• 제목/요약/키워드: Mammalian cell

검색결과 731건 처리시간 0.029초

Dual Inhibition of PI3K/Akt/mTOR Pathway and Role of Autophagy in Non-Small Cell Lung Cancer Cells

  • Jeong, Eun-Hui;Choi, Hyeong-Sim;Lee, Tae-Gul;Kim, Hye-Ryoun;Kim, Cheol-Hyeon
    • Tuberculosis and Respiratory Diseases
    • /
    • 제72권4호
    • /
    • pp.343-351
    • /
    • 2012
  • Background: The phosphoinositide 3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR) signaling axis has emerged as a novel target for cancer therapy. Agents that inhibit this pathway are currently under development for lung cancer treatment. In the present study, we have tested whether dual inhibition of PI3K/Akt/mTOR signaling can lead to enahnced antitumor effects. We have also examined the role of autophagy during this process. Methods: We analyzed the combination effect of the mTOR inhibitor, temsirolimus, and the Akt inhibitor, GSK690693, on the survival of NCI-H460 and A549 non-small cell lung cancer cells. Cell proliferation was determined by MTT assay and apoptosis induction was evaluated by flow cytometry and terminal deoxynucleotidyl transferase dUTP nick end labeling assay. Autophagy induction was also evaluated by acridine orange staining. Changes of apoptosis or autophagy-related proteins were evaluated by western blot analysis. Results: Combination treatment with temsirolimus and GSK690693 caused synergistically increased cell death in NCI-H460 and A549 cells. This was attributable to increased induction of apoptosis. Caspase 3 activation and poly(ADP-ribose) polymerase cleavage accompanied these findings. Autophagy also increased and inhibition of autophagy resulted in increased cell death, suggesting its cytoprotective role during this process. Conclusion: Taken together, our results suggest that the combination of temsirolimus and GSK690693 could be a novel strategy for lung cancer therapy. Inhibition of autophagy could also be a promising method of enhancing the combination effect of these drugs.

Pre-surface antigen 지역과 poly(A) addition site가 포함된 B형 간염 표면항원 유전자의 재조합 (Cloning of the Hepatitis B Surface Antigen Containing Pre-surface Antigen Region and Poly(A) Addition Site)

  • Kim, Sang-Hae;Kim, Yong-Sok;Park, Mee-Young;Park, Hyune-Mo
    • 한국동물학회지
    • /
    • 제28권3호
    • /
    • pp.166-178
    • /
    • 1985
  • 한국형 B형 간염바이러스(HBV) DNA의 표면항원 유전자를 포유동물 세포에서 발현시켜 항원의 검출과 유전자의 분자유전학적인 연구를 하기 위하여 pre-surface antigen 지역과 표면항원 유전자 그리고 poly(A) addition site가 포함된 DNA 조각을 simian virus 40(SV 40)의 DNA 복제 원점과 promoter가 포함된 유전자 운반체에 재조함 시켰다. 우선, HBV DNA가 들어있는 pHBV 107을 Bam HI으로 부분절단한뒤 self-ligation시켜 두 HBV DNA가 같은 방향으로 들어간 pHBVD 107을 만들었다. 이 plasmid를 Bgl II로 절단하였을때 pre-surface지역과 표면항원 유전자 그리고 poly(A) addition site가 함께 포함된 2.7 kb의 insert DNA 조각을 얻었다. 유전자 운반체로는 포유동물세포에서 복제할 수 있도록 하기 위하여 SV40의 DNA 복제 원점부위와 72 bp repeats(enhancer)가 포함된 pSVOE를 만든다음 이 vector의 Pvu II 절단자리에 Bam HI linker를 붙여 insert DNA가 vector의 SV40 late promoter지역 가까이에 들어갈 수 있도록 변형시킨 pSVOB를 만들었다. 이상과 같이 만들어진 pre-surface 지역-표면항원유전자-poly(A)-addition site가 포함된 2.7 kb DNA 절편을 pSVOB promoter 뒤의 Bam HI site에 삽입하여 재조합된 plasmid pSVBS를 얻었다. 예비실험으로 pSVBS를 T-antigen이 생산되는 COS cell에 이주시켰더니 $HB_sAg$가 발현됨을 보았다.

