• 제목/요약/키워드: MDA MB-231 cells

검색결과 245건 처리시간 0.029초

Evaluation of Anti-cancer and Anti-proliferative Activity of Medicinal Plant Extracts (Saffron, Green Tea, Clove, Fenugreek) on Toll Like Receptors Pathway

  • Ajmal, Sidra;Shafqat, Mahwish;Ajmal, Laiba;Younas, Hooria;Tasadduq, Raazia;Mahmood, Nasir
    • Natural Product Sciences
    • /
    • 제28권3호
    • /
    • pp.121-129
    • /
    • 2022
  • Despite considerable efforts, cancer remains an aggressive killer worldwide. Chemotherapeutic drugs that are currently in use lead to destructive side effects and have not succeeded in fulfilling expectations. For centuries, medicinal plants are used for treating various diseases and are also known to have anticancer activity. The main aim of this research was to evaluate antiproliferative activity of saffron, clove, fenugreek, and green tea on Vero and MDA-MB-231 cell lines and to subsequently analyze the effect of these extracts on IRAK-4, TAK1, IKK-alpha, IKK-beta, NF-Kappa B, IRF3, IRF7 genes in Toll Like Receptors (TLRs) pathway. Antiproliferative assay was done by Neutral Red Dye uptake assay. Methanolic extract of green tea was found to be most effective against both cell lines as IC50 was achieved at least concentration of the extract. For molecular studies, MDAMB-231 cells were sensitized with methanolic extract of green tea at same IC50, and RT-PCR was performed to determine the relative expression of genes. Expression of IRAK-4, TAK1, IKK-beta, NF-Kappa B, IRF3 genes was down regulated and IRF7 and IKKalpha was upregulated. Green tea has a potential cytotoxic effect on both cell lines which was demonstrated by its effect on the expression of (TLRs) pathway genes.

Synergistic Effects of Tamoxifen and Tranilast on VEGF and MMP-9 Regulation in Cultured Human Breast Cancer Cells

  • Darakhshan, Sara;Bidmeshkipour, Ali;Khazaei, Mozafar;Rabzia, Arezou;Ghanbari, Ali
    • Asian Pacific Journal of Cancer Prevention
    • /
    • 제14권11호
    • /
    • pp.6869-6874
    • /
    • 2013
  • Background: Vascular endothelial growth factor and matrix metalloproteinases are two important factors for angiogenesis associated with breast cancer growth and progression. The present study was aimed to examine the effects of tamoxifen and tranilast drugs singly or in combination on proliferation of breast cancer cells and also to evaluate VEGF and MMP-9 expression and VEGF secretion levels. Materials and Methods: Human breast cancer cell lines, MCF-7 and MDA-MB-231, were treated with tamoxifen and/or tranilast alone or in combination and percentage cell survival and proliferative activity were evaluated using LDH leakage and MTT assays. mRNA expression and protein levels were examined by real-time RT-PCR and ELISA assay, respectively. Results: LDH and MTT assays showed that the combined treatment of tamoxifen and tranilast resulted in a significant decrease in cell viability and cell proliferation compared with tamoxifen or tranilast treatment alone, with significant decrease in VEGF mRNA and protein levels. We also found that tamoxifen as a single agent rarely increased MMP-9 expression. A decrease in MMP-9 expression was seen after treatment with tranilast alone and in the combined treatment MMP-9 mRNA level was decreased. Conclusions: This combination treatment can able to inhibit growth, proliferation and angiogenesis of breast cancer cells.

Synergistic Induction of Apoptosis by the Combination of an Axl Inhibitor and Auranofin in Human Breast Cancer Cells

