• Title/Summary/Keyword: MDA-MB-231 cell

Search Result 240, Processing Time 0.03 seconds

Macrophage and Anticancer Activities of Feed Additives on β-Glucan from Schizophyllum commune in Breast Cancer Cells (치마버섯균 유래의 베타글루칸에 대한 사료첨가제로서의 대식세포 기능 활성 및 유방암 세포주에서의 항암효능 효과)

  • Lee, Jin-Seok;Lee, Seung-Ho;Jang, Yong-Man;Lee, Jong-Dae;Lee, Byoung-Hee;Jung, Ji-Youn
    • Journal of the Korean Society of Food Science and Nutrition
    • /
    • v.40 no.7
    • /
    • pp.949-955
    • /
    • 2011
  • [ ${\beta}$ ]Glucan is a polysaccharide expressed on the cell walls of fungi. It is known that ${\beta}$-glucan is recognized by a family of C-type lectin receptors, dectin-1, which is expressed mainly on myeloid immune cells, including macrophages, neutrophils and dendritic cells. Raw 264.7 cells were treated with ${\beta}$-glucan from Schizophyllum commune. ${\beta}$-Glucan was not cytotoxic up to 400 ${\mu}g$/mL as measured by MTT assay. To measure the activity of macrophages, NO and TNF-${\alpha}$ assays were performed in Raw 264.7 cells. Treatment with ${\beta}$-glucan for 24 hr significantly increased production of NO and TNF-${\alpha}$ compared with control groups (p<0.05), indicating activation of macrophages. To measure inhibition of breast cancer cell proliferation, MTT assay was performed in MDA-MB-231 cells. Cell viability was significantly decreased in the group treated with 400 ${\mu}g$/mL of ${\beta}$-glucan for 48 hr (p<0.05) compared to the control group. However, tumor volume was decreased in the groups administered 200 ${\mu}g$ of ${\beta}$-glucan/mouse compared to the control group. These results indicate that ${\beta}$-glucan inhibits breast cancer cell growth through the induction of apoptosis.

Non-Benzoquinone Geldanamycin Analog, WK-88-1, Induces Apoptosis in Human Breast Cancer Cell Lines

  • Zhao, Yu-Ru;Li, Hong-Mei;Zhu, Meilin;Li, Jing;Ma, Tao;Huo, Qiang;Hong, Young-Soo;Wu, Cheng-Zhu
    • Journal of Microbiology and Biotechnology
    • /
    • v.28 no.4
    • /
    • pp.542-550
    • /
    • 2018
  • Heat shock protein 90 (Hsp90) is treated as a molecular therapeutic target for the prevention and treatment of cancer. Geldanamycin (GA) was the first identified natural Hsp90 inhibitor, but hepatotoxicity has limited its clinical application. Nevertheless, a new GA analog (WK-88-1) with the non-benzoquinone skeleton, obtained from genetically engineered Streptomyces hygroscopicus, was found to have anticancer activity against two human breast cancer cell lines. WK-88-1 produced concentration-dependent inhibition of cell proliferation, cell cycle arrest, and apoptosis in estrogen receptor (ER)-positive MCF-7 and ER-negative MDA-MB-231 cell lines. Detailed analysis showed that WK-88-1 downregulated some key cell cycle molecules (CDK1 and cyclin B1) and lead to $G_2/M$ cell cycle arrest. Further studies also showed that WK-88-1 could induce human breast cancer cell apoptosis by downregulating Hsp90 client proteins (Akt, p-Akt, IKK, c-Raf, and Bcl-2), decreasing the ATP level, increasing reactive oxygen species production, and lowering the mitochondrial membrane potential. Meanwhile, we discovered that WK-88-1 significantly decreased the levels of Her-2 and $ER-{\alpha}$ in MCF-7 cells but not in MDA-MB-231 cells. In addition, WK-88-1 significantly increased caspase-3, -8, and -9 activities and the cleavage of PARP in a concentration-dependent manner (with the exception of caspase-3 and PARP in MCF-7 cells). Taken together, our preliminary results suggest that WK-88-1 has the potential to play a role in breast cancer therapy.

