Skip to main content

Table 1 Function of CDK1 based on cell line studies

From: A review on the role of cyclin dependent kinases in cancers

Tumor type

Targets/ Regulators and Signaling Pathways

Cell line

Function

References

Adrenocortical carcinoma

CENPF

SW13

CENPF/CDK1 signaling pathway was found to regulate the G2/M-phase, thus enhancing progression of adrenocortical carcinoma

[6]

Bladder cancer

PVT1/miR-31/CDK1 axis

RT4, T24, BIU‐87, and 5673

PVT1 facilitated proliferation, migration, and invasion via down-regulating miR-31 to enhance CDK1 expression

[7]

TFCP2L1

Murine R1, E14TG2a, and gcOct4‐GFP ESCs, HBlEpC, J82, T24, 5637, HT1197, HT1376, and RT4

CDK1-mediated TFCP2L1 phosphorylation was found to have essential role in bladder cancer

[8]

Cdc25C, Chk1, CDK1-cyclin B1 complex, Myt1, Wee1, phospho-Cdc25C (Ser216), Gadd45α, and 14-3-3 proteins

SCaBER, 5637, T24, UMUC3, TCCSUP, SV-HUC, T24, T24T, TCCSUP, UMUC1, and SV-HUC

Protein kinase D inhibitor “CRT0066101” suppressed expression of Cdc25C, which activates CDK1, but activated Chk1, that inhibits CDK1 and indirectly reduced the CDK1-cyclin B1 complex activity, so it inhibited bladder cancer growth by blocking cell cycle at G2/M

[9]

Breast cancer

KIAA1429

MCF-7, BT474, SUM1315,

MDA-MB-231 and MCF-10A

KIAA1429 was found to positively regulate CDK1

[10]

_

MCF-7 and MDA-MB-231

∆ CDK1: ↓ migration and invasion

[11]

UBAP2L

MCF-7, ZR-75-30, BT-474, T-47D and MDA-MB-468, and MCF-10A

∆ UBAP2L: ↓ proliferation, colony formation, CDK1 levels, and ↑ cell cycle arrest

[12]

miR-424

MDA-MB-231, HCC1937, MCF-10A, and HEK-293 T

↑↑ miR-424: ↓ proliferation and ↑ cell cycle arrest via targeting CDK1

[17]

NUSAP1, and DLGAP5

MCF-7

∆ NUSAP1: ↓ proliferation, migration, and invasion via regulating CDK1 and DLGAP5 expression and ↑ sensitivity to E-ADM

[18]

RBM7

SUM-1315, MCF-7, BT474, ZR-75-1, and MDA-MB-231

RBM7 was found to bind to the 3'-UTR of CDK1 transcript, which is involved in the stability of CDK1 mRNA

RBM7 plays its oncogenic role by increasing the levels of CDK1

[19]

Burkitt lymphoma

miR-129 and iASPP

Raji and CA46

miR-129 was found to target CDK1, so it is involved in inhibiting iASPP phosphorylation and reducing proliferation

∆ CDK1: ↓ iASPP S84/S113 phosphorylation, so blocked iASPP nucleus localization

[13]

Cancer stem cells

RAS/MAPK/CDK1 pathway, SOX2

p53 − / − MEFs, HRASV12-expressing p53 − / − MEF, TIG-3, and TIG-3–SMR, HCT116, SW480, DLD1, HCC827, and H460

RAS/MAPK/CDK1 pathway induces enhanced O-GlcNAc modification and is required for expression of SOX2 and cancer stem cells generation

[14]

miR-143-3p and miR-495-3p

HcerEpic, C4-1, HeLa, SiHa, and

Caski

CDK1 was a target of miR-143-3p and miR-495-3p

↑↑ miR-143-3p or miR-495-3p: ↓ proliferation, migration, invasion, viability and ↑ apoptosis

[20]

NCK1-AS1/miR-6857/CDK1 axis

CerEpiC, HeLa, C33A and SiHa and CaSki

∆ NCK1-AS1: ↓ proliferation and invasion, and ↑ cell cycle arrest

NCK1-AS1 was found to sponge miR-6857, so regulate CDK1/6 protein translation

[21]