  • Ryu, Yeon-Sang;Shin, Sangyun;An, Hong-Gyu;Kwon, Tae-Uk;Baek, Hyoung-Seok;Kwon, Yeo-Jung;Chun, Young-Jin
    • Biomolecules & Therapeutics
    • /
    • 제28권5호
    • /
    • pp.473-481
    • /
    • 2020
  • Axl receptor tyrosine kinase has been implicated in cancer progression, invasion, and metastasis in various cancer types. Axl overexpression has been observed in many cancers, and selective inhibitors of Axl, including R428, may be promising therapeutic agents for several human cancers, such as breast, lung, and pancreatic cancers. Here, we examined the cell growth inhibition mediated by R428 and auranofin individually as well as in combination in the human breast cancer cell lines MCF-7 and MDA-MB-231 to identify new advanced combination treatments for human breast cancer. Our data showed that combination therapy with R428 and auranofin markedly inhibited cancer cell proliferation. Isobologram analyses of these cells indicated a clear synergism between R428 and auranofin with a combination index value of 0.73. The combination treatment promoted apoptosis as indicated by caspase 3 activation and poly (ADP-ribose) polymerase cleavage. Cancer cell migration was also significantly inhibited by this combination treatment. Moreover, we found that combination therapy significantly increased the expression level of Bax, a mitochondrial proapoptotic factor, but decreased that of the X-linked inhibitor of apoptosis protein. Furthermore, the suppression of cell viability and induction of Bax expression by the combination treatment were recovered by treatment with N-acetylcysteine. In conclusion, our data demonstrated that combined treatment with R428 and auranofin synergistically induced apoptosis in human breast cancer cells and may thus serve as a novel and valuable approach for cancer therapy.

AKT1-targeted proapoptotic activity of compound K in human breast cancer cells

  • Choi, Eunju;Kim, Eunji;Kim, Ji Hye;Yoon, Keejung;Kim, Sunggyu;Lee, Jongsung;Cho, Jae Youl
    • Journal of Ginseng Research
    • /
    • 제43권4호
    • /
    • pp.692-698
    • /
    • 2019
  • Background: Breast cancer is a severe disease and the second leading cause of cancer death in women worldwide. To surmount this, various diagnosis and treatment options for breast cancer have been developed. One of the most effective strategies for cancer treatment is to induce apoptosis using naturally occurring compounds. Compound K (CK) is a ginseng saponin metabolite generated by human intestinal bacteria. CK has been studied for its cardioprotective, antiinflammatory, and liver-protective effects; however, the role of CK in breast cancer is not fully understood. Methods: To investigate the anticancer effects of CK in SKBR3 and MDA-MB-231 cells, cell viability assays and flow cytometry analysis were used. In addition, the direct targets of CK anticancer activity were identified using immunoblotting analysis and overexpression experiments. Invasion, migration, and clonogenic assays were carried out to determine the effects of CK on cancer metastasis. Results: CK-induced cell apoptosis in SKBR3 cells as determined through 3-(4-5-dimethylthiazol-2-yl)-2-5-diphenyltetrazolium bromide assays, propidium iodide (PI) and annexin V staining, and morphological changes. CK increased the cleaved forms of caspase-7, caspase-8, and caspase-9, whereas the expression of Bcl-2 was reduced by CK. In assays probing the cell survival pathway, CK activated only AKT1 and not AKT2. Moreover, CK inhibited breast cancer cell invasion, migration, and colony formation. Through regulation of AKT1 activity, CK exerts anticancer effects by inducing apoptosis. Conclusion: Our results suggest that CK could be used as a therapeutic compound for breast cancer.

Genetic disruption of ATAT1 causes RhoA downregulation through abnormal truncation of C/EBPβ

  • Jee-Hye Choi;Jangho Jeong;Jaegu Kim;Eunae You;Seula Keum;Seongeun Song;Ye Eun Hwang;Minjoo Ji;Kwon-Sik Park;Sangmyung Rhee
    • BMB Reports
    • /
    • 제57권6호
    • /
    • pp.293-298
    • /
    • 2024
  • Microtubule acetylation has been shown to regulate actin filament dynamics by modulating signaling pathways that control actin organization, although the precise mechanisms remain unknown. In this study, we found that the downregulation of microtubule acetylation via the disruption ATAT1 (which encodes α-tubulin N-acetyltransferase 1) inhibited the expression of RhoA, a small GTPase involved in regulating the organization of actin filaments and the formation of stress fibers. Analysis of RHOA promoter and chromatin immunoprecipitation assays revealed that C/EBPβ is a major regulator of RHOA expression. Interestingly, the majority of C/EBPβ in ATAT1 knockout (KO) cells was found in the nucleus as a 27-kDa fragment (referred to as C/EBPβp27) lacking the N-terminus of C/EBPβ. Overexpression of a gene encoding a C/EBPβp27-mimicking protein via an N-terminal deletion in C/EBPβ led to competitive binding with wild-type C/EBPβ at the C/EBPβ binding site in the RHOA promoter, resulting in a significant decrease of RHOA expression. We also found that cathepsin L (CTSL), which is overexpressed in ATAT1 KO cells, is responsible for C/EBPβp27 formation in the nucleus. Treatment with a CTSL inhibitor led to the restoration of RHOA expression by downregulation of C/EBPβp27 and the invasive ability of ATAT1 KO MDA-MB-231 breast cancer cells. Collectively, our findings suggest that the downregulation of microtubule acetylation associated with ATAT1 deficiency suppresses RHOA expression by forming C/EBPβp27 in the nucleus through CTSL. We propose that CTSL and C/EBPβp27 may represent a novel therapeutic target for breast cancer treatment.

