• Title/Summary/Keyword: AIF

Search Result 72, Processing Time 0.024 seconds

Topology Optimization of Shell Structures Using Adaptive Inner-Front(AIF) Level Set Method (적응적 내부 경계를 갖는 레벨셋 방법을 이용한 쉘 구조물의 위상최적설계)

  • Park, Kang-Soo;Youn, Sung-Kie
    • Proceedings of the Computational Structural Engineering Institute Conference
    • /
    • 2007.04a
    • /
    • pp.157-162
    • /
    • 2007
  • A new level set based topology optimization employing inner-front creation algorithm is presented. In the conventional level set based topology optimization, the optimum topology strongly depends on the initial level set distribution due to the incapability of inner-front creation during optimization process. In the present work, in this regard, an inner-front creation algorithm is proposed. in which the sizes. shapes. positions, and number of new inner-fronts during the optimization process can be globally and consistently identified by considering both the value of a given criterion for inner-front creation and the occupied volume (area) of material domain. To facilitate the inner-front creation process, the inner-front creation map which corresponds to the discrete valued criterion of inner-front creation is applied to the level set function. In order to regularize the design domain during the optimization process, the edge smoothing is carried out by solving the edge smoothing partial differential equation (PDE). Updating the level set function during the optimization process, in the present work, the least-squares finite element method (LSFEM) is employed. As demonstrative examples for the flexibility and usefulness of the proposed method. the level set based topology optimization considering lightweight design of 3D shell structure is carried out.

  • PDF

Selective Suppression of a Subset of Bax-dependent Neuronal Death by a Cell Permeable Peptide Inhibitor of Bax, BIP

  • Kim, Soo-Young;Kim, Hyun;Sun, Woong
    • Animal cells and systems
    • /
    • v.12 no.4
    • /
    • pp.211-217
    • /
    • 2008
  • Bax, a pro-apoptotic member of Bcl-2 family proteins, plays a central role in the mitochondria-dependent apoptosis. Apoptotic signals induce the translocation of Bax from cytosol into the mitochondria, which triggers the release of apoptogenic molecules such as cytochrome C and apoptosis-inducing factor, AIF. Bax-inhibiting peptide(BIP) is a cell permeable peptide comprised of five amino acids designed from the Bax-interaction domain of Ku70. Because BIP inhibits Bax translocation and Bax-mediated release of cytochrome C, BIP suppresses Bax-dependent apoptosis. In this study, we observed that BIP inhibited staurosporine-induced neuronal death in cultured cerebral cortex and cerebellar granule cells, but BIP failed to rescue granule cells from trophic signal deprivation-induced neuronal death, although both staurosporine-induced and trophic signal deprivation-induced neuronal death are dependent on Bax. These findings suggest that the mechanisms of the Bax activation may differ depending on the type of cell death induction, and thus BIP exhibits selective suppression of a subtype of Bax-dependent neuronal death.

An Epithermal Neutron Beam Design for BNCT Using $^2H(d,n)^3He$ Reaction

  • Han, Chi-Young;Kim, Jong-Kyung;Chung, Kyu-Sun
    • Nuclear Engineering and Technology
    • /
    • v.31 no.5
    • /
    • pp.512-521
    • /
    • 1999
  • A feasibility study was performed to design an epithermal neutron beam for BNCT using the neutron of 2.45 MeV on the average produced from $^2H(d,n)^3$He reaction induced by plasma focus in the z-pinch instead of the conventional accelerator-based $^3H(d, n)^4$He neutron generator. Flux and spectrum were analyzed to use these neutrons as the neutron source for BNCT. Neutronic characteristics of several candidate materials in this neutron source were investigated Using MCNP Code, and $^7LiF$ ; 40%Al + 60%$AIF_3$, and Pb Were determined as moderator, filter, and reflector in an epithermal neutron beam design for BNCT, respectively. The skin-skull-brain ellipsoidal phantom, which consists of homogeneous regions of skin-, bone-, or brain-equivalent material, was used in order to assess the dosimetric effect in brain. An epithermal neutron beam design for BNCT was proposed by the repeated work with MCNP runs, and the dosimetric properties (AD, AR, ADDR, and Dose Components) calculated within the phantom showed that the neutron beam designed in this work is effective in tumor therapy. If the neutron source flux is high enough using the z-pinch plasma, BNCT using the neutron source produced from $^2H(d,n)^3$He reaction will be very feasible.

