• 제목/요약/키워드: potential therapeutic target

검색결과 381건 처리시간 0.025초

Constitutive Activation of $p70^{S6k}$ in Cancer Cells

  • Kwon, Hyoung-Keun;Bae, Gyu-Un;Yoon, Jong-Woo;Kim, Yong-Kee;Lee, Hoi-Young;Lee, Hyang-Woo;Han, Jeung-Whan
    • Archives of Pharmacal Research
    • /
    • 제25권5호
    • /
    • pp.685-690
    • /
    • 2002
  • The mitogen-stimulated serine/threonine kinase $p70^{S6k}$ plays an important role in the progression of cells from $G_0/G$_1$$ to S phase of the cell cycle by translational up-regulation of a family of mRNA transcripts family of mRNA transcripts which contain polypyrimidine tract at their 5 transcriptional start site. Here, we report that $p70^{S6k}$ was constitutively phosphorylated and activated to various degrees in serum-deprived AGS, A2058, HT-1376, MG63, MCF7, MDA-MB-435S, MDA-MB-231 and MB-157. Rapamycin treatment induced a significant dephosphorylation and inactivation of $p70^{S6k}$ in all cancer cell lines, while wortmannin, a specific inhibitor of PI3-K, caused a mild dephosphorylation of $p70^{S6k}$ in AGS, MDA-MB-435S and MB-157. In addition, SQ20006, methylxanthine phosphodiesterase inhibitor, reduced the phosphorylation of $p70^{S6k}$ in all cancer cells tested. Consistent with inhibitory effect of rapamycin on $p70^{S6k}$ activity, rapamycin inhibited [$^3H$]-thymidine incorporation and increased the number of cells at $G_{0}G_{1}$ phase. Furthermore, these inhibitory effects were accompanied by the decrease in growth of cancer cells. Taken together, the results indicate that the antiproliferative activity of rapamycin might be attributed to cell cycle arrest at $G_{0}G_{1}$ phase in human cancer cells through the inhibition of constitutively activated $p70^{S6k}$ of cancer cells and suggest $p70^{S6k}$ as a potential target for therapeutic strategies aimed at preventing or inhibiting tumor growth.

ATM Signaling Pathway Is Implicated in the SMYD3-mediated Proliferation and Migration of Gastric Cancer Cells

  • Wang, Lei;Wang, Qiu-Tong;Liu, Yu-Peng;Dong, Qing-Qing;Hu, Hai-Jie;Miao, Zhi;Li, Shuang;Liu, Yong;Zhou, Hao;Zhang, Tong-Cun;Ma, Wen-Jian;Luo, Xue-Gang
    • Journal of Gastric Cancer
    • /
    • 제17권4호
    • /
    • pp.295-305
    • /
    • 2017
  • Purpose: We previously found that the histone methyltransferase suppressor of variegation, enhancer of zeste, trithorax and myeloid-nervy-deformed epidermal autoregulatory factor-1 domain-containing protein 3 (SMYD3) is a potential independent predictive factor or prognostic factor for overall survival in gastric cancer patients, but its roles seem to differ from those in other cancers. Therefore, in this study, the detailed functions of SMYD3 in cell proliferation and migration in gastric cancer were examined. Materials and Methods: SMYD3 was overexpressed or suppressed by transfection with an expression plasmid or siRNA, and a wound healing migration assay and Transwell assay were performed to detect the migration and invasion ability of gastric cancer cells. Additionally, an MTT assay and clonogenic assay were performed to evaluate cell proliferation, and a cell cycle analysis was performed by propidium iodide staining. Furthermore, the expression of genes implicated in the ataxia telangiectasia mutated (ATM) pathway and proteins involved in cell cycle regulation were detected by polymerase chain reaction and western blot analyses. Results: Compared with control cells, gastric cancer cells transfected with si-SMYD3 showed lower migration and invasion abilities (P<0.05), and the absence of SMYD3 halted cells in G2/M phase and activated the ATM pathway. Furthermore, the opposite patterns were observed when SMYD3 was elevated in normal gastric cells. Conclusions: To the best of our knowledge, this study provides the first evidence that the absence of SMYD3 could inhibit the migration, invasion, and proliferation of gastric cancer cells and halt cells in G2/M phase via the ATM-CHK2/p53-Cdc25C pathway. These findings indicated that SMYD3 plays crucial roles in the proliferation, migration, and invasion of gastric cancer cells and may be a useful therapeutic target in human gastric carcinomas.