Cholangiocarcinoma

_

CCKS-1, TFK-1 and HUCCT-1

∆ CDK1: ↓ proliferation and invasion, and ↑ cell cycle arrest

[22]

PSMC2

HUCCT1, QBC939, RBE, and HCCC-9810

∆ PSMC2: ↓ proliferation, cell migration, ↑ cell cycle arrest, and apoptosis

PSMC2 eas found to regulate its role via regulating CDK1

[23]

Colorectal cancer

KCTD12

HCT116 and HT29

Adefovir dipivoxil: ↓ proliferation, tumorigenesis, and ↑ G2 phase arrest via disrupting the CDK1-KCTD12 interaction

↑↑ CDK1: ↑ vemurafenib resistance

[16]

MEK/ERK pathway

HT-29, RKO, VACO432, WiDr, DLD1, SW620, DiFi, A375, A19, T29 and VACO432, VT1, NB7

∆ CDK1: ↑ sensitivity to apoptosis

A MEK/ERK inhibitor targeting CDK1 has effective role in reduction of cell proliferation

[15]

miR-378a-5p

SW480, HCT116, SW620, HT-29 and NCM460

CDK1 was a target of miR-378a-5p

↑↑ miR-378a-5p: ↓ proliferation and migration

↑↑ CDK1: ↑ proliferation and migration

[24]

DPP3

DLD-1, SW480, HCT 116, and RKO

∆ CDK1: ↓ inhibitory effects of DPP3 knockdown

∆ DPP3: ↓ proliferation, migration, ↑ apoptosis and cell cycle arrest

DPP3 was found to regulate CRC via CDK1

[25]

SNHG4/ miR-590-3p/CDK1 axis

FHC, HCT8, LoVo, HCT116, SW620, and HT29

∆ SNHG4: ↓ proliferation, viability, metastasis, and colony formation via targeting miR-590-3p and regulating CDK1

[26]

NFE2L3, DUX4

HCT116 and

HT29

∆ NFE2L3: ↑ levels of DUX4, which is an inhibitor of CDK1

[27]

SNRPA1

SW480, RKO, HT-29, HCT116, and HEK293T

∆ SNRPA1: ↓ proliferation, ↑ apoptosis

SNPRA1 was found to regulate CDK1 in CRC

[28]

Endometrial carcinoma

miR-1271

ECC-1, RL95-2, AN3 CA, and T-HESC

↑↑ miR-1271: ↓ cell proliferation, ↑ apoptosis via targeting CDK1

[29]

Esophageal squamous cell carcinoma

FAM135B, PI3K/Akt/mTOR signaling pathway

KYSE150, ECA109, TE-13, TE-10 and TE-1

∆ FAM135B: ↓ colony formation and ↓ cell cycle protein expression (pP53, CDK1), ↑ cell cycle arrest and ↑ radiosensitivity through regulating PI3K/Akt/mTOR

[30]

Gastric cancer

CASC11 and miR-340-5p

GES-1, MKN7, KATOIII and AZ521

∆ CASC11: ↓ proliferation, ↑ apoptosis and cell cycle arrest

CASC11 regulated CDK1 via targeting miR-340-5p

[31]

ESRRA, CDC25C-CDK1-Cyclin B1 pathway

HGC27, BGC823, MGC803, SGC7901 and GES-1

∆ ESRRA: ↓ cell viability, proliferation, migration, and invasion, EMT process, and ↑ apoptosis

ESRRA/DSN1/CDC25C-CDK1-Cyclin B1 pathway was involved in in GC development

[32]

CDCA5

MGC-803, SGC-7901, and BGC-823

∆ CDK1: ↓ proliferation, colon formation, migration, and invasion

CDK1 and CDCA5 were co-expressed in GC cells

[33]

ISL1

BGC823, MGC803, MKN28, and GES1

is CDK1 phosphorylated ISL1 at serine 269, thus promoted proliferation

[34]