Cytotoxic Effect of Bee (A. mellifera) Venom on Cancer Cell Lines

  • Borojeni, Sima Khalilifard;Zolfagharian, Hossein;Babaie, Mahdi;Javadi, Iraj
    • Journal of Pharmacopuncture
    • /
    • v.23 no.4
    • /
    • pp.212-219
    • /
    • 2020
  • Objectives: Nowadays cancer treatment is an important challenge in the medical world that needs better therapies. Many active secretions produced by insects such as honey bees used to discover new anticancer drugs. Bee venom (BV) has a potent anti inflammatory, anti cancer and tumor effects. The aim of present study is evaluation of anticancer effects induced by Apis mellifera venom (AmV) on cell Lines. Methods: AmV was selected for study on cancer cell lines. Total protein, molecular weight and LD50 of crude venom were determined. Then, cells were grown in Dulbecco's Modified Eagle medium supplemented with 10% fetal bovine serum and 1% antibiotics. The A549, HeLa and MDA-MB-231 cell Lines were exposed by different concentration of AmV. The morphology of cells was determined and cell viability was studed by MTT assay. Evaluation of cell death was determined by and DNA fragmentation. Results: The results from MTT assay showed that 3.125 ㎍/mL of A549, 12.5 for HeLa and 6.25 ㎍/mL of MDA-MB-231 killed 50% of cells (p < 0.05). Morphological analysis and the results from hoescht staining and DNA fragmentation indicated that cell death induced by AmV was significantly apoptosis. Conclusion: The data showed that using lower dosage of AmV during treatment period cause inhibition of proliferation in time and dose dependant manner. Findings indicated that some ingredients of AmV have anticancer effects and with further investigation it can be used in production of anticancer drugs.

Differential antiangiogenic and anticancer activities of the active metabolites of ginsenoside Rg3

  • Maryam Nakhjavani;Eric Smith;Kenny Yeo;Yoko Tomita;Timothy J. Price;Andrea Yool;Amanda R. Townsend;Jennifer E. Hardingham
    • Journal of Ginseng Research
    • /
    • v.48 no.2
    • /
    • pp.171-180
    • /
    • 2024
  • Background: Epimers of ginsenoside Rg3 (Rg3) have a low bioavailability and are prone to deglycosylation, which produces epimers of ginsenoside Rh2 (S-Rh2 and R-Rh2) and protopanaxadiol (S-PPD and R-PPD). The aim of this study was to compare the efficacy and potency of these molecules as anti-cancer agents. Methods: Crystal violet staining was used to study the anti-proliferatory action of the molecules on a human epithelial breast cancer cell line, MDA-MB-231, and human umbilical vein endothelial cells (HUVEC) and compare their potency. Cell death and cell cycle were studied using flow cytometry and mode of cell death was studied using live cell imaging. Anti-angiogenic effects of the drug were studied using loop formation assay. Molecular docking showed the interaction of these molecules with vascular endothelial growth factor receptor-2 (VEGFR2) and aquaporin (AQP) water channels. VEGF bioassay was used to study the interaction of Rh2 with VEGFR2, in vitro. Results: HUVEC was the more sensitive cell line to the anti-proliferative effects of S-Rh2, S-PPD and R-PPD. The molecules induced necroptosis/necrosis in MDA-MB-231 and apoptosis in HUVEC. S-Rh2 was the most potent inhibitor of loop formation. In silico molecular docking predicted a good binding score between Rh2 or PPD and the ATP-binding pocket of VEGFR2. VEGF bioassay showed that Rh2 was an allosteric modulator of VEGFR2. In addition, SRh2 and PPD had good binding scores with AQP1 and AQP5, both of which play roles in cell migration and proliferation. Conclusion: The combination of these molecules might be responsible for the anti-cancer effects observed by Rg3.

Screening of Cytotoxicity and Antimicrobial Effects of Hexane Extracts from Cornis fructus (산수유 헥산 추출물의 항균효과 및 세포독성)

  • Chun Hyun Ja;Choi Won Hyung;Lee Jeong Ho;Yang Hyun Ok;Baek Seung Hwa
    • Journal of Physiology & Pathology in Korean Medicine
    • /
    • v.17 no.2
    • /
    • pp.476-480
    • /
    • 2003
  • Cornis fructus was extracted by successive extraction and then fractionated with hexane extract to get active fractions. This study was performed to determine the cytotoxic effect of hexane extract from Cornis fructus on NIH 3T3 fibroblasts and cancer cell lines using MTT assay. Hexane extract showed cytotoxic effect against A549, B16 melanoma and MDA-MB-231. Futher fractionation with hexane extract was performed to obtain effective fraction, fraction 3 showed the cytotoxic effect against A549 and MDA-MB-231. In antimicrobial test of each fraction of hexane extract, fraction 5 showed antimicrobial activities against P. putida and P. aeruginosa.