  • PDF

Role of Annexin A5 on Mitochondria-Dependent Apoptosis Induced by Tetramethoxystilbene in Human Breast Cancer Cells

  • Hong, Mihye;Park, Nahee;Chun, Young-Jin
    • Biomolecules & Therapeutics
    • /
    • v.22 no.6
    • /
    • pp.519-524
    • /
    • 2014
  • We have previously shown that 2,4,3',5'-tetramethoxystilbene (TMS), a trans-stilbene analogue, induces apoptosis in human cancer cells. However, the detailed mechanisms of mitochondria-dependent apoptosis induced by TMS are not fully understood. In the present study, the possible roles of annexin A5 in TMS-mediated apoptosis were investigated in MCF7 human breast cancer cells. Quantitative real-time PCR analysis and Western blot analysis showed that the expression of annexin A5 was strongly increased in TMS-treated cells. TMS caused a strong translocation of annexin A5 from cytosol into mitochondria. Confocal laser scanning microscopic analysis clearly showed that TMS induced translocation of annexin A5 into mitochondria. TMS increased the expression and oligomerization of voltage-dependent anion channel (VDAC) 1, which may promote mitochondria-dependent apoptosis through disruption of mitochondrial membrane potential. When cells were treated with TMS, the levels of Bax, and Bak as well as annexin A5 were strongly enhanced. Moreover, we found that the cytosolic release of apoptogenic factors such as cytochrome c, or apoptosis-inducing factor (AIF) in mitochondria was markedly increased. Annexin A5 depletion by siRNA led to decreased proapoptotic factors such as Bax, Bak, and annexin A5. Taken together, our results indicate that annexin A5 may play an important role in TMS-mediated mitochondrial apoptosis through the regulation of proapoptotic proteins and VDAC1 expression.

Role of HIV Vpr as a Regulator of Apoptosis and an Effector on Bystander Cells

  • Moon, Ho Suck;Yang, Joo-Sung
    • Molecules and Cells
    • /
    • v.21 no.1
    • /
    • pp.7-20
    • /
    • 2006
  • The major event in human immunodeficiency virus type 1 (HIV-1) infection is the death of many cells related to host immune response. The demise of these cells is normally explained by cell suicide mechanism, apoptosis. Interestingly, the decrease in the number of immune cells, such as non-CD4+ cells as well as CD4+ T cells, in HIV infection usually occurs in uninfected bystander cells, not in directly infected cells. It has, therefore, been suggested that several soluble factors, including viral protein R (Vpr), are released from the infected cells and induce the death of bystander cells. Some studies show that Vpr interacts directly with adenine nucleotide translocator (ANT) to induce mitochondrial membrane permeabilization (MMP). The MMP results in release of some apoptogenic factors such as cytochrome-c (cyt-c) and apoptosis-inducing factor (AIF). Vpr also has indirect effect on mitochondria through enhancing the level of caspase-9 transcription and suppressing nuclear factor-kappa B (NF-${\kappa}B$). The involvement of p53 in Vpr-induced apoptosis remains to be studied. On the other hand, low level of Vpr expression has anti-apoptotic effect, whereas it's high level of expression induces apoptosis. Extracellular Vpr also exhibits cytotoxicity to uninfected bystander cells through apoptotic or necrotic mechanism. The facts that Vpr has cytotoxic effect on both infected cells and bystander cells, and that it exhibits both proand anti-apoptotic activity may explain its role in viral survival and disease progression.