Glioblastoma

p50, BCL-3, NF-κB

U87, A172, T98, U251, and GBM34

CDK1 was found to be up-regulated by temozolomide in an NF-κB related manner

∆ CDK1: ↑ sensitivity cells to temozolomide

[35]

Glioma

FOXD2-AS1/miR-31/CDK1 axis

SVG p12, T98,

LN229, U87, U251, and 293FT

∆ FOXD2-AS1: ↓ proliferation, and ↑ cell cycle arrest

FOXD2-AS1 was found to sponge miR-31, so regulated CDK1 levels

[36]

Hepatocellular carcinoma

PDK1/β-Catenin

MHCC97H (97H), LO2 and 97H liver cancer stem cells

∆ CDK1/PDK1/β-Catenin: ↓ EMT process

RO3306 and sorafenib combination: ↓ 97H CSC growth

[37]

DEPDC1B

HEP3B2.1-7, SK-HEP-1, huh-7, and HCCLM3

∆ DEPDC1B: ↓ proliferation, migration, colony formation, and ↑ G2 phase arrest, and cell apoptosis

The function of DEPDC1B was found to be mediated by CDK1

[38]

miR-1271-5p

SMMC-7721 and HuH-7

↑↑ miR-1271-5p: ↓ proliferation and ↑ radiosensitivity via targeting CDK1

[39]

CDK1-PLK1/SGOL2/ANLN pathway

SK-Hep1

∆ CDK1: ↓ expression of PLK1, ANLN, and SGOL2 and resulted in a disordered cell cycle

[40]

Upf1/SNORD52/CDK1 pathway

Huh7, HepG2, Hep3B, SK-Hep1, HCCLM9, HCCLM3, and HL-7702

∆ SNORD52: ↓

↓ migration and invasion, and ↑ cell cycle arrest

SNORD52 was found to regulate CDK1 by increasing the stability of CDK1 proteins

[41]

Leukemia

PLK1, Aurora B, and TRF1

HL-60

∆ CDK1: ↓ proliferation, ↑ cell cycle arrest via reducing the phosphorylation of PLK1 and Aurora B and negatively regulating TRF1

[42]

Lung cancer

Sox2

A549 and NCI-H520

∆ CDK1: ↑ chemotherapeutic sensitivity

CDK1/Sox2 axis was found to regulate the stemness

[43]

CASC11, miR-302

A547, H157, SPC-A-1 and 16HBE

∆ CASC11: ↓ proliferation via targeting miR-302 and regulating CDK1

[44]

miR-34c-3p

A549, CALU-1, and HCC827

↑↑ miR-34c-3p: ↓ proliferation, ↑ apoptosis and in KRASmut cells via targeting CDK1

[45]

NCK1-AS1

A549, NCI-H1299, PC-9 and NCI-H1650

∆ NCK1-AS1 (which regulated CDK1): ↓ proliferation

[46]

miR-186

A549, H1299, H460, and

BEAS-2

Lycorine treatment: ↑ levels of miR-186 and ↓ levels of CDK1: ↓ proliferation and ↑ apoptosis

CDK1 was a target of miR-186

[47]

GP130/STAT3 signaling pathway

A549, 1792, and HEK293T

↑↑ Iron-dependent CDK1 activity: ↑ activaty of the GP130/STAT3 signaling

[48]

TMPO-AS1 and miR-143-3p

16HBE, H1299, A549, 95D, and H125

∆ TMPO-AS1: ↓ cell viability, ↑ apoptosis

TMPO-AS1 regulated CDK1 via targeting miR-143-3p

[49]

miR-181a

16HBE,, H1299, and A549

↑↑ miR-181a: ↓ proliferation, colony formation, and invasion

[50]

miR-143 and miR-506

HFL-1, A549, H358, H69-AR, H358, H1975, and Calu-3

↑↑ miR-143 and miR-506: ↓ cell growth via targeting CDK1 and CDK4

[51]

miR-143 and miR-506

A549, HUVECs

↑↑ miR-143 and miR-506: ↓ angiogenesis, and ↑ cell cycle arrest via targeting CDK1, 4/6 genes, respectively