Antiestrogen Interaction with Estrogen Receptors and Additional Antiestrogen Binding sites in Human Breast Cancer MCF-7 Cells

  • Ahn, Mee-Ryung;Sheen, Yhun-Yhong
    • Archives of Pharmacal Research
    • /
    • v.20 no.6
    • /
    • pp.579-585
    • /
    • 1997
  • To gain further insight into the mechanism of action of antiestrogens, we examined the interaction of antiestrogen with the estrogen receptor system and with estrogen- noncompetable antiestrogen binding sites. In addition to binding directly to the estrogen receptor, antiestrogens can be found associated with binding sites that are distinct from the estrogen receptor. In contrast to the restriction of estrogen receptors to estrogen target cells, such as those of uterus and mammary glands, antiestrogen binding sites are present in equal amounts in estrogen receptor-positive and -negative human breast cancer cell lines, such as MCF-7, T47D, and MDA-MB-231 that differ markedly in their sensitivity to antiestrogens. In order to gain greater insight into the role of these antiestrogen binding sites in the action of antiestrogens, we have examined the biopotency of different antiestrogens for the antiestrogen binding sites and that is CI628 > tamoxifen > trans-hydroxy tamoxifen > CI628M > H1285 > LY117018. This order of affinities does not parallel the affinity of these compounds for the estrogen receptor nor the potency of these compounds as antiestrogens. Indeed, compounds with high affinity for the estrogen receptor and greatest antiestrogenic potency have low affinities for these antiestrogen binding sites. Antiestrogenic potency correlates best with estrogen receptor affinity and not with affinity for antiestrogen binding sites. In summary, our findings suggested that interaction with the estrogen receptor is most likely the mechanism through which antiestrogens evoke their growth inhibitory effects.

  • PDF

Anti-cancer Activity of the Leave Extracts of Rodgersia podophylla through β-catenin Proteasomal Degradation in Human Cancer Cells

  • Kim, Jeong Dong;Park, Su Bin;Kim, Ha Na;Jeong, Jin Boo
    • Proceedings of the Plant Resources Society of Korea Conference
    • /
    • 2019.10a
    • /
    • pp.68-68
    • /
    • 2019
  • In this study, we evaluated the effect of Rodgersia podophylla leave extracts (RPL) on ${\beta}$-catenin level in human cancer cells. RPL dose-dependently inhibited cell proliferation in SW480, A549, MDA-MB-231, PC-3 and AsPC-1 cells. RPL dramatically decreased ${\beta}$-catenin protein level in all cancer cells. However, decreased level of ${\beta}$-catenin mRNA expression was observed in A549 and AsPC-1 cells. In addition, RPL dramatically attenuated cyclin D1 mRNA expression in all cancer cells. MG132 decreased the downregulation of ${\beta}$-catenin protein level induced by RPL in all cancer cells, while RPL-induced downregulation of ${\beta}$-catenin was inhibited by the inhibition of $GSK-3{\beta}$ by LiCl in MDA-MB-231 cells. RPL phosphorylated ${\beta}$-catenin and $GSK-3{\beta}$. In addition, the inhibition of $GSK-3{\beta}$ by LiCl attenuated RPL-induced ${\beta}$-catenin phosphorylation. Based on these findings, RPL may be a potential candidate for the development of chemopreventive or therapeutic agents for human cancer.

  • PDF

6,8-Dihydroxy-7-methoxy-1-methyl-azafluorenone Induces Caspase-8- and -9-mediated Apoptosis in Human Cancer Cells

  • Banjerdpongchai, Ratana;Khaw-on, Patompong;Ristee, Chantrarat;Pompimon, Wilart
    • Asian Pacific Journal of Cancer Prevention
    • /
    • v.14 no.4
    • /
    • pp.2637-2641
    • /
    • 2013
  • 6,8-Dihydroxy-7-methoxy-1-methyl-azafluorenone (DMMA), a purified compound from Polyalthia cerasoides roots, is cytotoxic to various cancer cell lines. The aims of this study were to demonstrate the type of cancer cell death and the mechanism(s) involved. DMMA inhibited cell growth and induced apoptotic death in human leukemic cells (HL-60, U937, MOLT-4), human breast cancer MDA-MB231 cells and human hepatocellular carcinoma HepG2 cells in a dose dependent manner, with $IC_{50}$ values ranging between 20-55 ${\mu}M$. DMMA also decreased cell viability of human peripheral blood mononuclear cells. The morphology of cancer cells induced by the compound after staining with propidium iodide and examined under a fluorescence microscope was condensed nuclei and apoptotic bodies. Mitochondrial transmembrane potential (MTP) was decreased after 24h exposure in all five types of cancer cells. DMMA-induced caspase-3, -8, and -9 activity was strongly induced in human leukemic HL-60 and MOLT-4 cells, while in U937-, MDA-MB231- and HepG2-treated cells there was partial induction of caspase. In conclusion, DMMA-induced activation of caspase-8 and -9 resulted in execution of apoptotic cell death in human leukemic HL-60 and MOLT-4 cell lines via extrinsic and intrinsic pathways.

