リケラボ論文検索は、全国の大学リポジトリにある学位論文・教授論文を一括検索できる論文検索サービスです。

リケラボ 全国の大学リポジトリにある学位論文・教授論文を一括検索するならリケラボ論文検索大学・研究所にある論文を検索できる

リケラボ 全国の大学リポジトリにある学位論文・教授論文を一括検索するならリケラボ論文検索大学・研究所にある論文を検索できる

大学・研究所にある論文を検索できる 「Anti-diabetic drug metformin inhibits cell proliferation and tumor growth in gallbladder cancer via G0/G1 cell cycle arrest」の論文概要。リケラボ論文検索は、全国の大学リポジトリにある学位論文・教授論文を一括検索できる論文検索サービスです。

コピーが完了しました

URLをコピーしました

論文の公開元へ論文の公開元へ
書き出し

Anti-diabetic drug metformin inhibits cell proliferation and tumor growth in gallbladder cancer via G0/G1 cell cycle arrest

山下 拓磨 香川大学 DOI:10.1097/CAD.0000000000000870

2020.03.24

概要

Gallbladder cancer is the most common biliary tract cancer with poor prognosis and wide variation in incidence rates worldwide, being very high in some countries in Latin America and Asia. Treatment of type 2 diabetes with metformin causes a reduction in the incidence of cancer. Till date, there are no reports on the anti-tumor effects of metformin in gall bladder cancer. Therefore, this study evaluated the effects of metformin on the proliferation of human gallbladder adenocarcinoma cells in vitro and in vivo, as well as explored the microRNAs associated with the anti-tumor effects of metformin. Metformin inhibited the proliferation in gallbladder adenocarcinoma cell lines NOZ, TGBC14TKB, and TGBC24TKB, and blocked the G0 to G1 transition in the cell cycle. This was accompanied by strong reduction in the expression of G1 cyclins, especially cyclin D1 and its catalytic subunits including cyclin-dependent kinase 4, and in retinoblastoma protein phosphorylation. In addition, metformin reduced the phosphorylation of receptor tyrosine kinases, especially Tie-2, ALK, PYK, EphA4, and EphA10, as well as angiogenesis-related proteins, including RANTES, TGF-β, and TIMP-1. Moreover, metformin also markedly altered microRNA expression profile leading to an anti-tumor effect. Treatment of athymic nude mice bearing xenograft tumors with metformin inhibited tumor growth. These results suggest that metformin may be used clinically for the treatment of gallbladder adenocarcinoma.

この論文で使われている画像

参考文献

1 Bal MM, Ramadwar M, Deodhar K, Shrikhande S. Pathology of Gallbladder

Carcinoma: Current Understanding and New Perspectives. Pathol Oncol Res.

2015;21:509-525.

2 Ha TY, Yoon YI, Hwang S, Park YJ, Kang SH, Jung BH, et al. Effect of Reoperation

on Long-Term Outcome of pT1b/T2 Gallbladder Carcinoma After Initial Laparoscopic

Cholecystectomy. J Gastrointest Surg. 2015;19:298–305.

3 Doval DC, Azam S, Sinha R, Batra U, Mehta A. Expression of epidermal growth

factor receptor, p53, Bcl2, vascular endothelial growth factor, cyclooxygenase-2, cyclin

D1, human epidermal receptor-2 and Ki-67: Association with clinicopathological

profiles and outcomes in gallbladder carcinoma. J Carcinog. 2014;13:10:3903-9.

Levy

AD,

Murakata

LA,

Rohrmann

CA

Jr.

Gallbladder

carcinoma:

radiologic-pathologic correlation. Radiographics. 2001;21(2):295–314. questionnaire,

549–555.

5 Goetze TO. Gallbladder carcinoma: Prognostic factors and therapeutic options. World

J Gastroenterol. 2015;21(43):12211–7.

26

6 Soraya J Kaewpitoon, Ryan A Loyd, Ratana Rujirakul, Sukij Panpimanmas, Likit

Matrakool,

Taweesak

Tongtawee,

et

al.