  • PDF

Keratinization of Lung Squamous Cell Carcinoma Is Associated with Poor Clinical Outcome

  • Park, Hye Jung;Cha, Yoon-Jin;Kim, Seong Han;Kim, Arum;Kim, Eun Young;Chang, Yoon Soo
    • Tuberculosis and Respiratory Diseases
    • /
    • 제80권2호
    • /
    • pp.179-186
    • /
    • 2017
  • Background: Although the World Health Organization (WHO) classification of lung squamous cell carcinoma (SCC) was revised in 2015, its clinical implications for lung SCC subsets remain unclear. We investigated whether the morphologic characteristics of lung SCC, including keratinization, were associated with clinical parameters and clinical outcome of patients. Methods: A total of 81 patients who underwent curative surgical resection of diagnosed lung SCC, were enrolled in this study. Attributes such as keratinization, tumor budding, single cell invasion, and nuclear size within the tumor, as well as immunohistochemistry of Bcl-xL and pS6 expressions, were evaluated. Results: The keratinizing and nonkeratinizing subtypes did not differ with respect to age, sex, TNM stage, and morphologic parameters such as nuclear diameter, tumor budding, and single cell invasion at the tumor edge. Most patients with the keratinizing subtype (98.0%) had a history of smoking, whereas the nonkeratinizing group had a relatively higher proportion of never-smokers relative to the keratinizing group (24.0% vs. 2.0%; p=0.008, chi-square test). Expression of pS6 (a surrogate marker of mammalian target of rapamycin complex 1 [mTORC1] signaling that regulates keratinocyte differentiation), and Bcl-xL (a key anti-apoptotic molecule that may inhibit keratinization), did not correlate significantly with the presence of keratinization. Patients with the keratinizing subtype had a significantly shorter overall survival (85.2 months vs. 135.7 months, p=0.010, log-rank test), and a multivariate analysis showed that keratinization was an independent, poor prognostic factor (hazard ratio, 2.389; 95% confidence interval, 1.090-5.233; p=0.030). Conclusion: In lung SCC, keratinization is associated with a poor prognosis, and might be associated with smoking.

Characterization of the Effects of Silver Nanoparticles on Liver Cell Using HR-MAS NMR Spectroscopy

  • Kim, Si-Won;Kim, So-Sun;Lee, Sang-Mi;Kwon, Bo-Bae;Choi, Jin-Hee;Hyun, Jin-Won;Kim, Suhk-Mann
    • Bulletin of the Korean Chemical Society
    • /
    • 제32권6호
    • /
    • pp.2021-2026
    • /
    • 2011
  • AgNPs (silver nanoparticles) has been widely used for the commercial products, which have antimicrobial agent, medical devices, food industry and cosmetics. Despite, AgNPs have been reported as toxic to the mammalian cell, lung, liver, brain and other organs and many researchers have investigated the toxicity of AgNPs. In this study, we investigated toxicity of the AgNPs to the liver cell using metabolomics based on HRMAS NMR (High Resolution Magic Angle Spinning Nuclear Magnetic Resonance) technics, which could apply to the intact tissues or cells, to avoid the sample destruction. Target profiling and multivariative statistical analysis were performed to analyze the 1D $^1H$ spectrum. The results show that the concentrations of many metabolites were affected by the AgNPs in the liver cell. The concentrations of glutathione (GSH), lactate, taurine, and glycine were decreased and most of amino acids, choline analogues, and pyruvate were increased by the AgNPs. Moreover, the levels of the metabolites were recovered upto similar level of metabolites in the normal cell by the pre-treatment of NAC, external antioxidant. The results suggest that the depletion of the GSH by the AgNPs might induce the conversion of lactate and taurine to the pyruvate.