Antiestrogen, Trans-Tamoxifen Modulation of Human Breast Cancer Cell Growth

  • Lee, Hyung-Ok;Sheen, Yhun-Yhong
    • Archives of Pharmacal Research
    • /
    • v.20 no.6
    • /
    • pp.572-578
    • /
    • 1997
  • To gain further insight into how antiestrogens modulate cell function, the effects of antiestrogen on cell proliferation were studied in human breast cancer cells. We examined the effects of trans-tamoxifen on the proliferation of three human breast cancer cell lines that differed in their estrogen receptor contents. Trans-tamoxifen $(1{\mu}M)$ markedly inhibited the estrogen stimulated proliferation of MCF-7 human breast cancer cells that contained high levels of estrogen receptor $(1.15{\pm}0.03 pmole/mg protein)$ over that of control. In T47D cells that contained low levels of estrogen receptor $(0.23{\pm}0.05 pmole/mg protein)$, trans-tamoxifen $(1{\mu}M)$ showed minimal inhibition of estrogen stimulated cell proliferation over that of control. MDA-MB-231 cells, that contained no detectable levels of estrogen receptors, had their growth unaffected by trans-tamoxifen treatment. These results showed their sensitivity to growth inhibition by antiestrogen conrrelated well with their estrogen receptor content. Also we examined the effect of antiestrogen on cellular progestrone receptor level as well as plasminogen activator activity in MCF-7 cells. Trans-tamoxifen $(1{\mu}M)$ showed maximal inhibition of estrogen stimulated progestrone receptor level as well as plasminogen activator activity in MCF-7 cells that were stimulated by estrogen. It is not clear whether these inhibitions of progestrone receptor and plasminogen activator activity by estrogen are related to the antiestrogen inhibition of cell proliferation of MCF-7 cells. From the results of this study, it is clearly demonstrated that trans-tamoxifen is an antiestrogen in MCF-7 human breast cancer cells. Our data suggest that the biological effectiveness of trans-tamoxifen appear to result from its affinity of interaction with the estrogen receptor.

  • PDF

shRNA Mediated RHOXF1 Silencing Influences Expression of BCL2 but not CASP8 in MCF-7 and MDA-MB-231 Cell Lines

  • Ghafouri-Fard, Soudeh;Abdollahi, Davood Zare;Omrani, Mirdavood;Azizi, Faezeh
    • Asian Pacific Journal of Cancer Prevention
    • /
    • v.13 no.11
    • /
    • pp.5865-5869
    • /
    • 2012
  • RHOXF1 has been shown to be expressed in embryonic stem cells, adult germline stem cells and some cancer lines. It has been proposed as a candidate gene to encode transcription factors regulating downstream genes in the human testis with antiapoptotic effects. Its expression in cancer cell lines has implied a similar role in the process of tumorigenesis. The human breast cancer cell lines MDA-MB-231 and MCF-7 were cultured in DMEM medium and transfected with a pGFP-V-RS plasmid bearing an RHOXF1 specific shRNA. Quantitative real-time RT-PCR was performed for RHOXF1, CASP8, BCL2 and HPRT genes. Decreased RHOXF1 expression was confirmed in cells after transfection. shRNA knock down of RHOXF1 resulted in significantly decreased BCL2 expression in both cell lines but no change in CASP8 expression. shRNA targeting RHOXF1 was shown to specifically mediate RHOXF1 gene silencing, so RHOXF1 can mediate transcriptional activation of the BCL2 in cancers and may render tumor cells resistant to apoptotic cell death induced by anticancer therapy. shRNA mediated knock down of RHOXF1 can be effective in induction of apoptotic pathway in cancer cells via BCL2 downregulation, so it can have potential therapeutic utility for human breast cancer.