• 제목/요약/키워드: p27KIP1

검색결과 69건 처리시간 0.023초

마우스 피부암 발생과정에 있어서 2,3,7,8-Tetrachlorodibenzo-p­Dioxin (TCDD) 처리에 의한 유전자발현 변화 연구 (Effects of 2,3,7,8-Tetrachlorodibenzo-p-Dioxin (TCDD) on Gene Expression in Mouse Skin Carcinogenesis)

  • Ryeom Tai Kyung;Kim Ok Hee;Kong Mi Kyung;Park Mi Sun;Jee Seung Wan;Eom Mi Ok;Kang Ho Il
    • 한국환경성돌연변이발암원학회지
    • /
    • 제25권1호
    • /
    • pp.40-46
    • /
    • 2005
  • 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) displays high toxicity in animals and has been implicated in human carcinogenesis. Although the mechanism of carcinogenesis by TCDD is unclear, it is considered to be a non-genotoxic compound and tumor promoter. In our experiment, we investigated the effects of TCDD on gene expression in mouse skin carcinogenesis. We used cDNA microarray to detect the differential gene expression in tumors induced in hairless mouse skin by MNNG plus TCDD protocol. We found that erb-2, c-ets2 and p27$^{kip1}$ were significantly up-regulated, but TNFR2, AKT-l, integrin $\beta$l, maspin, IGF-l, c-raf-l, Rb were significantly down-regulated, in tumor region, respectively. We also found that the expression of 53 genes involved in cen cycle, signal transduction, apoptosis, adhesion molecule, angiogenesis, and invasion, were changed two fold more, in tumor surrounding region. These data suggest that TCDD alters the expression of a large array of genes involved in apoptosis, cytokine production and angiogenesis in mouse skin carcinogenesis.

  • PDF

Lysophosphatidic acid increases mesangial cell proliferation in models of diabetic nephropathy via Rac1/MAPK/KLF5 signaling

  • Kim, Donghee;Li, Hui Ying;Lee, Jong Han;Oh, Yoon Sin;Jun, Hee-Sook
    • Experimental and Molecular Medicine
    • /
    • 제51권2호
    • /
    • pp.9.1-9.10
    • /
    • 2019
  • Mesangial cell proliferation has been identified as a major factor contributing to glomerulosclerosis, which is a typical symptom of diabetic nephropathy (DN). Lysophosphatidic acid (LPA) levels are increased in the glomerulus of the kidney in diabetic mice. LPA is a critical regulator that induces mesangial cell proliferation; however, its effect and molecular mechanisms remain unknown. The proportion of ${\alpha}-SMA^+/PCNA^+$ cells was increased in the kidney cortex of db/db mice compared with control mice. Treatment with LPA concomitantly increased the proliferation of mouse mesangial cells (SV40 MES13) and the expression of cyclin D1 and CDK4. On the other hand, the expression of $p27^{Kip1}$ was decreased. The expression of $Kr{\ddot{u}}ppel$-like factor 5 (KLF5) was upregulated in the kidney cortex of db/db mice and LPA-treated SV40 MES13 cells. RNAi-mediated silencing of KLF5 reversed these effects and inhibited the proliferation of LPA-treated cells. Mitogen-activated protein kinases (MAPKs) were activated, and the expression of early growth response 1 (Egr1) was subsequently increased in LPA-treated SV40 MES13 cells and the kidney cortex of db/db mice. Moreover, LPA significantly increased the activity of the Ras-related C3 botulinum toxin substrate (Rac1) GTPase in SV40 MES13 cells, and the dominant-negative form of Rac1 partially inhibited the phosphorylation of p38 and upregulation of Egr1 and KLF5 induced by LPA. LPA-induced hyperproliferation was attenuated by the inhibition of Rac1 activity. Based on these results, the Rac1/MAPK/KLF5 signaling pathway was one of the mechanisms by which LPA induced mesangial cell proliferation in DN models.