Apoptotic Effect of Co-treatment with Curcumin and Cisplatin on SCC25 Human Tongue Squamous Cell Carcinoma Cell Line

  • Sohn, Hyeon-Jin;Kim, In-Ryoung;Kim, Yong-Ho;Kim, Gyoo-Cheon;Kwak, Hyun-Ho;Park, Bong-Soo
    • International Journal of Oral Biology
    • /
    • v.39 no.3
    • /
    • pp.159-167
    • /
    • 2014
  • Curcumin is a widely used flavoring agent in food, and it has been reported to inhibit cell growth, to induce apoptosis, and to have antitumor activity in many cancers. Cisplatin is one of the most potent known anticancer agents and shows significant clinical activity against a variety of solid tumors. This study was undertaken to investigate the synergistic apoptotic effects of co-treatment with curcumin and cisplatin on human tongue SCC25 cells. To investigate whether the co-treatment efficiently reduced the viability of the SCC25 cells compared with the two treatments separately, an MTT assay was conducted. The induction and the augmentation of apoptosis were confirmed by DNA electrophoresis, Hoechst staining, and an analysis of DNA hypoploidy. Western blot, MMP and immunofluorescence tests were also performed to evaluate the expression levels and the translocation of apoptosis-related proteins following the co-treatment. In this study, following the co-treatment with curcumin and cisplatin, the SCC25 cells showed several forms of apoptotic manifestation, such as nuclear condensation, DNA fragmentation, reduction of MMP, increased levels of Bax, decreased levels of Bcl-2, and decreased DNA content. In addition, they showed a release of cytochrome c into the cytosol, translocation of AIF and DFF40 (CAD) to the nuclei, and activation of caspase-7, caspase-3, PARP, and DFF45 (ICAD). In contrast, separate treatments of $5{\mu}M$ of curcumin or $4{\mu}g/ml$ of cisplatin, for 24 hours, did not induce apoptosis. Therefore, our data suggest that combination therapy with curcumin and cisplatin could be considered as a novel therapeutic strategy for human oral squamous cell carcinoma.

Factors in Selection of Surgical Approaches for Lower Lumbar Burst Fractures (하부 요추 방출 골절의 수술방법 결정시 고려 요인들)

  • Jahng, Tae-Ahn;Kim, Jong-Moon
    • Journal of Korean Neurosurgical Society
    • /
    • v.29 no.8
    • /
    • pp.1055-1062
    • /
    • 2000
  • Objectives : Burst fracture of the lower lumbar spine(L3-L5) is rare and has some different features compare to that of thoracolumbar junction. Lower lumbar spine is flexible segments located deeply, and has physiologic lordosis. All of these contribute to making surgical approach difficult. Generally, lower lumbar burst fracture is managed either anteriorly or posteriorly with various fixation and fusion methods. But there is no general guideline or consensus regarding the proper approach for such lesion. We have tried to find out the influencing factors for selecting the surgical approach through the analysis of lower lumbar burst fractures treated for last 4 years(1994.3-1998.3). Method : This study includes 15 patients(male : 10, female : 5, age range 20-59 years with mean age of 36.7 years, L3 : 8 cases, L4 : 5 cases, L5 : 2 cases). Patients were classified into anterior(AO) and posterior operated(PO) groups. We investigated clinical findings, injured column, operation methods, and changes in follow-up radiologic study (kyphotic angle) to determine the considerable factors in selecting the surgical approaches. Results : There were 5 AO and 10 PO patients. Anterior operation were performed with AIF with Kaneda or Z-plate and posterior operation were done with pedicle screw fixation with PLIF with cages or posterolateral fusion. Canal compression was 46.6% in AO and 38.8% in PO. The degree of kyphotic angle correction were 10.7 degree(AO) and 8.5 degree(PO), respectively. There was no statistical difference between anterior and posterior operation group. All patients showed good surgical outcome without complications. Conclusion : Anterior operation provided good in kyphotic angle correction and firm anterior strut graft, but it difficulty arose in accessing the lesions below L4 vertebra. While posterior approach showed less correction of kyphotic angle, it required less time and provided better results for accompanied adjacent lesion and pathology such as epidural hematoma. The level of injury, canal compression, biomechanics, multiplicity, and pathology are considered to be important factors in selection of the surgical approach.