Benefits

of

Metformin

Use

for

Cholangiocarcinoma. Asian Pac J Cancer Prev. 2015;16(18):8079–83.

7 Ren HB, Yu T, Liu C, Li YQ. Diabetes mellitus and increased risk of biliary tract

cancer: systematic review and meta-analysis. Cancer Causes Control. 2011;22:837–47.

8 Palmer WC, Patel T. Are common factors involved in the pathogenesis of primary

liver cancers? A meta-analysis of risk factors for intrahepatic cholangiocarcinoma. J

Hepatol. 2012;57:69–76.

9 Ben Sahra I, Laurent K, Loubat A, Giorgetti-Peraldi S, Colosetti P, Auberger P, et al.

The antidiabetic drug metformin exerts an antitumoral effect in vitro and in vivo

through a decrease of cyclin D1 level. Oncogene. 2008;27:3576–86.

10 Anisimov VN, Egormin PA, Piskunova TS, Popovich IG, Tyndyk ML, Yurova MN,

et al. Metformin extends life span of HER-2/neu transgenic mice and in combination

with melatonin inhibits growth of transplantable tumors in vivo. Cell Cycle.

2010;9:188–97.

11 Z Bao B, Wang Z, Ali S, Ahmad A, Azmi AS, Sarkar SH, et al. Metformin Inhibits

27

Cell Proliferation, Migration and Invasion by Attenuating CSC Function Mediated by

Deregulating miRNAs in Pancreatic Cancer Cells. Cancer Prev Res (Phila).

2012;5(3):355–64.

12 Kato K, Gong J, Iwama H, Kitanaka A, Tani J, Miyoshi H, et al. The antidiabetic

drug metformin inhibits gastric cancer cell proliferation in vitro and in vivo. Mol

Cancer Ther. 2012;11:549–60.

13 Fujihara S, Kato K, Morishita A, Iwama H, Nishioka T, Chiyo T, et al. Antidiabetic

drug metformin inhibits esophageal adenocarcinoma cell proliferation in vitro and in

vivo. Int J Oncol. 2015;46(5):2172–80.

14 Miyoshi H, Kato K, Iwama H, Maeda E, Sakamoto T, Fujita K, et al. Effect of the

anti-diabetic drug metformin in hepatocellular carcinoma in vitro and in vivo. Int J

Oncol. 2014;45:322–32.

15 Morishita A, Masaki T. miRNA in hepatocellular carcinoma. Hepatol Res.

2015;45:128–41.

16 Fujimori T, Kato K, Fujihara S, Iwama H, Yamashita T, Kobayashi K, et al.

Antitumor effect of metformin on cholangiocarcinoma: In vitro and in vivo studies.

28

Oncol Rep. 2015;34(6):2987–96.

17 Pernicova I, Korbonits M. Metformin-mode of action and clinical implications for

diabetes and cancer. Nat Rev Endocrinol. 2014;10:143–56.

18 Kourelis TV, Siegel RD. Metformin and cancer: new applications for an old drug.

Med Oncol. 2012;29:1314–27.

19 Hui AM, Cui X, Makuuchi M, Li X, Shi YZ, Takayama T. Decreased p27(Kip1)

expression and cyclin D1 overexpression, alone and in combination, influence

recurrence and survival of patients with resectable extrahepatic bile duct carcinoma.

Hepatology. 1999;30(5):1167–73.

20 Sugimachi K, Aishima S, Taguchi K, Tanaka S, Shimada M, Kajiyama K, et al. The

role of overexpression and gene amplification of cyclin D1 in intrahepatic

cholangiocarcinoma. J Hepatol. 2001;35(1):74–9.

21 Ling S, Feng T, Ke Q, Fan N, Li L, Li Z, et al Metformin inhibits proliferation and

enhances chemosensitivity of intrahepatic cholangiocarcinoma cell lines. Oncol Rep.

2014;31(6):2611–8.