Endometrial profilin 1: A key player in embryo-endometrial crosstalk

  • Lee, Chang-Jin;Hong, Seon-Hwa;Yoon, Min-Ji;Lee, Kyung-Ah;Ko, Jung-Jae;Koo, Hwa Seon;Kim, Jee Hyun;Choi, Dong Hee;Kwon, Hwang;Kang, Youn-Jung
    • Clinical and Experimental Reproductive Medicine
    • /
    • 제47권2호
    • /
    • pp.114-121
    • /
    • 2020
  • Objective: Despite extensive research on implantation failure, little is known about the molecular mechanisms underlying the crosstalk between the embryo and the maternal endometrium, which is critical for successful pregnancy. Profilin 1 (PFN1), which is expressed both in the embryo and in the endometrial epithelium, acts as a potent regulator of actin polymerization and the cytoskeletal network. In this study, we identified the specific role of endometrial PFN1 during embryo implantation. Methods: Morphological alterations depending on the status of PFN1 expression were assessed in PFN1-depleted or control cells grown on Matrigel-coated cover glass. Day-5 mouse embryos were cocultured with Ishikawa cells. Comparisons of the rates of F-actin formation and embryo attachment were performed by measuring the stability of the attached embryo onto PFN1-depleted or control cells. Results: Depletion of PFN1 in endometrial epithelial cells induced a significant reduction in cell-cell adhesion displaying less formation of colonies and a more circular cell shape. Mouse embryos co-cultured with PFN1-depleted cells failed to form actin cytoskeletal networks, whereas more F-actin formation in the direction of surrounding PFN1-intact endometrial epithelial cells was detected. Furthermore, significantly lower embryo attachment stability was observed in PFN1-depleted cells than in control cells. This may have been due to reduced endometrial receptivity caused by impaired actin cytoskeletal networks associated with PFN1 deficiency. Conclusion: These observations definitively demonstrate an important role of PFN1 in mediating cell-cell adhesion during the initial stage of embryo implantation and suggest a potential therapeutic target or novel biomarker for patients suffering from implantation failure.

적소두 추출물이 톨유사수용체 활성 및 염증유발 사이토카인의 생성에 미치는 영향 (Effect of Vigna angularis on Toll-like Receptor Activation and Pro-inflammatory Cytokine Production)

  • 김미화;정시화;이승웅;김현규;박찬선;전병훈;오현미;노문철
    • 동의생리병리학회지
    • /
    • 제26권4호
    • /
    • pp.511-518
    • /
    • 2012
  • The mechanisms of Toll-like receptor (TLR) signaling have been the focus of extensive studies because TLRs are the target of therapeutic intervention on multiple diseases. In this study, we investigated the inhibitory potential of Vigna angularis (azuki bean) on the TLR signaling. The effect of Vigna angularis extract (JSD) on TLR activation was investigated by assessing NF-${\kappa}B$ and AP-1 inducible secreted embryonic alkaline phosphatase (SEAP) activity. JSD significantly inhibited SEAP activity induced by poly I:C (TLR3 ligand) and poly I (TLR7 ligand) in a dose-dependent manner at concentration below 100 ${\mu}g/ml$ with no sign of cytotoxicity. Pretreatment of JSD markedly suppressed mRNA expressions of pro-inflammatory cytokines and adhesive molecules such as TNF-${\alpha}$, IL-6, RANTES, MCP-1 and ICAM-1 induced by TLR ligands. It also diminished the phosphorylation of $I{\kappa}B$ kinase and $I{\kappa}B$, and followed by $I{\kappa}B$-mediated nuclear translocation of p50, p65, and phosphorylation of p38, JNK, and IRF signaling pathway. In conclusion, our results suggest that Vigna angularis has inhibitory activity on TLR-3 and -7 signaling and it can be further developed as a remedy in curing TLR-related multiple diseases.

