• Title/Summary/Keyword: Antitumor Effect

검색결과 586건 처리시간 0.037초

마우스 간암에서 항암제-방사선 복합요법을 이용한 치료 효과 향상 (Enhancement of Tumor Radioresponse by Combined Chemotherapy in Murine Mepatocarcinorna)

  • 성진실;김성희;서창옥
    • Radiation Oncology Journal
    • /
    • 제18권4호
    • /
    • pp.329-336
    • /
    • 2000
  • 목적 : 마우스 간암에서 방사선과 각종 항암제와의 복합요법을 시행하여, 방사선 감수성을 증가시킬 수 있는 약물을 탐색하고자 하였다. 방법 : C3H/HeJ마우스에 마우스 간암인 HCa-1을 이식하고, 평균 직경 8 mm에 이르렀을 때, 방사선 조사(25 Gy), 항암 약물(5-Fu, 150 mg/kg; adriamycln, 8 mg/kg; paclltaxel, 40 mg/kg; gemcltablne, 50 mg/kg), 또는 방사선과 항암 약물의 복합 치료를 시행하였다 치료에 대한 종양 반응은 종양 성장 지연과 항진 요인으로 분석하였다. 항진 효과를 보인 약물에 대하여 그 기전 연구는 조직 절편에서 apoptotic 수준을 평가하고, 또한 조절물질의 발현을 분석하였다. p53, Bcl-2, Bax, Bcl-XL, Bcl-XS, p21$^{WAF1/CIP1}$의 발현 분석은 westeblotting으로 하였다. 결과 : Gemcltabine 만이 방사선 감수성을 증가시키는 것으로 나타났다(항진요인:1.6). Gemcltabine과 방사선의 복합 치료는 apoptosis 유도에서는 부가적 수준만을 보였다. 조절울질의 발현 양상은 방사선 단독에 비하여 방사선과 gemcitabine의 병용시 p21$^{WAF1/CIP1}$의 증가가 유의하게 관찰되었다. 결론 : Gemcitabiue은 마우스 간암에서 방사선 감수성을 증가시키는 것으로 나타났다. 이를 조절하는 요소로서 p21$^{WAF1/CIP1}$ 이 관여할 것으로 생각 된다.

  • PDF

Siamese Crocodile White Blood Cell Extract Inhibits Cell Proliferation and Promotes Autophagy in Multiple Cancer Cell Lines

  • Phosri, Santi;Jangpromma, Nisachon;Chang, Leng Chee;Tan, Ghee T.;Wongwiwatthananukit, Supakit;Maijaroen, Surachai;Anwised, Preeyanan;Payoungkiattikun, Wisarut;Klaynongsruang, Sompong
    • Journal of Microbiology and Biotechnology
    • /
    • 제28권6호
    • /
    • pp.1007-1021
    • /
    • 2018
  • Cancer represents one of the most significant threats to human health on a global scale. Hence, the development of effective cancer prevention strategies, as well as the discovery of novel therapeutic agents against cancer, is urgently required. In light of this challenge, this research aimed to evaluate the effects of several potent bioactive peptides and proteins contained in crocodile white blood cell extract (cWBC) against LU-1, LNCaP, PC-3, MCF-7, and CaCo-2 cancer cell lines. The results demonstrate that 25, 50, 100, and $200{\mu}g/ml$ cWBC exhibits a strong cytotoxic effect against all investigated cell lines ($IC_{50}$ $70.34-101.0{\mu}g/ml$), while showing no signs of cytotoxicity towards noncancerous Vero and HaCaT cells. Specifically, cWBC treatment caused a significant reduction in the cancerous cells' colony forming ability. A remarkable suppression of cancerous cell migration was observed after treatment with cWBC, indicating potent antimetastatic properties. The mechanism involved in the cancer cell cytotoxicity of cWBC may be related to apoptosis induction, as evidenced by typical apoptotic morphology features. Moreover, certain cWBC concentrations induced significant overproduction of ROS and significantly inhibited the $S-G_2/M$ transition in the cancer cell. The molecular mechanisms of cWBC in apoptosis induction were to decrease Bcl-2 and XIAP expression levels and increase the expression levels of caspase-3, caspase-8, and p53. These led to a decrease in the expression level of the cell cycle-associated gene cyclin-B1 and the arrest of cell population growth. Consequently, these findings demonstrate the prospect of the use of cWBC for cancer therapy.