오령산에 의한 고포도당 유도 사구체간질세포 이상증식 개선효과 (Oryeong-san Ameliorates High Glucose-induced Mesangial Cell Proliferation)

  • 윤정주;이윤정;이소민;김대환;이호섭;강대길
    • 대한한의학방제학회지
    • /
    • 제21권2호
    • /
    • pp.53-62
    • /
    • 2013
  • Objectives : Diabetic nephropathy is associated with morbidity and mortality of diabetes mellitus patients. Mesangial cell proliferation is known as the major pathologic features such as glomerulosclerosis. Oryeong-san, Korean formula, is widely used for the treatment of nephrosis, edema, and uremia. Oryeong-san is composed of five herbs: Alismatis Rhizoma, Polyporus, Atractylodis Rhizoma Alba, Hoelen, and Cinnamomi Cortex. Methods : The present study was performed to investigate potent inhibitory effect of Oryeong-san on high glucose (HG)-induced rat mesangial cells (RMC) proliferation. Results : RMC proliferation under 25 mM glucose was significantly accelerated compared with 5.5 mM glucose, which was inhibited by Oryeong-san in dose dependent manner. Pre-treatment of Oryeong-san induced down-regulation of cyclins/CDKs and up-regulation of CDK inhibitor, p21waf1/cip1 and p27kip1 expression. In addition, Oryeong-san reduced HG-induced RMC proliferation by suppressed the mitogen-activated protein kinase (MAPK) phospholyration such as extracellular signal regulated kinase (ERK), Jun N-terminal Kinase (JNK), and p38. Oryeong-san significantly suppressed HG-induced ROS production. Conclusions : Oryeong-san consequently inhibited HG-induced mesangial cell proliferation through the inhibition of MAPK and ROS signaling pathway. These results suggest that Oryeong-san may be effective in the treatment of renal dysfunction leading to diabetic nephropathy.

Panduratin A Inhibits Cell Proliferation by Inducing G0/G1 Phase Cell Cycle Arrest and Induces Apoptosis in Breast Cancer Cells

  • Liu, Qiuming;Cao, Yali;Zhou, Ping;Gui, Shimin;Wu, Xiaobo;Xia, Yong;Tu, Jianhong
    • Biomolecules & Therapeutics
    • /
    • 제26권3호
    • /
    • pp.328-334
    • /
    • 2018
  • Because of the unsatisfactory treatment options for breast cancer (BC), there is a need to develop novel therapeutic approaches for this malignancy. One such strategy is chemotherapy using non-toxic dietary substances and botanical products. Studies have shown that Panduratin A (PA) possesses many health benefits, including anti-inflammatory, anti-bacterial, anti-oxidant and anticancer activities. In the present study, we provide evidence that PA treatment of MCF-7 BC cells resulted in a time- and dose-dependent inhibition of cell growth with an $IC_{50}$ of $15{\mu}M$ and no to little effect on normal human MCF-10A breast cells. To define the mechanism of these anti-proliferative effects of PA, we determined its effect critical molecular events known to regulate the cell cycle and apoptotic machinery. Immunofluorescence and flow cytometric analysis of Annexin V-FITC staining provided evidence for the induction of apoptosis. PA treatment of BC cells resulted in increased activity/expression of mitochondrial cytochrome C, caspases 7, 8 and 9 with a significant increase in the Bax:Bcl-2 ratio, suggesting the involvement of a mitochondrial-dependent apoptotic pathway. Furthermore, cell cycle analysis using flow cytometry showed that PA treatment of cells resulted in G0/G1 arrest in a dose-dependent manner. Immunoblot analysis data revealed that, in MCF-7 cell lines, PA treatment resulted in the dose-dependent (i) induction of $p21^{WAF1/Cip1}$ and p27Kip1, (ii) downregulation of Cyclin dependent kinase (CDK) 4 and (iii) decrease in cyclin D1. These findings suggest that PA may be an effective therapeutic agent against BC.