Suicidal gene therapy with rabbit cytochrome P450 4B1/2-aminoanthracene or 4-ipomeanol system in human colon cancer cell

  • Jang, Su Jin;Kang, Joo Hyun;Moon, Byung Seok;Lee, Yong Jin;Kim, Kwang Il;Lee, Tae Sup;Choe, Jae Gol;Lim, Sang Moo
    • 대한방사성의약품학회지
    • /
    • 제1권2호
    • /
    • pp.118-122
    • /
    • 2015
  • Suicidal gene therapy is based on the transduction of tumor cells with "suicide" genes encoding for prodrug-activating enzymes that render target cells susceptible to prodrug treatment. Suicidal gene therapy results in the death of tumor with the expression of gene encoding enzyme that converts non-toxic prodrug into cytotoxic product. Cytochrome P450 4B1 (CYP4B1) activates 4-ipomeanol (4-IPO) or 2-aminoanthracene (2-AA) to cytotoxic furane epoxide and unsaturated dialdehyde intermediate.In this study, therapeutic effects of suicidal gene therapy with rabbit CYP4B1/2-AA or 4-IPO system were evaluated in HT-29 (human colon cancer cell). pcDNA-CYP4B1 vector was transfected into HT-29 by lipofection and stable transfectant was selected by treatment of hygromycin ($500{\mu}g/mL$) for 3 weeks. Reverse transcription polymerase chain reaction (RT-PCR) analysis was performed for confirmation of CYP4B1 expression in CYP4B1 gene transduced cell. The cytotoxic effects of CYP4B1 transduced cell were determined using dye-exclusion assay after treatment of 2-AA or 4-IPO for 96 hrs. Dye-exclusion assay showed that $IC_{50}$ of HT-29 and CYP4B1 transduced HT-29 was 0.01 mM and 0.003 mM after 4-IPO or 2-AA treatment at 96 hrs exposure, respectively. In conclusion, CYP4B1 based prodrug gene therapy probably have the potential for treatment of colorectal adenocarcinoma.

결장암세포에서 sanguinarine에 의한 종양억제 유전자 p53 의존적 apoptosis 유도 (Induction of Tumor Suppressor Gene p53-dependent Apoptosis by Sanguinarine in HCT116 Human Colorectal Cancer Cells)

  • 최영현
    • 생명과학회지
    • /
    • 제31권4호
    • /
    • pp.400-409
    • /
    • 2021
  • 천연 benzophenanthridine alkaloid의 일종인 sanguinarine에 의한 인간 암세포에서의 세포사멸 유도는 암 치료를 위한 잠재적 치료 가능성으로 여겨져 왔으나 기본적인 항암 기전은 여전히 불분명하다. 종양 억제제 p53의 결실 또는 돌연변이는 결장암세포의 항암제 내성에 대한 주요 원인으로 작용하다. 따라서, 본 연구에서는 정상 p53을 가진 HCT116 (p53+/+) 및 p53이 결여된 HCT116 (p53-/-) 결장암세포를 대상으로 sanguinarine에 의해 유도되는 세포사멸에서 p53의 역할을 조사하였다. 본 연구의 결과에 의하면, sanguinarine은 HCT116 (p53-/-) 세포에 비하여 HCT116 (p53+/+) 세포의 생존력을 현저히 감소시켰다. 아울러 sanguinarine은 HCT116 (p53-/-) 세포보다 HCT116 (p53+/+) 세포에서 p53 및 cyclin-dependent kinase 억제제 p21WAF1/CIP1의 발현을 증가시키면서 DNA 손상 및 세포사멸의 유도를 증가시켰다. Sanguinarine은 HCT116 (p53+/+) 세포에서 외인성 및 내인성 세포사멸의 개시에 관여하는 caspase-8 및 caspase-9의 활성을 증가시켰으며, 전형적인 효과기 caspase인 caspase-3을 활성화시켰다. 또한, sanguinarine은 HCT116 (p53+/+) 세포에서 Bax/Bcl-2의 발현 비율을 증가시키고 미토콘드리아 손상을 유발하였지만, HCT116 (p53-/-) 세포에서는 이러한 현상이 관찰되지 않았다. 결론적으로 본 연구의 결과는 sanguinarine은 HCT116 결장암세포에서 p53 의존적으로 외인성 및 내인성 세포사멸의 경로 활성을 통하여 세포사멸을 유도하였음을 의미한다.