누룩(Rhizopus oryzae KSD-815)으로부터 분리한 지질화합물의 세포독성 및 항염증 활성 (Cytotoxic and Anti-inflammatory Activities of Lipids from the Nuruk (Rhizopus oryzae KSD-815))

  • 곽호영;이상진;이대영;배낙현;정낙훈;홍성렬;김계원;백남인
    • Applied Biological Chemistry
    • /
    • 제51권2호
    • /
    • pp.142-147
    • /
    • 2008
  • Rhizopus oryzae KSD-815를 밀에 접종하여 만든 누룩(Rhizopus oryzae KSD-815)에서 4종의 지질 화합물을 분리하여 화학구조를 동종하였다. 건조된 누룩을 실온에서 80% MeOH 수용액으로 추출하고 이추출물을 EtOAc 분획, n-BuOH 분획, $H_2O$ 분획으로 나누었다. EtOAc분획은 다시 80% MeOH 과 n-hexane 으로 분획하였다. n-Hexane 분획에 대해 silica gel 및 ODS column chromatography를 반복 실시하여 4종의 지질 화합물을 분리, 정제하였다. NMR, IR, GC/MS 등을 통하여 화합물 1(linolenic acid methyl ester), 화합물 2(palmitic acid methyl ester), 화합물 3(linoleic acid), 화합물 4(palmitic acid)의 구조를 결정하였다. 이 지방산 화합물의 세포독성을 평가하기 위해 인체 유래 유방암(MDA-MB-231)과 간암(SK-HEP-1)세포주에 대해 MTT assay를 수행하였다. 두 암 세포주에서 화합물 1(linolenic acid methyl ester)과 화합물 3(linoleic acid)은 농도의존적인 세포독성을 확인하였다. 또 염증반응 매개체의 일종인 nitric oxice(NO)의 생성 억제 활성을 Griess 방법으로 평가한 결과, 화합물 3(linoleic acid)은 LPS와 IFN-${\gamma}$에 의해 유도 된 NO 생성도 저해하는 것을 RAW264.7 세포에서 확인하였다.

암억제 유전자 p53에 의한 insulin-like growth factor binding protein-3의 발현과 glycosylation를 통한 항암작용 (The anti-tumor mechanisms of p53 through the regulation of expression and glycosylation of insulin-like growth factor binding protein-3)