Anti-Cancer Effect of IN-2001 in T47D Human Breast Cancer

  • Joung, Ki-Eun;Min, Kyung-Nan;Kim, Dae-Kee;Sheen, Yhun-Yhong
    • Biomolecules & Therapeutics
    • /
    • 제20권1호
    • /
    • pp.81-88
    • /
    • 2012
  • Histone deacetylases (HDACs) are enzymes involved in the remodelling of chromatin, and have a key role in the epigenetic regulation of gene expression. Histone deacetylase (HDAC) inhibitors are emerging as an exciting new class of potential anti-cancer agents. In recent years, a number of structurally diverse HDAC inhibitors have been identifi ed and these HDAC inhibitors induce growth arrest, differentiation and/or apoptosis of cancer cells in vitro and in vivo. However, the underlying molecular mechanisms remain unclear. This study aimed at investigating the anti-tumor activity of various HDAC inhibitors, IN-2001, using T47D human breast cancer cells. Moreover, the possible mechanism by which HDAC inhibitors exhibit anti-tumor activity was also explored. In estrogen receptor positive T47D cells, IN-2001, HDAC inhibitor showed anti-proliferative effects in dose-and time-dependent manner. In T47D human breast cancer cells showed anti-tumor activity of IN-2001 and the growth inhibitory effects of IN-2001 were related to the cell cycle arrest and induction of apoptosis. Flow cytometry studies revealed that IN-2001 showed accumulation of cells at $G_2$/M phase. At the same time, IN-2001 treatment time-dependently increased sub-$G_1$ population, representing apoptotic cells. IN-2001-mediated cell cycle arrest was associated with induction of cdk inhibitor expression. In T47D cells, IN-2001 as well as other HDAC inhibitors treatment significantly increased $p21^{WAF1}$ and $p27^{KIP1}$ expression. In addition, thymidylate synthase, an essential enzyme for DNA replication and repair, was down-regulated by IN-2001 and other HDAC inhibitors in the T47D human breast cancer cells. In summary, IN-2001 with a higher potency than other HDAC inhibitors induced growth inhibition, cell cycle arrest, and eventual apoptosis in human breast cancer possibly through modulation of cell cycle and apoptosis regulatory proteins, such as cdk inhibitors, cyclins, and thymidylate synthase.

사람혀편평상피세포암종세포에서 proteasome 억제제인 lactacystin에 의해 유도된 세포자멸사의 기전에 대한 연구 (Mechanism Underlying a Proteasome Inhibitor, Lactacystin-Induced Apoptosis on SCC25 Human Tongue Squamous Cell Carcinoma Cells)

  • 백철중;김규천;김인령;이승은;곽현호;박봉수;태일호;고명연;안용우
    • Journal of Oral Medicine and Pain
    • /
    • 제34권3호
    • /
    • pp.261-276
    • /
    • 2009
  • Sreptomyces라는 세균에서 추출한 lactacystin은 선택적인 proteasome 억제제로서 많은 연구에서 사용되어져 왔다. Proteasome 억제제는 최근의 많은 연구를 통해서 암세포증식의 억제에 대한 효과가 증명되었으며, 특히 다른 항암제와 병용처리 시, 상호작용에 의한 상승효과가 있다고 알려져 있다. 현재 proteasome 억제제는 새로운 강력한 항암제로서 분류되어 있다. 본 연구는 사람혀편평세포암종세포(SCC25 cells)에서 lactacystin의 세포독성과 성장억제 효과, 그리고 세포자멸사의 유도에 대한 분자생물학적 기전을 밝히기 위해 실험을 시행하였다. SCC25 세포, 사람정상각화세포 (HaCaT cells) 그리고 사람치은섬유모세포(HGF-1 cells)의 생존율 측정은 MTT법을 시행하였고, SCC25 세포의 성장억제를 확인하기 위해서는 clonogenic assay를 사용하였다. lactcystin이 SCC25 세포에서 세포자멸사가 유도되는지를 확인하기 위해서 hoechst 염색법, hemacolor 염색법 그리고 TUNEL법을 시행하였다. 그리고 SCC25 세포에 lactacystin을 적용한 후, Western blot 분석, 세포면역화학염색, 공초점레이저주사현미경 검경, FACScan flow cytometry, 사립체막 전위변화, proteasome 활성도 측정 등을 시행하였다. Lactacystin으로 처리된 SCC25 세포는 시간 및 용량 의존적인 세포생존율의 감소, 용량의존적인 세포성장억제 그리고 세포자멸사에 의한 세포죽음을 보였다. 흥미롭게도 lactacytin은 정상세포인 HaCat 세포와 HGF-1 세포에서는 세포독성을 전혀 보이지 않았다. 그리고 lactacystin이 적용된 SCC25세포에서 핵 응축, DNA의 조각남, 사립체막전위와 proteasome 활성도의 감소, DNA 양의 감소, cytochrome c의 사립체에서의 세포질로의 유리, AIF와 DFF40 (CAD)의 핵으로의 이동, Bax의 증가, caspase-7, caspase-3, PARP, lamin A/C 그리고 DFF45 (ICAD)의 활성화 혹은 파괴와 같은 아주 다양한 세포자멸사 증거를 보였다. Flow cytometry 분석에서는 CDK 억제제인 $p21^{WAF1/CIP1}$$p27^{KIP1}$의 발현 증가와 관계있는 것으로 추정되어 지는 G1 세포주기 정지를 보였다. 이러한 결과는 lactacytin이 SCC25 세포에서 G1 세포주기정지와 proteasome, 사립체 및 caspase 경로의 연속반응을 통한 세포자멸사를 유도함을 명확하게 증명하고 있다. 이와 같은 세포주기 정지와 세포자멸사 유도능은 lactacytin이 사람혀편평상피세포암종의 새로운 치료전략으로서의 가능성을 제공한다고 생각한다.