Non-Polar Myxococcus fulvus KYC4048 Metabolites Exert Anti-Proliferative Effects via Inhibition of Wnt/β-Catenin Signaling in MCF-7 Breast Cancer Cells

  • Park, Juha;Yoo, Hee-Jin;Yu, Ah-Ran;Kim, Hye Ok;Park, Sang Cheol;Jang, Young Pyo;Lee, Chayul;Choe, Wonchae;Kim, Sung Soo;Kang, Insug;Yoon, Kyung-Sik
    • Journal of Microbiology and Biotechnology
    • /
    • 제31권4호
    • /
    • pp.540-549
    • /
    • 2021
  • The Wnt/β-catenin signaling pathway is involved in breast cancer and Myxococcus fulvus KYC4048 is a myxobacterial strain that can produce a variety of bioactive secondary metabolites. Although a previous study revealed that KYC4048 metabolites exhibit anti-proliferative effects on breast cancer, the biochemical mechanism involved in their effects remains unclear. In the present study, KYC4048 metabolites were separated into polar and non-polar (ethyl acetate and n-hexane) fractions via liquid-liquid extraction. The effects of these polar and non-polar KYC4048 metabolites on the viability of breast cancer cells were then determined by MTT assay. Expression levels of Wnt/β-catenin pathway proteins were determined by Western blot analysis. Cell cycle and apoptosis were measured via fluorescence-activated cell sorting (FACS). The results revealed that non-polar KYC4048 metabolites induced cell death of breast cancer cells and decreased expression levels of WNT2B, β-catenin, and Wnt target genes (c-Myc and cyclin D1). Moreover, the n-hexane fraction of non-polar KYC4048 metabolites was found most effective in inducing apoptosis, necrosis, and cell cycle arrest, leading us to conclude that it can induce apoptosis of breast cancer cells through the Wnt/β-catenin pathway. These findings provide evidence that the n-hexane fraction of non-polar KYC4048 metabolites can be developed as a potential therapeutic agent for breast cancer via inhibition of the Wnt/β-catenin pathway.

Ubiquitin D Promotes Progression of Oral Squamous Cell Carcinoma via NF-Kappa B Signaling

  • Song, An;Wang, Yi;Jiang, Feng;Yan, Enshi;Zhou, Junbo;Ye, Jinhai;Zhang, Hongchuang;Ding, Xu;Li, Gang;Wu, Yunong;Zheng, Yang;Song, Xiaomeng
    • Molecules and Cells
    • /
    • 제44권7호
    • /
    • pp.468-480
    • /
    • 2021
  • Ubiquitin D (UBD) is highly upregulated in many cancers, and plays a pivotal role in the pathophysiological processes of cancers. However, its roles and underlying mechanisms in oral squamous cell carcinoma (OSCC) are still unclear. In the present study, we investigated the role of UBD in patients with OSCC. Quantitative real-time polymerase chain reaction and Western blot were used to measure the expression of UBD in OSCC tissues. Immunohistochemistry assay was used to detect the differential expressions of UBD in 244 OSCC patients and 32 cases of normal oral mucosae. In addition, CCK-8, colony formation, wound healing and Transwell assays were performed to evaluate the effect of UBD on the cell proliferation, migration, and invasion in OSCC. Furthermore, a xenograft tumor model was established to verify the role of UBD on tumor formation in vivo. We found that UBD was upregulated in human OSCC tissues and cell lines and was associated with clinical and pathological features of patients. Moreover, the overexpression of UBD promoted the proliferation, migration and invasion of OSCC cells; however, the knockdown of UBD exerted the opposite effects. In this study, our results also suggested that UBD promoted OSCC progression through NF-κB signaling. Our findings indicated that UBD played a critical role in OSCC and may serve as a prognostic biomarker and potential therapeutic target for OSCC treatment.