The Antitumor Effects of Selenium Compound $Na_5SeV_5O_{18}{\cdot}3H_2O$ in K562 Cell

  • Yang, Jun-Ying;Wang, Zi-Ren
    • Archives of Pharmacal Research
    • /
    • 제29권10호
    • /
    • pp.859-865
    • /
    • 2006
  • With an approach to study the anti-tumor effects and mechanism of selenium compound, we investigated the anti-tumor activity and mechanism of $Na_5SeV_5O_{18}{\cdot}3H_2O$ (NaSeVO) in K562 cells. The results showed that $0.625{\sim}20\;mg/L$ NaSeVO could significantly inhibit the proliferation of K562 cells in vitro in a time- and concentration-dependent manner as determined by microculture tetrazolium (MTT) assay, the IC50 values were 14.41 (4.45-46.60) and 3.45 (2.29-5.22) mg/L after 48 hand 72 h treatment with NaSeVO respectively. In vivo experiments demonstrated that i.p. administration of 5, 10 mg/kg NaSeVO exhibited an significant inhibitory effect on the growth of transplantation tumor sarcoma 180 (S180) and hepatoma 22 (H22) in mice, with inhibition rate 26.8% and 58.4% on S180 and 31.3% and 47.4% on H22, respectively. Cell cycle studies indicated that the proportion of G0/G1 phase was increased at 2.5 mg/L while decreased at 10 mg/L after treatment for 24, 48 h. Whereas S phase was decreased at 2.5-5 mg/L and markedly increased at 10 mg/L after treatment for 48 h. After treatment for 24 h, 10 mg/L NaSeVO also markedly increased S and G2/M phases. Take together, the result clearly showed that NaSeVO markedly increased S and G2/M phases at 10 mg/L. The study of immunocytochemistry showed that the expression bcl-2 is significantly inhibited by 10 mg/L NaSeVO, and bax increased. Morphology observation also revealed typical apoptotic features. NaSeVO also significantly caused the accumulation of $Ca^{2+}$ and $Mg^{2+}$, reactive oxygen species (ROS) and the reduction of pH value and mitochondrial membrane potential in K562 cells as compared with control by confocal laser scanning microscope. These results suggest that NaSeVO has anti-tumor effects and its mechanism is attributed partially to apoptosis induced by the elevation of intracellular $Ca^{2+}$, $Mg^{2+}$ and ROS concentration, and a reduction of pH value and mitochondria membrane potential (MMP).

인간 말초혈액 단핵구 유래 수지상세포의 면역반응에 미치는 Gefitinib의 영향 (The Effect of Gefitinib on Immune Response of Human Peripheral Blood Monocyte-Derived Dendritic Cells)

  • 조진훈;김미현;이광하;김기욱;전두수;박혜경;김윤성;이민기;박순규
    • Tuberculosis and Respiratory Diseases
    • /
    • 제69권6호
    • /
    • pp.456-464
    • /
    • 2010
  • Background: Synergistic antitumor effects of the combined chemoimmunotherapy based on dendritic cells have been reported recently. The aim of this study is to search new applicability of gefitinib into the combination treatment through the confirmation of gefitinib effects on the monocyte derived dendritic cells (moDCs); most potent antigen presenting cell (APC). Methods: Immature and mature monocyte-derived dendritic cell (im, mMoDC)s were generated from peripheral blood monocyte (PBMC) in Opti-MEM culture medium supplemented with IL-4, GM-CSF and cocktail, consisting of TNF-${\alpha}$ (10 ng/mL), IL-$1{\beta}$ (10 ng/mL), IL-6 (1,000 U/mL) and $PGE_2$ ($1{\mu}/mL$). Various concentrations of gefitinib also added on day 6 to see the influence on immature and mature MoDCs. Immunophenotyping of DCs under the gefitinib was performed by using monoclonal antibodies (CD14, CD80, CD83, CD86, HLA-ABC, HLA-DR). Supernatant IL-12 production and apoptosis of DCs was evaluated. And MLR assay with $[^3H]$-thymidine uptake assay was done. Results: Expression of CD83, MHC I were decreased in mMoDCs and MHC I was decreased in imMoDCs under gefitinib. IL-12 production from mMoDCs was decreased under $10{\mu}M$ of gefitinib sinificantly. Differences of T cell proliferation capacity were not observed in each concentration of geftinib. Conclusion: In spite of decreased expressions of some dendritic cell surface molecules and IL-12 production under $10{\mu}M$ of gefitinib, significant negative influences of gefitinib in antigen presenting capacity and T cell stimulation were not observed.