Anti-Cancer Effect of IN-2001 in MDA-MB-231 Human Breast Cancer

  • Min, Kyung-Nan;Joung, Ki-Eun;Kim, Dae-Kee;Sheen, Yhun-Yhong
    • Biomolecules & Therapeutics
    • /
    • 제20권3호
    • /
    • pp.313-319
    • /
    • 2012
  • In recent years, inhibition of HDACs has emerged as a potential strategy to reverse aberrant epigenetic changes associated with cancer, and several classes of HDAC inhibitors have been found to have potent and specific anticancer activities in preclinical studies. But their precise mechanism of action has not been elucidated. In this study, a novel synthetic inhibitor of HDAC, 3-(4-dimethylamino phenyl)-N-hydroxy-2-propenamide [IN-2001] was examined for its antitumor activity and the underlying molecular mechanisms of any such activity on human breast cancer cell lines. IN-2001 effectively inhibited cellular HDAC activity ($IC_{50}$ = 0.585 nM) inMDA-MB-231 human breast cancer cells. IN-2001 caused a significant dose-dependent inhibition of cell proliferation in estrogen receptor (ER) negative MDA-MB-231human breast cancer cells. Cell cycle analysis revealed that the growth inhibitory effects of IN-2001 might be attributed to cell cycle arrest at $G_0/G_1$ and/or $G_2$/Mphase and subsequent apoptosis in human breast cancer cells. These events are accompanied by modulating several cell cycle and apoptosis regulatory genes such as CDK inhibitors $p21^{WAF1}$ and $p27^{KIP1}$ cyclin D1, and other tumor suppressor genes such as cyclin D2. Collectively, IN-2001 inhibited cell proliferation and induced apoptosis in human breast cancer cells and these findings may provide new therapeutic approaches, combination of antiestrogen together with a HDAC inhibitor, in the hormonal therapy-resistant ER-negative breast cancers. In summary, our data suggest that this histone deacetylase inhibitor, IN-2001, is a novel promising therapeutic agent with potent antitumor effects against human breast cancers.