Surface Display of Heme- and Diflavin-Containing Cytochrome P450 BM3 in Escherichia coli: A Whole-Cell Biocatalyst for Oxidation

  • Yim, Sung-Kun;Kim, Dong-Hyun;Jung, Heung-Chae;Pan, Jae-Gu;Kang, Hyung-Sik;Ahn, Tae-Ho;Yun, Chul-Ho
    • Journal of Microbiology and Biotechnology
    • /
    • 제20권4호
    • /
    • pp.712-717
    • /
    • 2010
  • Cytochrome P450 enzymes (P450s) are involved in the synthesis of a wide variety of valuable products and in the degradation of numerous toxic compounds. The P450 BM3 (CYP102A1) from Bacillus megaterium was the first P450 discovered to be fused to its redox partner, a mammalian-like diflavin reductase. Here, we report the development of a whole-cell biocatalyst using ice-nucleation protein (Inp) from Pseudomonas syringae to display a hemeand diflavin-containing oxidoreductase, P450 BM3 (a single, 119-kDa polypeptide with domains of both an oxygenase and a reductase) on the surface of Escherichia coli. The surface localization and functionality of the fusion protein containing P450 BM3 were verified by flow cytometry and measurement of enzymatic activities. The results of this study comprise the first report of microbial cell-surface display of a heme- and diflavin-containing enzyme. This system should allow us to select and develop oxidoreductases containing heme and/or flavins into practically useful whole-cell biocatalysts for extensive biotechnological applications, including selective synthesis of new chemicals and pharmaceuticals, bioconversion, bioremediation, live vaccine development, and biochip development.

Tunicamycin-Induced ER Stress Upregulates the Expression of Mitochondrial HtrA2 and Promotes Apoptosis Through the Cytosolic Release of HtrA2

  • Han, Chul;Nam, Min-Kyung;Park, Hyo-Jin;Seong, Young-Mo;Kang, Seong-Man;Rhim, Hyang-Shuk
    • Journal of Microbiology and Biotechnology
    • /
    • 제18권6호
    • /
    • pp.1197-1202
    • /
    • 2008
  • Recent studies provide some evidence that the HtrA2 protein is intimately associated with the pathogenesis of neurodegenerative disorders and that endoplasmic reticulum (ER) quality control and ER stress-associated cell death play critical roles in neuronal cell death. However, little is known about the intimate relationship between HtrA2 and ER stress-associated cellular responses. In the present study, we have demonstrated that the HtrA2 protein level was gradually and significantly increased by up to to-fold in the mitochondria under tunicamycin (Tm)-induced ER stress, which eventually promoted cell death through the release of HtrA2 into the cytoplasm. Using an ecdysone-inducible mammalian expression system, we demonstrate that the extent of cell death in 293-HtrA2 cells was approximately 20 times higher under Tm-induced ER stress, indicating that the increase in the HtrA2 protein level in the mitochondria itself is necessary but not sufficient for the promotion of cell death. Taken together, these results suggest that HtrA2 may serve as a mediator of ER stress-induced apoptosis and ER-mitochondrial cross-talk in some cellular processes.

Functional Analyses of Centrosomal Proteins, Nek2 and NuMA in Development of Mouse Gametes and Early Embryos