  • PDF

Mechanism Underlying Shikonin-induced Apoptosis and Cell Cycle Arrest on SCC25 Human Tongue Squamous Cell Carcinoma Cell Line

  • Oh, Sang-Hun;Park, Sung-Jin;Yu, Su-Bin;Kim, Yong-Ho;Kim, In-Ryoung;Park, Bong-Soo
    • International Journal of Oral Biology
    • /
    • v.40 no.1
    • /
    • pp.51-61
    • /
    • 2015
  • Shikonin, a major ingredient in the traditional Chinese herb Lithospermumerythrorhizon, exhibits multiple biological functions including antimicrobial, anti-inflammatory, and antitumor effects. It has recently been reported that shikonin displays antitumor properties in many cancers. This study was aimed to investigate whether shikonin could inhibit oral squamous carcinoma cell (OSCC) growth via mechanisms of apoptosis and cell cycle arrest. The effects of shikonin on the viability and growth of OSCC cell line, SCC25 cells were assessed by MTT assay and clonogenic assays, respectively. Hoechst staining and DNA electrophoresis indicated that the shikonin-treated SCC25 cells were undergoing apoptosis. Western blotting, immunocytochemistry, confocal microscopy, flow cytometry, MMP activity, and proteasome activity also supported the finding that shikonin induces apoptosis. Shikonin treatment of SCC25 cells resulted in a time- and dose-dependent decrease in cell viability, inhibition of cell growth, and increase in apoptotic cell death. The treated SCC25 cells showed several lines of apoptotic manifestation as follows: nuclear condensation; DNA fragmentation; reduced MMP and proteasome activity; decrease in DNA contents; release of cytochrome c into cytosol; translocation of AIF and DFF40 (CAD) onto the nuclei; a significant shift in Bax/Bcl-2 ratio; and activation of caspase-9, -7, -6, and -3, as well as PARP, lamin A/C, and DFF45 (ICAD). Shikonin treatment also resulted in down-regulation of the G1 cell cycle-related proteins and up-regulation of $p27^{KIP1}$. Taken together, our present findings demonstrate that shikonin strongly inhibits cell proliferation by modulating the expression of the G1 cell cycle-related proteins, and that it induces apoptosis via the proteasome, mitochondria, and caspase cascades in SCC25 cells.

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
    • /
    • v.35 no.2
    • /
    • 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.

Mechanism Underlying Curcumin-induced Apoptosis and Cell Cycle Arrest on SCC25 Human Tongue Squamous Cell Carcinoma Cell Line

  • Moon, Jung-Bon;Lee, Kee-Hyun;Kim, In-Ryoung;Kim, Gyoo-Cheon;Kwak, Hyun-Ho;Park, Bong-Soo
    • International Journal of Oral Biology
    • /
    • v.39 no.1
    • /
    • pp.23-33
    • /
    • 2014
  • Several studies have shown that curcumin, which is derived from the rhizomes of turmeric, possesses antimicrobial, antioxidant and anti-inflammatory properties. The antitumor properties of curcumin have also now been demonstrated more recently in different cancers. This study was undertaken to investigate the modulation of cell cycle-related proteins and the mechanisms underlying apoptosis induction by curcumin in the SCC25 human tongue squamous cell carcinoma cell line. Curcumin treatment of the SCC25 cells resulted in a time- and dose-dependent reduction in cell viability and cell growth, and onset of apoptotic cell death. The curcumin-treated SCC25 cells showed several types of apoptotic manifestations, such as nuclear condensation, DNA fragmentation, reduced MMP and proteasome activity, and a decreased DNA content. In addition, the treated SCC25 cells showed a release of cytochrome c into the cytosol, translocation of AIF and DFF40/CAD into the nuclei, a significant shift in the Bax/Bcl-2 ratio, and the activation of caspase-9, caspase-7, caspase-6, caspase-3, PARP, lamin A/C, and DFF45/ICAD. Furthermore, curcumin exposure resulted in a downregulation of G1 cell cycle-related proteins and upregulation of $p27^{KIP1}$. Taken together, our findings demonstrate that curcumin strongly inhibits cell proliferation by modulating the expression of G1 cell cycle-related proteins and inducing apoptosis via proteasomal, mitochondrial, and caspase cascades in SCC25 cells.