고해상도 항공자력탐사를 이용한 Italia Vulcano-Lipari 화산 복합체의 천부 지하 구조 (Shallow subsurface structure of the Vulcano-Lipari volcanic complex, Italy, constrained by helicopter-borne aeromagnetic surveys)

  • 대웅무웅;중총 정;구택정부;삼원광언;중야 준
    • 지구물리와물리탐사
    • /
    • 제9권1호
    • /
    • pp.129-138
    • /
    • 2006
  • 남부 Italia Aeolian 군도의 Vulcano-Lipari 화산 복합체의 천부 지하구조를 잘 이해하고 또한 이 지역의 화산활동을 모니터링하기 위해서 고해상도 항공자력탐사가 3 년간의 간격을 두고 두 번 수행되었다. 두 개의 서로 다른 자력탐사 자료가 화산활동의 변화를 지시하는 어떠한 의미있는 차이를 보이지 않기 때문에, 자료들은 서로 합쳐져서 단일 자료보다 넓은 영역에 대한 항공자력도로 만들어졌다. 지형보정된 자력이상으로부터 겉보기 자화강도 분포도가 만들어졌으며 이로부터 Fossa 원추구의 이질성을 제시하는 국부적인 고 자화 이상을 볼 수 있었다. 이중 세 개의 고자화 이상에 대해 자력 모델링이 수행되었다. 각 모델은 Fossa 화구의 화산쇄설류로 덮혀있는 화산생성물의 존재를 밝히는 데에 적용되었다. Fossa 화구 지역에 대한 모델로부터 현재 화구의 남쪽 가장자리에는 조면암질 용암류가 묻혀있다는 것이 제시되었다. Forgia Vecchia에서 적용한 자력모델은 수증기 폭발성 원추구가, Fossa 칼데라를 메운 레타이트질 용암류(현무암질조면암과 안산암질조면암을 통칭)에 덮혀버린 한 분출중심으로부터 형성되었다는 것을 제시해 준다. 하지만 용암류의 분포는 기존의 시추 결과들로부터 알려진 것보다도 적은 지역에 국한되는 것처럼 나타난다. 이는 Porto Levante에 인접한 지열지역에서 알 수 있듯이, 강렬한 열수활동으로 인한 용암류의 부분적인 변질에 기인한 것으로 설명될 수 있다. Fossa 원추구 북동부에서의 모델은 두꺼운 용암류가 Fossa 화산활동의 초기단계에 또 다른 분출중심에서 집적되어 있다는 것을 암시한다. 최근의 전기탐사는 마지막 두 자력모델 지역에서 고비저항대를 보여준다.를 재활성화 시키는 $CO_2$의 탄성파 반응 또한 예측될 수 있다. 이 논문에서는 암석물리학 모의실험장치를 적용했던 현장(해상과 육상의 잠재적 $CO_2$ 격리 지역)의 사례를 보여주고 있다. 4차원 탄성파 반응들이 모니터링 프로그램의 설계를 돕기 위하여 만들어 졌다. 액체상의 $CO_2$ 주입은 공기로 포화된 상태에 비해 속도-유효응력 반응을 평균 약 8% 정도 낮게 한다. 실험자료들은 높은 유효응력에서 Gassmann 계산들과 일치한다. 이러한 이론과 일치하는 "임계" 유효응력은 사암의 종류에 따라 달라진다. 이 차이는 각각의 사암 종류의 미세구조에서 미세 균열 수의 차이에 기인한 것이라 생각된다. 높은 유효응력에서의 이론과 의미있게 일치하였으며, $CO_2$ 주입 시 현장에서의 탄성파 거동을 예상하는데 있어서 어느 정도 확신을 준다.극압 증가의 최대 허용치를 결정하는데는 사용할 수 없다고 주장하고자 한다. 초기폐사율이 낮음을 확인할수 있었으며, 상기 결과를 토대로, 넙치 치어의 경우 ${\beta}-1,3$ 글루칸을 0.05% 이상 0.1% 미만을 사료에 첨가하는 것이 성장, 사료효율 증진, 항산화능 및 질병저항성에 가장 좋은 효과를 나타낼 수 있을 것을 사료된다./Cip1}(-)/p27^{kip1}(-)$인 경우는 미만형인 경우(87.0%)가 장형(54.9%)의 경우보다 많은 비율을 차지하였다(P<0.05). 5년 장기 생존율에 있어서는 각각의 $p21^{Waf1/Cip1}$$p27^{kip1}$의 발현 유무에 따른 통계적인 유의성은 없었고 복합