[52]

Melanoma

Sox2

1205Lu, WM239A, A375, and HCT116

CDK1 was found to be a new regulator of Sox2, so had tumor-initiating capacity in melanoma

[53]

CHPF

A375

CHPF was found to play its oncogenic role by regulating of CDK1 in malignant melanoma

[54]

Myeloid leukemia

EZH2 and DNMT3A

NIH3T3, 293T, and OCI-AML3

↑↑ DNMT3A mutation-induced CDK1: ↑ proliferation and ↓ apoptosis via modulating the interaction between EZH2 and DNMT3A

[55]

Nasopharyngeal carcinoma

cyclin B1

5-8F and 6-10B NPC

Proteasome inhibitors were found to participate in the accretion of CDK1/cyclin B1, so decreased paclitaxel-induced cell death

[56]

CDC25C/CDK1/Cyclin B1 pathway

CNE1 and CNE2

appropriate dose of tetrandrine and irradiation treatment: ↓ phosphorylation of CDK1 and CDC25C and ↑ expression of cyclin B1, ↑ cell cycle arrest

[57]

miR-195-3p

5–8 F, 6–10B, CNE1, CNE2, C666-1, and NP69

↑↑ miR-195-3p: ↑ radiosensitivity via targeting CDK1

[58]

Ovarian cancer

UBE2C

KOV3, A2780, SKOV3/DDP, and A2780/

DDP

∆ UBE2C: ↓ proliferation, cisplatin resistance, and ↑ apoptosis via downregulating CDK1

[59]

Chk1-CDC25C and P53-P21WAF1 signaling pathway

SK-OV-3 and OVCAR-3

∆ CDK1: ↓ proliferation, ↑ cell cycle arrest, and cell apoptosis

[60]

TONSL-AS1 and miR-490-3p

OVCAR3 OEC cell line

↑↑ TONSL-AS1: ↑ proliferation via targeting miR-490-3p and regulating CDK1

[61]

DLEU1/miR-490-3p/CDK1 axis

OVCAR3 and A2780

↑↑ DLEU1: ↑ proliferation, migration, and invasion, and ↓ apoptosis

DLEU1 was found to sponge miR-490-3p, so regulate CDK1

[62]

Pancreatic cancer

KRas

MiaPaCa2, Panc1, L3.6pl, A549, A427, H460, Calu6, SW620, DLD1, HCT8

AT7519, (a CDK1, 2, 7, and 9 inhibitor) induces apoptosis

CDK hyperactivation was linked with mt KRas dependency

[63]

miR-143 and miR-506

Panc-1 and MIA-PaCa-2

↑↑ miR-143 and miR-506: ↓ cell growth via targeting CDK1 and CDK4

[51]

Pancreatic ductal adenocarcinoma

_

PATU-T, Hs766T, and HPAF-II

Oxadiazole-based topsentin derivative (compound 6b): ↓ CDK1 expression, and ↑ apoptosis

[64]

_

different cell lines

Inaciclib was found to be an immune checkpoint inhibitor

∆ CDK1/2/5: ↓ UN-dependent STAT1 expression and activation, ↑ caspase-dependent apoptosis and histone-dependent ICD

[65]

Prostate cancer

TPX2, ERK/GSK3β/SNAIL signaling pathway

BPH-1, LNCaP, C4-2, PC-3, 22RV1

∆ TPX2: ↓ cell activity and migration, EMT process, ↓ expression of CDK1, ↓ the phosphorylation of ERK/GSK3β/SNAIL

[66]

ABCC5

C4-2, VCaP, ENZ-R, C4-2 and 22Rv1

↑↑ ABCC5: ↑ progression of cancer and resistance to Enzalutamide via the CDK1-mediated phosphorylation of AR

ABCC5 was found to inhibit ubiquitination of CDK1 via binding to CDK1

∆ CDK1: ↑ sensitivity to enzalutamide

[67]

  1. ∆ knock-down or deletion, ICD immunogenic cell death, EMT epithelial-mesenchymal transition, GC Gastric cancer, CRC Colorectal cancer