 U2OS cells Cells expressing Numb-S265A/S284A are more sensitive to doxorubicin

- DOX + DOX

WT ** 250 * 200

150 AA 100

50 # of colonies of #

0 WT+DOX AA+DOX DD+DOX DD Tumors carrying Numb-S265A/S284A are more sensitive to chemotherapy

500

450

400 FLAG β-actin 350

300 WT DOX injection AA 250 DD 200

WT+DOX volume (mm3) volume 150 AA+DOX 100 DD+DOX ** Tumor Tumor * 50 * 0 0 5 10 15 20 25 30 35 40 45 50 55 Days post-inoculation

600 *

WT WT+DOX 500 * 400 300

AA AA+DOX 200 Tumor weight(mg) Tumor 100 0 DD DD+DOX Tumors carrying Numb-S265A/S284A are more sensitive toFigure chemotherapy 7 a

WT S265A/S284AAA S265D/S284DDD

20 * 18 16

DOX 14

- 12 10 8 WT+DOX AA+DOX DD+DOX cells(%) tumor Positive 6 4 2

0 + DOX DOX +

Cleaved Caspase 3 IHC Plk1 phosphorylatesWorking model MDC1 at T4

Publications: Shao et al., 2018, 37, 810.

Tumors carrying a high level of Plk1 and WT p53 Plk1

P P Dr. Chen Shao S265 S284 Numb p53 Low response Numb p53 Degradation Degradation to chemotherapy

Plk1 BI2536

S265 S284 Numb p53 Enhanced response Numb p53 Stabilization Stabilization to chemotherapy Acknowledgements

Funding: NIH