22 Laemmli UK. Cleavage of structural proteins during the assembly of the head of

29

bacteriophage T4. Nature. 1970;227:680–5.

23 Towbin H, Staehelin T, Gordon J. Electrophoretic transfer of proteins from

polyacrylamide gels to nitrocellulose sheets: procedure and some applications. Proc

Natl Acad Sci USA. 1979;76:4350–4.

24 D'Incalci M, Colombo T, Ubezio P, Nicoletti I, Giavazzi R, Erba E, et al. The

combination of yondelis and cisplatin is synergistic against human tumor xenografts.

Eur J Cancer. 2003;39:1920–6.

25 Zhou G, Myers R, Li Y, Chen Y, Shen X, Fenyk-Melody J, et al. Role of

AMP-activated protein kinase in mechanism of metformin action. J Clin Invest.

2001;108(8):1167–74.

26 Chaiteerakij R, Yang JD, Harmsen WS, Slettedahl SW, Mettler TA, Fredericksen ZS,

et al. Risk factors for intrahepatic cholangiocarcinoma: association between metformin

use and reduced cancer risk. Hepatology. 2013;57(2):648–55.

27 Han S, Kim HY, Park K, Lee MS, Kim HJ, Kim YD. Expression of p27Kip1 and

cyclin D1 proteins is inversely correlated and is associated with poor clinical outcome

in human gastric cancer. J Surg Oncol. 1999;71:147–54.

30

28 Aoyagi K, Koufuji K, Yano S, Murakami N, Terasaki Y, Yamasaki Y, et al.

Immunohisto- chemical study on the expression of cyclin D1 and E in gastric cancer.

Kurume Med J. 2000;47:199–203.

29 Masaki T, Shiratori Y, Rengifo W, Igarashi K, Yamagata M, Kurokohchi K, et al.

Cyclins and cyclin- dependent kinases: Comparative study of hepatocellular carcinoma

versus cirrhosis. Hepatology. 2003;37:534–43.

30 Masaki T, Shiratori Y, Rengifo W, Igarashi K, Matsumoto K, Nishioka M, et al.

Hepatocellular carcinoma cell cycle: study of Long-Evans cinnamon rats. Hepatology.

2000;32:711–20.

31 Sherr CJ. Mammalian G1 cyclins. Cell. 1993;73:1059–65.

32 Sherr CJ. G1 phase progression: cyclin g on cue. Cell. 1994;79:551–5.

33 Pollak M. Insulin and insulin-like growth factor signaling in neoplasia. Nat Rev

Cancer. 2008;8:915–28.

34 Narayanan R, Kim HN, Narayanan NK, Nargi D, Narayanan B. Epidermal growth

factor-stimulated human cervical cancer cell growth is associated with EGFR and cyclin

D1 activation, independent of COX-2 expression levels. Int J Oncol. 2011;40:13–20.

31

35 Oki M, Yamamoto H, Taniguchi H, Adachi Y, Imai K, Shinomura Y. Overexpression

of the receptor tyrosine kinase EphA4 in human gastric cancers. World J Gastroenterol.

2008;14:5650–6.

36 Li Y, Jin L, Ye F, Ma Q, Yang Z, Liu D, et al. Isoform expression of EPHA 10 protein

mediate breast cancer progression by regulating the E-cadherin and

β-catenin complex.

Oncotarget. 2017;8:30344–56.

37 Zhang JH, Wang LH, Li XJ, Wang AP, Reng LQ, Xia FG, et al. Expression of

Ang-2/Tie-2 and PI3K/AKT in Colorectal Cancer. ASIAN PACIFIC JOURNAL OF

CANCER PREVENTION. 2014:15:8651-6.

38 Lipimski CA, Tran NL, Menashi E, Rohl C, Kloss J, Bay RC, et al. The tyrosine

kinase pyk2 promotes migration and invasion of grioma cells. Neoplasia.

2005;7:435–45.