  • Youn, Hong-Hee;Oh, Hwa-Soon;Lee, Kwang-Hee;Son, Chae-Ick;Lee, Sang-Ho
    • 한국동물번식학회:학술대회논문집
    • /
    • 한국동물번식학회 2003년도 학술발표대회 발표논문초록집
    • /
    • pp.96-96
    • /
    • 2003
  • Nek2 (NIMA-related protein) is a mammalian cell cycle-regulated kinase that involves in chromosome condensation and centrosome regulation and NuMA (nuclear mitotic apparatus protein) is involved in spindle assembly during a cell cycle. The cellular distribution and organization of the centrosomal components is completely unknown during fertilization and embryonic development. We examined distribution of two well-known centrosomal proteins, Nek2 and NuMA in mouse gametes and embryos to get an insight in the reorganization of centrosomal proteins during germ cell development and early fertilization. Spermatogenic cells, gametes, and embryos were analyzed with anti-Nek2 or -NuMA antibodies by immunological assay, RT-PCR, and overexpression through gene transfection. Mitotically or meiotically active spermatogenic cells were intensively stained with these antibodies in both centrosomes and cytoplasm, whereas the oocytes showed different staining patterns depending on the meiotic stages. During maturation, GV, GVBD, and MI stage were clearly stained with NuMA antibody in the nucleus or cytoplasm at MII. Also, Nek2 was detectable in cytoplasm as scattered spots or chromosome associated at MII. In early developmental embryo, NuMA was detected in nucleus of each blastomere, while Nek2 was detected in cytoplasm. In contrast to previously reported results, Nek2 and NuMA were detected in both decondensing head, and the centriole of demembranated and decondensed sperm or whole body of trypsin-treated sperm for Nek2. During meiotic progress in oocytes, transcripts levels were the highest in MI stage and then downregulated in MII. Also, it shows dramatically change in early developmental embryos, firstly, it was increased until 4 cell stage and reduced in 8 cell stage, and finally, transcript levels were upregulated until blastoscyst. This finding suggests that cnetrosomal component may play an important role in reorganizing of functional centrosome during fertilization process and subsequent development.

  • PDF

Induction of cytoprotective autophagy by morusin via AMP-activated protein kinase activation in human non-small cell lung cancer cells

  • Park, Hyun-Ji;Park, Shin-Hyung
    • Nutrition Research and Practice
    • /
    • 제14권5호
    • /
    • pp.478-489
    • /
    • 2020
  • BACKGROUND/OBJECTIVES: Morusin, a marker component of Morus alba L., possesses anti-cancer activity. The objective of this study was to determine autophagy-inducing effect of morusin in non-small cell lung cancer (NSCLC) cells and investigate the underlying mechanism. SUBJECTS/METHODS: Autophagy induction and the expression of autophagy-related proteins were analyzed by LC3 immunofluorescence and western blot, respectively. The role of autophagy and AMP-activated protein kinase (AMPK) was determined by treating NSCLC cells with bafilomycin A1, an autophagy inhibitor, and compound C, an AMPK inhibitor. Cytotoxicity and apoptosis induction were determined by MTT assay, trypan blue exclusion assay, annexin V-propidium iodide (PI) double staining assay, and cell cycle analysis. RESULTS: Morusin increased the formation of LC3 puncta in the cytoplasm and upregulated the expression of autophagy-related 5 (Atg5), Atg12, beclin-1, and LC3II in NSCLC cells, demonstrating that morusin could induce autophagy. Treatment with bafilomycin A1 markedly reduced cell viability but increased proportions of sub-G1 phase cells and annexin V-positive cells in H460 cells. These results indicate that morusin can trigger autophagy in NSCLC cells as a defense mechanism against morusin-induced apoptosis. Furthermore, we found that AMPK and its downstream acetyl-CoA carboxylase (ACC) were phosphorylated, while mammalian target of rapamycin (mTOR) and its downstream p70S6 kinase (p70S6K) were dephosphorylated by morusin. Morusin-induced apoptosis was significantly increased by treatment with compound C in H460 cells. These results suggest that morusin-induced AMPK activation could protect NSCLC cells from apoptosis probably by inducing autophagy. CONCLUSIONS: Our findings suggest that combination treatment with morusin and autophagy inhibitor or AMPK inhibitor might enhance the clinical efficacy of morusin for NSCLC.