나노 구조체를 이용한 산화질소 전달체에 대한 연구 및 바이오메디컬 응용 (Nitric Oxide Delivery using Nanostructures and Its Biomedical Applications)

  • 최윤서;정혜중;박경태;홍진기
    • Korean Chemical Engineering Research
    • /
    • 제57권3호
    • /
    • pp.305-312
    • /
    • 2019
  • 산화질소(NO)가 혈관 확장, 혈소판 응집 억제, 면역 반응 조절, 상처 치료, 항암 등의 주요 병리 생리학적 프로세스에 관여한다는 사실이 밝혀지면서 최근 산화질소 전달에 대한 국내외 연구진들의 관심이 높아지고 있다. 그러나 인체에 이상적으로 적용될 수 있는 산화질소 전달체의 개발은 산화질소의 높은 반응성과 짧은 반감기로 인하여 여전히 난제로 남아 있다. 본 논문에서는 다양한 산화질소 전달체 중에서도 최근 바이오메디컬 분야에서 연구가 활발히 이루어지고 있는 나노 구조체를 이용한 전달체의 연구 결과 및 응용 방향에 대해서 소개하고자 한다. 나노 크기의 구조체가 다른 전달체와 비교하여 가지는 장점은 표면 대 부피 비율이 높아 산화질소를 효율적으로 탑재할 수 있고, 표면 개조능력이 뛰어나 산화질소의 방출 양상을 효과적으로 제어할 수 있다는 것이다. 특히 이 글에서는 다양한 나노 구조체 중에서도 나노입자 형태, 마이크로에멀젼 형태, 그리고 다층필름 형태의 나노 구조체에 대해서 다룸으로써 각 구조체의 산화질소 방출 양상을 비교하고 그 특징에 대해서 자세히 알아보고자 한다. 이와 같은 나노 구조체의 개발은 산화질소의 급격한 방출을 방지하고 지효성을 띠게 함으로써 타겟 부위에서의 효과를 높일 수 있을 것으로 기대되며, 더 나아가 차후 다양한 바이오메디컬 분야에서 유망한 치료 기제로서 적용될 수 있을 것으로 보인다.

스코폴라민으로 유도한 기억력 손상 모델에서 소엽 추출물의 보호 효과 (Perilla Frutescens Extract Protects against Scopolamine-Induced Memory Deficits in Mice)

  • 이지혜;이은홍;정은미;김동현;김성규;박미희;정지욱
    • 동의생리병리학회지
    • /
    • 제35권3호
    • /
    • pp.97-103
    • /
    • 2021
  • Perilla frutescens (P. frutescens) is an important herb used for many purposes such as medicinal, aromatic, and functional food in Asian countries and has beneficial effects such as antioxidant activity, anti-inflammation activity, anti-depression activity, and anxiolytic activity. However, there have been no studies on the protective effect of P. frutescens extract (PFE) on amnesia in vivo. The present study aimed to investigate whether PFE protects memory deficit using a scopolamine-induced mice model and elucidate the underlying mechanisms involved. The protective effect of PFE against scopolamine-induced memory deficits was investigated using Y-maze, passive avoidance, and Morris water maze tests. Furthermore, the potential mechanisms of PFE in improving memory capabilities related to the cholinergic system and antioxidant activity were examined. PFE significantly increased spontaneous alternation in the Y-maze test, step-through latency in the passive avoidance test, and swimming time in the target quadrant in the probe test when compared to the scopolamine-treated group. Likewise, PFE significantly decreased escapes latency in the Morris water maze test. PFE could not regulate cholinergic function in acetylcholine level and acetylcholine esterase activity. However, PFE increased DPPH radical scavenging activity dose-dependently and total polyphenol content was 127.7±1.2 ㎍ GAE/mg. The results showed that the PFE could be a preventive and/or therapeutic candidate for memory and cognitive dysfunction in Alzheimer's disease.