A Case of Ischemic Colitis Associated with Paclitaxel Loaded Polymeric Micelle ($Genexol-PM^{(R)}$) Chemotherapy

  • Park, Choel-Kyu;Kang, Hyun-Wook;Kim, Tae-Ok;Ki, Ho-Seok;Kim, Eun-Young;Ban, Hee-Jung;Yoon, Byeong-Kab;Oh, In-Jae;Choi, Yoo-Deok;Kwon, Yong-Soo;Kim, Yoo-Il;Lim, Sung-Chul;Kim, Young-Chul;Kim, Kyu-Sik
    • Tuberculosis and Respiratory Diseases
    • /
    • 제69권2호
    • /
    • pp.115-118
    • /
    • 2010
  • Paclitaxel has been widely used for treating many solid tumors. Although colonic toxicity is an unusual complication of paclitaxel-based chemotherapy, the reported toxicities include pseudomembranous colitis, neutropenic enterocolitis and on rare occasions ischemic colitis. $Genexol-PM^{(R)}$, which is a recently developed cremophor-free, polymeric micelle-formulated paclitaxel, has shown a more potent antitumor effect because it can increase the usual dose of paclitaxel due to that $Genexol-PM^{(R)}$ does not include the toxic cremophor compound. We report here on a case of a 57-year-old man with advanced non-small cell lung cancer and who developed ischemic colitis after chemotherapy with $Genexol-PM^{(R)}$ and cisplatin. He complained of hematochezia with abdominal pain on the left lower quadrant. Colonoscopy revealed diffuse mucosal hemorrhage and edema from the sigmoid colon to the splenic flexure. After bowel rest, he recovered from his symptoms and the follow-up colonoscopic findings showed that the mucosa was healing. Since then, he was treated with pemetrexed monotherapy instead of a paclitaxel compound and platinum.

Helixor A는 시험관 내에서 thrombospondin-1의 상승조절을 통해 신혈관생성을 억제한다. (Helixor A Inhibits Angiogenesis in vitro Via Upregutation of Thrombospondin-1)

  • 염동훈;홍경자
    • 생명과학회지
    • /
    • 제15권6호
    • /
    • pp.895-903
    • /
    • 2005
  • Thrombospondin-1은 암성장과 신혈관생성 억제조절인자로 병리학적 조건과 세포외에서의 자극에 의해 세포 특이적으로 조절되어지고 이 TSP-1 유전자 발현조절은 암치료제 개발을 위한 새로운 접근으로 중요하다. Mistletoe는 기생식물로 면역조절과 항암제로 사용되어지고 있다. Helixor A는 mistletoe 추출물의 수용액부분이다. 여기서 우리는 Helixor A를 투여하면 간암세포주 (Hep3B)와 혈관내피세포주 (BAE)에서 TSP-1의 mRWA와 단백질 발현이 유도되는 것을 관찰하였다. TSP-1 promoter 활성분석으로 TSP-1유전자의 발현이 Helixor A에 의해 전사단계에서 조절되어 진다는 것을 확인하였다. 세포 침윤 분석에서 Helixor A를 처리한 배지를 두 세포주에 처리함으로 침윤된 세포의 수가 현저하게 줄어드는 것을 관찰하였고, Matrigel에서 혈관 내피 세포 (BAE)의 모세관 형성을 억제하는 것을 관찰하였다. 또한 세포 침윤과 모세관 형성에서 Helixor A를 처리했던 배지의 억제 효과가 TSP-1 중화 항체에 의해 그 효과가 상실되었다. 그러므로, 이 결과는 TSP-1이 Helixor A에 의해 유도되어진 혈관신생 억제 효과에 연관이 있음을 제시하였다. 이러한 결과를 종합 하였을 때 Helixor A가 TSP-1의 상승조절을 통해 혈관신생 억제 효과를 가질 것이라고 생각된다.