Mechanism Underlying NaF-Induced Apoptosis in Human Oral Squamous Cell Carcinoma

  • Hur, Young-Joo;Kim, Do-Kyun;Lee, Seung-Eun;Kim, In-Ryoung;Jeong, Na-Young;Kim, Ji-Young;Park, Bong-Soo
    • International Journal of Oral Biology
    • /
    • 제35권2호
    • /
    • pp.51-60
    • /
    • 2010
  • Few studies have evaluated the apoptosis-inducing efficacy of NaF on cancer cells in vitro but there has been no previous investigation of the apoptotic effects of NaF on human oral squamous cell carcinoma cells. In this study, we have investigated the mechanisms underlying the apoptotic response to NaF treatment in the YD9 human squamous cell carcinoma cell line. The viability of YD9 cells and their growth inhibition were assessed by MTT and clonogenic assays, respectively. Hoechst staining, DNA electrophoresis and TUNEL staining were conducted to detect apoptosis. YD9 cells were treated with NaF, and western blotting, immunocytochemistry, confocal microscopy, FACScan flow cytometry, and MMP and proteasome activity assays were performed sequentially. The NaF treatment resulted in a time- and dose-dependent decrease in YD9 cell viability, a dose-dependent inhibition of cell growth, and the induction of apoptotic cell death. The apoptotic response of these cells was manifested by nuclear condensation, DNA fragmentation, the reduction of MMP and proteasome activity, a decreased DNA content, the release of cytochrome c into the cytosol, the translocation of AIF and DFF40 (CAD) into the nucleus, a significant shift of the Bax/Bcl-2 ratio, and the activation of caspase-9, caspase-3, PARP, Lamin A/C and DFF45 (ICAD). Furthermore, NaF treatment resulted in the downregulation of G1 cell cyclerelated proteins, and upregulation of p53 and the Cdk inhibitor $p27^{KIP1}$. Taken collectively, our present findings demonstrate that NaF strongly inhibits YD9 cell proliferation by modulating the expression of G1 cell cycle-related proteins and inducing apoptosis via mitochondrial and caspase pathways.

The Antimicrobial Peptide CopA3 Inhibits Clostridium difficile Toxin A-Induced Viability Loss and Apoptosis in Neural Cells

  • Yoon, I Na;Hwang, Jae Sam;Lee, Joon Ha;Kim, Ho
    • Journal of Microbiology and Biotechnology
    • /
    • 제29권1호
    • /
    • pp.30-36
    • /
    • 2019
  • Numerous studies have reported that enteric neurons involved in controlling neurotransmitter secretion and motility in the gut critically contribute to the progression of gut inflammation. Clostridium difficile toxins, which cause severe colonic inflammation, are also known to affect enteric neurons. Our previous study showed that C. difficile toxin A directly induces neural cell toxicities, such as viability loss and apoptosis. In the current study, we attempted to identify a potent inhibitor of toxin A-induced neural cell toxicity that may aid in managing toxin A-induced gut inflammation. In our recent study, we found that the Korea dung beetle-derived antimicrobial peptide CopA3 completely blocked neural cell apoptosis caused by okadaic acid or 6-OHDA. Here, we examined whether the antimicrobial peptide CopA3 inhibited toxin A-induced neural cell damage. In neuroblastoma SH-SY5Y cells, CopA3 treatment protected against both apoptosis and viability loss caused by toxin A. CopA3 also completely inhibited activation of the pro-apoptotic factor, caspase-3. Additionally, CopA3 rescued toxin A-induced downregulation of neural cell proliferation. However, CopA3 had no effect on signaling through ROS/p38 $MAPK/p27^{kip1}$, suggesting that CopA3 inhibits toxin A-induced neural cell toxicity independent of this well-characterized toxin A pathway. Our data further suggest that ability of CopA3 to rescue toxin A-induced neural cell damage may also ameliorate the gut inflammation caused by toxin A.