39 Yoshida R, Sasaki T, Minami Y, Hibino Y, Okumura S, Sado M, et al. Activation of

Src signaling mediates acquired to ALK inhibition in lung cancer. Int J Oncol.

2017;51:1533–40.

40 Simone V, Brunetti O, Lupo L, Testini M, Maiorano E, Simone M, et al. Targeting

32

Angiogenesis in Biliary Tract Cancers: An Open Option. Int J Mol Sci. 2017;18(2):418.

41 Lv D, Zhang Y, Kim HJ, Zhang L, Ma X. CCL5 as a potential immunotherapeutic

target in triple-negative breast cancer. Cell Mol Immunol. 2013;10:30310.

42 Principe DR, Doll JA, Bauer J, Jung B, Munshi HG, Bartholin L, et al. TGF-β:

duality of function between tumor prevention and carcinogenesis. J Natl Cancer Inst.

2014;106, doi:10.1093/jnci/djt369.

43 Gong Y, Scott E, Lu R, Xu Y, Oh WK, Yu Q. TIMP-1 promote accumulation of

cancer associated fibroblast and cancer progression. PLoS One. 2013;8: e77366. 2013.

44 He D, Wang J, Zhang C, Shan B, Deng X, Li B, et al. Down-regulation of

miR-675-5p contributes to tumor progression and development by targeting

pro-tumorigenic GPR55 in non-small cell lung cancer. Mol Cancer. 2015;14:73.

45 Zhu M, Chen Q, Liu X, Sun Q, Zhao X, Deng R, et al. lncRNA H19/miR-675 axis

represses prostate cancer metastasis by targeting TGFBI. FEBS J. 2014;281:3766–75.

46 Schmitz KJ, Helwig J, Bertram S, Sheu SY, Suttorp AC, Seggewiss J, et al.

Differential expression of microRNA-675, microRNA-139-3p and microRNA-335 in

benign and malignant adrenocortical tumours. J Clin Pathol. 2011;64:529–35.

33

Figure legends

Figure 1. Metformin inhibits the proliferation of cultured GBC cells. NOZ,

TGBC14TKB, and TGBC24TKB cells were seeded in 96-well plates. Metformin (1, 5,

and 10 mmol/L) or vehicle was added to the culture medium; cells were subjected to the

CCK assay at 24–72 h after metformin treatment. Cell viability was assayed daily from

0 to 72 h; viability of metformin-treated cells at each time point differed significantly

from the control cells (P* < 0.05).

34

Figure 2. (A) Flow cytometric analysis of proliferating NOZ cells at 24 h after addition

of 10 mmol/L metformin (Met). Metformin blocked the cell cycle at G0/G1. Results are

representative of three independent experiments. (B) Expressions of cyclin D1, Cdk4,

Cdk6, cyclin E, phosphorylated Rb (pRb), and Rb in NOZ cells at 24, 48, and 72 h after

the addition of 10 mmol/L metformin. Protein expression was assayed by western

blotting.

Figure 3. (A) Template showing the location of tyrosine kinase antibodies spotted onto a

human phospho-RTK array. (B) Representative expression of various phosphorylated

tyrosine kinase receptors in NOZ cells treated with or without metformin. (C) The

densitometric of ratios p-Tie, p-PYK, p-ALK, p-EGFR, p-EphA4, and p-EphA10 spots

of metformin-treated to untreated cells were 31.8 %, 32.1 %, 8.4 %, 72.1 %,1.3 %, and

14.6%, respectively.

Figure 4. (A) Template showing the location of angiogenesis related proteins spotted

onto a human angiogenesis array. (B) Representative expression of various

35

angiogenesis-related proteins in NOZ cells treated with or without metformin. (C) The

densitometric ratios of RANTES, TGF-β, and TIMP spots of the metformin-treated cells

to untreated cells were 0.21 times,, 20 times and 0.37 times, respectively (Fig. 4C).

, respectively.

Figure 5. Hierarchical clustering of NOZ cells with and without metformin treatment.