Cyclophosphamide가 마우스의 면역기억에 미치는 영향 (Effects of Cyclophosphamide on Immunological Memory in Mice)

  • 박영민;박윤규;안우섭;하대유
    • 대한미생물학회지
    • /
    • 제22권2호
    • /
    • pp.175-184
    • /
    • 1987
  • The use of alkylating agent cyclophosphamide(CY), a widely used antitumor drug is well known as a potent immunosuppressant and has been used as a probe for investigating the functional capabilities of lymphocyte subsets of both T and B cells that play an important role in the regulation of the immune response. The present study was undertaken in an effort to assess the effects of CY on immunological memory in murine model. CY, given as a single dose of CY(250mg/kg) before sensitization with sheep red blood cells(SRBC) enhanced the primary response of Arthus and delayed-type hypersensitivity(DTH), as measured by footpad swelling reaction, but suppressed their tertiary DTH response. The similar CY pretreatment enhanced both the primary and tertiary hemagglutinin(HA) responses to SRBC, and the tertiary antibody response against polyvinylpyrroridone(PVP), a thymus-independent antigen but not the primary response against PVP. CY, given as a single dose of 250mg/kg 2 days before the primary immunization and two doses of 100mg/kg 2 days before the secondary and tertiary immunization, markedly suppressed the tertiary DTH and HA responses to SRBC. However, CY, given as small multiple daily doses(10mg/kg) over 4 days before sensitization but not after sensitization, enhanced the secondary HA response to SRBC. Contact sensitivity to dinitrofluorobenzene(DNFB) was suppressed by the drug, given either as a single large dose(300mg/kg) or as multiple dose(10mg/kg) administered 2 days before, together with or after DNFB sensitization. This suppression was more pronounced and more significant when CY was given as multiple dose. However, the enhancement of the secondary contact sensitivity to DNFB by CY was not clear-cut. The splenectomy appears to increase the enhancing effect of CY on contact sensitivity. These results suggest that CY selectively influences the immune response depending on the time of the drug administration relative to immunization and that the secondary or tertiary immune response involve memory cells with different susceptibilities to CY. Moreover, these results suggest that multiple low doses may sesectivley inhibit suppressor T cell proliferation involving DTH, HA or contact sensitivity without effecting helper T cells, but high doses presumably inhibit helper T cells and suppressor T cells with effecting B cells.

  • PDF

인체폐암 A549 세포에서 Baicalein에 의한 세포사멸 유도: Apoptosis와 Autophagy 경로의 역할 (Baicalein induces cell death in Human Lung Carcinoma A549 Cells: Role of Apoptosis and Autophagy pathway)

  • 김철환;황병수;정용태;김민진;신수영;오영택;엄정혜;이승영;최경민;조표연;정진우
    • 한국자원식물학회:학술대회논문집
    • /
    • 한국자원식물학회 2019년도 춘계학술대회
    • /
    • pp.112-112
    • /
    • 2019
  • Baicalein is one of the main flavonoids derived from roots of Scutellaria baicalensis Georgi, a traditional Oriental medicine. Although baicalein has high antitumor effect on several human carcinomas, the mechanism responsible for this property is not unclear. In this study, the data revealed that baicale-ininduced growth inhibition was associated with the induction of apoptosis connecting with cytochrome c release, down-regulation of anti-apoptotic Bcl-xl and increased the percentage of cells with a loss of mitochondria membrane permeabilization. Baicalein also induced the proteolytic activation of caspases and cleavage of PARP; however, blockage of caspases activation by z-VAD-fmk inhibited baicalein-induced apoptosis. In addition, baicalein enhanced the formation of autophagosomes and up-regulated LC3-II/LC3-I ratio. Interestingly, the pretreatment of bafilomycin A1 recovered baicalein-induced cell death suggesting that autophagy by baicalein roles as protective autophagy. Taken together, our results indicated that this flavonoid induces apoptosis and cell protective autophagy. These data means combination treatment with baicalein and autophagy inhibitor might be a promising anticancer drug.