Effects of C-Terminal Residues of 12-Mer Peptides on Antibacterial Efficacy and Mechanism

  • Son, Kkabi;Kim, Jieun;Jang, Mihee;Chauhan, Anil Kumar;Kim, Yangmee
    • Journal of Microbiology and Biotechnology
    • /
    • 제29권11호
    • /
    • pp.1707-1716
    • /
    • 2019
  • The development of new antimicrobial agents is essential for the effective treatment of diseases such as sepsis. We previously developed a new short peptide, Pap12-6, using the 12 N-terminal residues of papiliocin, which showed potent and effective antimicrobial activity against multidrug-resistant Gram-negative bacteria. Here, we investigated the antimicrobial mechanism of Pap12-6 and a newly designed peptide, Pap12-7, in which the 12th Trp residue of Pap12-6 was replaced with Val to develop a potent peptide with high bacterial selectivity and a different antibacterial mechanism. Both peptides showed high antimicrobial activity against Gram-negative bacteria, including multidrug-resistant Gram-negative bacteria. In addition, the two peptides showed similar anti-inflammatory activity against lipopolysaccharide-stimulated RAW 264.7 cells, but Pap12-7 showed very low toxicities against sheep red blood cells and mammalian cells compared to that showed by Pap12-6. A calcein dye leakage assay, membrane depolarization, and confocal microscopy observations revealed that the two peptides with one single amino acid change have different mechanisms of antibacterial action: Pap12-6 directly targets the bacterial cell membrane, whereas Pap12-7 appears to penetrate the bacterial cell membrane and exert its activities in the cell. The therapeutic efficacy of Pap12-7 was further examined in a mouse model of sepsis, which increased the survival rate of septic mice. For the first time, we showed that both peptides showed anti-septic activity by reducing the infiltration of neutrophils and the production of inflammatory factors. Overall, these results indicate Pap12-7 as a novel non-toxic peptide with potent antibacterial and anti-septic activities via penetrating the cell membrane.

In vitro and in vivo pharmacokinetic characterization of LMT-28 as a novel small molecular interleukin-6 inhibitor

  • Ahn, Sung-Hoon;Heo, Tae-Hwe;Jun, Hyun-Sik;Choi, Yongseok
    • Asian-Australasian Journal of Animal Sciences
    • /
    • 제33권4호
    • /
    • pp.670-677
    • /
    • 2020
  • Objective: Interleukin-6 (IL-6) is a T cell-derived B cell stimulating factor which plays an important role in inflammatory diseases. In this study, the pharmacokinetic properties of LMT-28 including physicochemical property, in vitro liver microsomal stability and an in vivo pharmacokinetic study using BALB/c mice were characterized. Methods: LMT-28 has been synthesized and is being developed as a novel therapeutic IL-6 inhibitor. The physicochemical properties and in vitro pharmacokinetic profiles such as liver microsomal stability and Madin-Darby canine kidney (MDCK) cell permeability assay were examined. For in vivo pharmacokinetic studies, pharmacokinetic parameters using BALB/c mice were calculated. Results: The logarithm of the partition coefficient value (LogP; 3.65) and the apparent permeability coefficient values (Papp; 9.7×10-6 cm/s) showed that LMT-28 possesses a moderate-high cell permeability property across MDCK cell monolayers. The plasma protein binding rate of LMT-28 was 92.4% and mostly bound to serum albumin. The metabolic half-life (t1/2) values of LMT-28 were 15.3 min for rat and 21.9 min for human at the concentration 1 μM. The area under the plasma drug concentration-time curve and Cmax after oral administration (5 mg/kg) of LMT-28 were 302±209 h·ng/mL and 137±100 ng/mL, respectively. Conclusion: These data suggest that LMT-28 may have good physicochemical and pharmacokinetic properties and may be a novel oral drug candidate as the first synthetic IL-6 inhibitor to ameliorate mammalian inflammation.