NOZ cells were clustered according to the expression profiles of 35 miRNAs

differentially expressed in NOZ cells with and without metformin treatment. The

analyzed samples are shown in columns and the miRNAs are presented in rows. The

miRNA clustering color scale shown at the top depicts relative expression levels of the

miRNAs, with red and blue representing high and low expression levels, respectively.

Figure 6. In vivo anti-tumor effects of metformin on established gall bladder cancer

xenografts in nude mice. NOZ cells were implanted subcutaneously into the flanks of

nude mice. When tumors became palpable, 0 or 2 mg metformin was injected

intraperitoneally for 23 days, 5 times per week. (A) Representative images of gross

36

NOZ tumors from nude mice treated with vehicle (i) or 2 mg metformin (ii). (B) Tumor

growth curves in control and metformin groups. Tumor volume (mm3) was calculated as

(tumor length (mm) × tumor width (mm) 2)/2. Tumors were significantly smaller in

metformin-treated than in vehicle-treated mice. Each point represents the mean ±

standard deviation of six animals. *P = 0.01 by two-way ANOVA

Table 1. Statistical results and chromosomal locations of miRNAs in NOZ cells treated

with or without metformin.

Figure 1

Met (-)

G0/G1:36.3%

S:47.4%

G2/M:16.3%

72h

Met (+)

Figure 2

G0/G1:50.6%

S:35.2%

G2/M:14.1%

Met

(10mM)

(-)

24h

(+)

(-)

48 h

(+)

(-)

72h

(+)

β-actin

Rb

pRb

Cdk6

Cdk4

CyclinE

CyclinD1

Tie-2

Tie-2

Met (-)

ALK

150

Axl

PYK

PYK

Reference

Spots

Met (+)

ALK

EphB3

EphB1

TrkB

MSP R

PYK

EphB2

TrkC

PDGF Rα

EphA10

EphA10

EphB4

VEGF R1

PDGF Rβ

Flt-3

met+

50

100

met-

EphA10

met+

150

50

50

met+

100

100

met-

150

150

ALK

met-

50

EphA5

EphA2

ROR1

50

Tie-2

DDR2

EphA1

C-Ret

100

DDR1

MuSK

M-CSF R

100

150

ALK

VEGF R3

Figure 3

EphA4

EphA4

EphB6

VEGF R2

SCF R

EphA10

EphA3

ROR2

met-

EphA4

met-

PYK

PBS

EphA4

Tie-1

EphA7

IGF-1 R

EphA6

Insulin R

TrkA

FGF R4

Tie-2

FGF R3

HGF R

FGF R2α

Mer

FGF R1

Dtk

ErbB4

ErbB2

EGF R

ErbB3

Reference

Spots

Reference

Spots

met+

met+

RANTES

IFN gamma

RANTES

RANTES

BLANK

BLANK

GRO

PLGF

PLGF

BLANK

BLANK

Met (-)

IFN gamma

GRO

TIMP-1

POS

POS

TGF beta1

POS

POS

RANTES

BLANK

BLANK

Figure 4

TIMP-1

BLANK

BLANK

TIMP-1

TIMP-1

IL-6

IL-6

NEG

NEG

Met (+)

TGF beta1

TGF beta 1

TGF beta 1

IGF-1

IGF-1

NEG

NEG

BLANK

BLANK

TIMP-2

TIMP-2

IL-8

IL-8

ANG

ANG

BLANK

BLANK

THPO

THPO

Leptin

Leptin

EGF

EGF

50

100

150

1000

2000

3000

50

100

150

NEG

NEG

VEGF

VEGF

POS

POS

VEGF-D

VEGF-D

met-

met-

met+

met+

TIMP-1

TGF beta1

met+

PDGF-BB

PDGF-BB

b FGF

b FGF

RANTES

met-

MCP-1

MCP-1

ENA-78

ENA-78

Metformin

Figure 5

Control

Figure 6

i) Control

ii) Met 2 mg/d

...

参考文献をもっと見る

全国の大学の
卒論・修論・学位論文

一発検索!

この論文の関連論文を見る