  • 김선영;김세림;이정창;이호근;이대열;황평한
    • Clinical and Experimental Pediatrics
    • /
    • 제49권4호
    • /
    • pp.431-438
    • /
    • 2006
  • 목 적 : 새로운 암억제 유전자로 알려진 IGFBP-3의 주요 기능은 IGF-I과 IGF-II와 결합을 하여 IGF의 기능을 조절하는 IGF dependent mechanism과 IGFBP-3 자체가 IGF와 결합과는 무관하게 세포의 apoptosis를 유도하는 IGF independent mechanism이 보고되었다. 암억제 유전자 p53의 대표적인 항암 기전의 하나는 직접 IGFBP-3의 발현을 증가시키며, 발현된 IGFBP-3는 암세포의 apoptosis를 유도시킨다. IGFBP-3의 항암작용은 보고되었지만, IGFBP-3의 변역 후 변형에 의한 항암기전은 전혀 밝혀져 있지 않다. 본 연구에서는 p53의 항암기전과 관련하여 IGFBP-3의 당화에 관련된 기전을 밝히고, IGFBP-3 당화의 의미를 규명하였다. 방 법 : 실험 세포주로는 p53의 변이를 보이며 p53의 발현이 일반세포에 비교하여 낮은 특징을 갖고 있는 사람의 유방암세포인 MDA-MB-231를 사용하였으며, Ad/p53과 Ad/IGFBP-3 아데노바이러스를 감염시킨 후 IGFBP-3의 발현 변화와 apoptosis 기전을 분석하였다. glycosylation 억제제로 알려져 있는 tunicamycin을 처리하여 당화의 정도를 조사하였다. 결 과 : 실험 세포에 Ad/p53을 감염시켜 p53을 발현시킨 결과 성장의 억제와 apoptosis가 유도되었고, IGFBP-3의 발현이 현저하게 증가되었으며, 특히 IGFBP-3의 당화 형태를 증가시켰다. 당화된 IGFBP-3의 증가는 세포의 apoptosis의 유도가 촉진되었으며, 이러한 IGFBP-3의 당화는 p53과 IGFBP-3의 발현을 동시에 유도시킨 결과 더욱 항진되었다. 결 론 : 이상의 연구에서 IGFBP-3의 암억제 능력의 향상은 p53에 의한 IGFBP-3의 당화의 증가를 통하여 안정화됨으로서 나타나고 있음을 알 수 있었다. 이는 p53과 IGFBP-3를 이용한 혼합유전자 치료가 가능할 것으로 사료된다.

마늘추출물에 의한 암세포의 이동 저하 (Inhibition of Cancer Cell Migration by Compounds from Garlic Extracts)

  • 김은경;윤성지;하정민;진인혜;김영환;김선근;박다정;최영환;윤식;김치대;배순식
    • 생명과학회지
    • /
    • 제21권6호
    • /
    • pp.767-774
    • /
    • 2011
  • 세포의 이동은 많은 생리적 반응뿐만 아니라 암 세포 침윤과 전이에 중요한 역할을 수행한다. 본 연구에서는 마늘이 암세포의 이동에 미치는 영향을 확인하기 위해, 표준 마늘과 흑마늘을 준비하고 이들을 각각 물을 이용하여 추출하거나 건조하여 추출한 추출물 4 종류를 이용하여 항침윤성과 항전이성에 대해 조사하였다. 실험결과, 암세포의 이동은 건조 후 헥산으로 추출한 분획에 암세포의 이동 억제 활성이 관찰되었다. 이 분획을 박막 크로마토그래피를 이용하여 분리정제하였으며, 이를 inhibitor of cancer metastasis from garlic #27 (ICMG-27)이라 명명하였다. ICMG-27 (6 ${\mu}g/ml$)을 세포에 처리하였을 때, IGF-1에 의한 OVCAR-3와 NIH-3T3 세포의 이동을 억제함을 확인하였다. 그러나 ICMG-27은 mouse embryonic fibroblast (MEF) 세포에서 IGF-1에 의한 이동에는 영향을 주지 않았다. 이러한 ICMG-27은 OVCA-3, SKOV-3와 MDA-MB-231 세포와 같은 암세포에서 모두 IGF-1에 의한 이동을 억제함을 관찰하였다. 마지막으로 세포이동을 일으키는 인자에 따른 ICMG-27의 영향을 확인한 것으로, IGF-1, lysophosphatidic acid (LPA), sphingosine-1-phosphate (S1P), leukotriene B4 (LTB4) 그리고, angiotensinII (AngII)에 의한 OVCAR-3 세포의 이동을 모두 억제하였다. 이러한 결과를 바탕으로, ICMG-27은 암세포의 이동을 유도하는 많은 인자들에 의한 필수적인 단계를 차단함으로써, 암세포의 이동을 억제하는 것을 확인 할 수 있었으며, ICMG-27에 의한 암세포의 항 침윤 메커니즘의 규명은 암환자의 치료에 기초적인 발판을 제공할 것입니다.