  • PDF

20(S)-ginsenoside Rh2 induces caspase-dependent promyelocytic leukemia-retinoic acid receptor A degradation in NB4 cells via Akt/Bax/caspase9 and TNF-α/caspase8 signaling cascades

  • Zhu, Sirui;Liu, Xiaoli;Xue, Mei;Li, Yu;Cai, Danhong;Wang, Shijun;Zhang, Liang
    • Journal of Ginseng Research
    • /
    • 제45권2호
    • /
    • pp.295-304
    • /
    • 2021
  • Background: Acute promyelocytic leukemia (APL) is a hematopoietic malignancy driven by promyelocytic leukemia-retinoic acid receptor A (PML-RARA) fusion gene. The therapeutic drugs currently used to treat APL have adverse effects. 20(S)-ginsenoside Rh2 (GRh2) is an anticancer medicine with high effectiveness and low toxicity. However, the underlying anticancer mechanisms of GRh2-induced PML-RARA degradation and apoptosis in human APL cell line (NB4 cells) remain unclear. Methods: Apoptosis-related indicators and PML-RARA expression were determined to investigate the effect of GRh2 on NB4 cells. Z-VAD-FMK, LY294002, and C 87, as inhibitors of caspase, and the phosphatidylinositol 3-kinase (PI3K) and tumor necrosis factor-α (TNF-α) pathways were used to clarify the relationship between GRh2-induced apoptosis and PML-RARA degradation. Results: GRh2 dose- and time-dependently decreased NB4 cell viability. GRh2-induced apoptosis, cell cycle arrest, and caspase3, caspase8, and caspase9 activation in NB4 cells after a 12-hour treatment. GRh2-induced apoptosis in NB4 cells was accompanied by massive production of reactive oxygen species, mitochondrial damage and upregulated Bax/Bcl-2 expression. GRh2 also induced PML/PML-RARA degradation, PML nuclear bodies formation, and activation of the downstream p53 pathway in NB4 cells. Z-VAD-FMK inhibited caspase activation and significantly reversed GRh2-induced apoptosis and PML-RARA degradation. GRh2 also upregulated TNF-α expression and inhibited Akt phosphorylation. LY294002, an inhibitor of the PI3K pathway, enhanced the antitumor effects of GRh2, and C 87, an inhibitor of the TNF-α pathway, reversed NB4 cell viability, and GRh2-mediated apoptosis in a caspase-8-dependent manner. Conclusion: GRh2 induced caspase-dependent PML-RARA degradation and apoptosis in NB4 cells via the Akt/Bax/caspase9 and TNF-α/caspase8 pathways.

P3H4 promotes renal cell carcinoma progression and suppresses antitumor immunity via regulating GDF15-MMP9-PD-L1 axis

  • Tian, Shuo;Huang, Yan;Lai, Dong;Wang, Hanfeng;Du, Songliang;Shen, Donglai;Chen, Weihao;Xuan, Yundong;Lu, Yongliang;Feng, Huayi;Zhang, Xiangyi;Zhao, Wenlei;Wang, Chenfeng;Wang, Tao;Wu, Shengpan;Huang, Qingbo;Niu, Shaoxi;Wang, Baojun;Ma, Xin;Zhang, Xu
    • Advances in nano research
    • /
    • 제12권6호
    • /
    • pp.639-652
    • /
    • 2022
  • The prolyl 3-hydroxylase family member 4 (P3H4), is associated with post-translational modification of fibrillar collagens and aberrantly activated in cancer leading to tumor progression. However, its role in clear cell renal cell carcinoma (ccRCC) is still unknown. Here we reported that P3H4 was highly expressed in renal cancer tissues and significantly positive correlated with poor prognosis. Knockdown of P3H4 inhibited the proliferation, migration and metastasis of renal cancer cells in vitro and in vivo, and also, overexpression of it enhanced the oncogenic process. Mechanistically, P3H4 depletion decreased the levels of GDF15-MMP9 axis and repressed its downstream signaling. Further functional studies revealed that inhibition of GDF15 suppressed renal cancer cell growth and GDF15 recombinant human protein (rhGDF15) supplementation effectively rescued the inhibitory effect induced by P3H4 knockdown. Moreover, decreased levels of MMP9 caused by inhibition of P3H4-GDF15 signaling constrained the expression of PD-L1 and suppression of P3H4 accordingly promoted anti-tumor immunity via stimulating the infiltration of CD4+ and CD8+ T cells in syngeneic mice model. Taken together, our findings firstly demonstrated that P3H4 promotes ccRCC progression by activating GDF15-MMP9-PD-L1 axis and targeting P3H4-GDF15-MMP9 signaling pathway can be a novel strategy of controlling ccRCC malignancy.