Ethanol but not Aqueous Extracts of Tubers of Sauromatum Giganteum(Engl.) Cusimano and Hett Inhibit Cancer Cell Proliferation

  • Gao, Shi-Yong;Li, Jun;Wang, Long;Sun, Qiu-Jia;Gong, Yun-Fei;Gang, Jian;Su, Yi-Jun;Ji, Yu-Bin
    • Asian Pacific Journal of Cancer Prevention
    • /
    • 제15권24호
    • /
    • pp.10613-10619
    • /
    • 2015
  • Background: Both alcohol and aqueous extracts of Sauromatum giganteum(Engl.) Cusimano and Hett, the dried root tuber of which is named Baifuzi in Chinese, have been used for folklore treatment of cancer in Northeast of China. However, little is known about which is most suitable to the cancer therapy. Materials and Methods: Serum pharmacology and MTT assays were adopted to detect the effects of ethanol and aqueous extracts of Sauromatum giganteum(Engl.) Cusimano and Hett, prepared by heat reflux methods, on proliferation of different cancer cells. Results: Cancer cells treated with medium supplemented with 10%, 20%, 40% serum(v/v) containing ethanol extract had a decline in viability, with inhibition rates of 7.69%, 21.8%, 41.9% in MCF-7 cells, 42.8%, 48.1%, 51.8% in SGC-7901 cells, 44.1%, 49.2%, 53.7% in SMMC-7721 cells, 6.8%, 15.2%, 39.8% in HepG2 cells, 7.57%, 16.3%, 36.2% in HeLa cells, 6.24%, 12.5%, 27.4% in A549 cells, and 7.20%, 17.5%, 31.3% in MDA-MB-231 cells, respectively. Viability in the aqueous extract groups was no different with that of controls. Conclusions: An ethanol extract of Sauromatum giganteum(Engl.) Cusimano and Hett inhibited the proliferation of SMMC-7721, SGC-7901 and MCF-7 cells, which supports the use of alcoholic but not aqueous extracts for control of sensive cancers, which might include hepatocarcinoma, gastric cancer and breast cancer.

Anti-Proliferative and Pro-Apoptotic Activities of 4-Methyl-2,6-bis(1-phenylethyl)phenol in Cancer Cells

  • Sung, Nak Yoon;Kim, Seung Cheol;Kim, Yun Hwan;Kim, Gihyeon;Lee, Yunmi;Sung, Gi-Ho;Kim, Ji Hye;Yang, Woo Seok;Kim, Mi Seon;Baek, Kwang-Soo;Kim, Jong-Hoon;Cho, Jae Youl
    • Biomolecules & Therapeutics
    • /
    • 제24권4호
    • /
    • pp.402-409
    • /
    • 2016
  • It has been found that 4-isopropyl-2,6-bis(1-phenylethyl)phenol (KTH-13), a novel compound isolated from Cordyceps bassiana, is able to suppress tumor cell proliferation by inducing apoptosis. To mass-produce this compound, we established a total synthesis method. Using those conditions, we further synthesized various analogs with structural similarity to KTH-13. In this study, we aimed to test their anti-cancer activity by measuring anti-proliferative and pro-apoptotic activities. Of 8 compounds tested, 4-methyl-2,6-bis(1-phenylethyl)phenol (KTH-13-Me) exhibited the strongest anti-proliferative activity toward MDA-MB 231 cells. KTH-13-Me also similarly suppressed the survival of various cancer cell lines, including C6 glioma, HCT-15, and LoVo cells. Treatment of KTH-13-Me induced several apoptotic signs in C6 glioma cells, such as morphological changes, induction of apoptotic bodies, and nuclear fragmentation and chromatin condensation. Concordantly, early-apoptotic cells were also identified by staining with FITC-Annexin V/PI. Moreover, KTH-13-Me highly enhanced the activation of caspase-3 and caspase-9, and decreased the protein level of Bcl-2. In addition, the phosphorylation levels of Src and STAT3 were diminished in KTH-13-Me-treated C6 cells. Therefore, these results suggest that KTH-13-Me can be developed as a novel anti-cancer drug capable of blocking proliferation, inducing apoptosis, and blocking cell survival signaling in cancer cells.