• Title/Summary/Keyword: 미토콘드리아 의존기전

Search Result 24, Processing Time 0.023 seconds

Involvement of apoptotic signals in cyclosporin A-induced proliferation of human gingival fibroblast (사람 치은 섬유모세포에서 Cyclosporin-A 유도 세포증식에 대한 항세포고사 기전)

  • Jeong, Tea-Sul;Chung, Hyun-Ju;Kim, Won-Jae
    • Journal of Periodontal and Implant Science
    • /
    • v.35 no.3
    • /
    • pp.731-745
    • /
    • 2005
  • Cyclosproin A(CsA)는 세포 이식거부방지를 위한 면역 억제제 및 자가 면역질환 치료제로 널리 사용되어 왔다. CsA는 매양된 사람 치은섬유아세포를 증식시킴이 알려져 있지만 CsA에 의한 세포증식기전에 대한 세포사멸기전 및 Bcl-2의 역할은 연구되어 있지 않다. 이번 연구는 사람 섬유아세포에서 CsA에 의한 세포증삭기전에 세포고사기전 및 Bcl-2 family가 관여하는지 밝히는 데에 목적이 있다. 세포 생장력은 MTT 방법으로 측정하였다. Bcl-2 family와 Fas 발현 정도는 RT-PCR 방법이나 western blot으로 확인하였다. Caspase-3 및 -9의 활성은 ELISER reader로, reactive oxygen species(ROS)는 fluorescence spectrometer에 의해 측정되었다. 미토콘드리아에서 세포질로 분비된 cytochrome c는 Western blot으로 조사하였다. CsA는 $0.1{\sim}10\;{\mu}M$에서 사람 섬유아세포의 생존률을 시간과 농도 의존적으로 증가시켰으며, 50 ${\mu}M$ CsA에서는 오히려 세포가 죽였다. 또한, CsA 처리로 미토콘드리아에서 세포질로 유리되는 cytochrome c 양과 VDAC 1 및 3 발현량이 감소되었고, caspase-9과 caspase-3의 활성도도 감소되었다. 한편, CsA 처리한 섬유아세포에서 death receptor 구성요소인 Fas 발현이 감소되었다. Bcl-2 family에 대한 RT-PCR, western blot 분석결과, 세포고사를 억제하는 Bel-2 발현은 증가되었으나 세포고사를 자극하는 Bax와 Bid의 발현은 감소되었다. 이러한 결과들은 사람 섬유아세포에서 CsA유도 세포증식에 Bcl-2 family와 ROS가 매개하는 미토콘드리아 의존 및 death receptor 의존 세포고사기전이 관여함을 시사하였다.

Effects of epigallocatechin gallate on $CoCl_2-induced$ apoptosis in PC12 cells (PC12 세포에서 $CoCl_2$ 유발 세포자멸사에 대한 epigallocatechin-gallate의 역할)

  • Mo, Hyun-Chul;Choi, Nam-Ki;Kim, Seon-Mi;Kim, Won-Jae;Yang, Kyu-Ho
    • Journal of the korean academy of Pediatric Dentistry
    • /
    • v.33 no.1
    • /
    • pp.13-24
    • /
    • 2006
  • Neuronal apoptotic events, consequently resulting in neuronal cell death, are occurred in hypoxic/ischemic condition. This cell death has been shown to be accompanied with the production of reactive oxygen species (ROS), which can attack cellular components such as nucleic acids, proteins and phospholipid. However, the underlying mechanisms of apoptosis induced in hypoxic/ischemic condition and its treatment methods are unsettled. Cobalt chloride $(CoCl_2)$ has been known to mimic hypoxic condition including the production of ROS. Epigallocatechin gallate (EGCG), a green tea polyphenol, has diverse pharmacologial activities in cell growth and death. This study was aimed to investigate the apoptotic mechanism by $CoCL_2$ and effects of EGCG on $CoCl_2-induced$ apoptosis in PC12 cells. Administration of $CoCl_2$ decreased cell survival in dose- and time-dependent manners and induced genomic DNA fragmentation. Treatment with $100{\mu}M$ EGCG for 30 min before PC12 cells were exposed to $150{\mu}M$ $CoCl_2$, being resulted in the cell viability and DNA fragmentation being rescued. $CoCl_2$ caused morphologic changes such as cell swelling and condensed nuclei whereas EGCG attenuated morphologic changes by $CoCl_2$. EGCG suppressed the apoptotic peak and a loss of ${\Delta}{\psi}_m$ induced by $CoCl_2$. $CoCl_2$ decreased Bcl-2 expression but Bax expression was not changed in $CoCl_2$- treated cells. EGCG attenuated the Bcl-2 underexpression by $CoCl_2$. $CoCl_2$ augumented the cytochrome c release from mitochondria into cytoplasm and increased caspase-8, -9 and caspase-3 activity a marker of the apoptotic executing stage. EGCG ameliorated the incruement in caspase-8, -9 and -3 activity, and cytochrome c release by $CoCl_2$ NAC (N-acetyl-cysteine), a scavenger of ROS, attenuated $CoCl_2-induced$ apoptosis in consistent with those of EGCG. These results suggest that $CoCl_2$ induces apoptotic cell death through both mitochondria- and death receptor-dependent pathway and EGCG has neuroprotective effects against $CoCl_2-induced$ apoptosis in PC12 cells.

  • PDF

A study on the mechanism of NO-induced apoptosis in human gingival fibroblast (사람 치은 섬유아세포에서 산화질소 유도 세포고사에 대한 연구)

  • Kim, Kang-Moon;Chung, Hyun-Ju;Kim, Won-Jae
    • Journal of Periodontal and Implant Science
    • /
    • v.34 no.4
    • /
    • pp.807-818
    • /
    • 2004
  • 산화질소는 생리적 농도에서 세포내 신호전달자로 작용하지만 높은 농도에서는 세포독성을 일으킨다. 최근 치은 섬유아세포와 치주인대 섬유아세포는 산화질소 합성효소를 가지고 있고 세균의 lipopolysaccharide나 cytokine에 의해 대량의 높은 농도의 산화질소가 합성된다는 보고가 있음에도 지금까지 치은 조직에서 산화질소의 세포독성에 대한 연구는 아직 이루어 지지않고 있다. 본 연구는 사람의 치은 섬유아세포에서, 산화질소유도세포 고사기전을 밝히는데 목적이 있다. 세포 생장력은 MTT 방법으로 측정하였고, 세포의 형태적 변화는 Diff-Quick 염색법으로 조사하였다. Bcl-2 famly와 Fas 발현 정도는 RT-PCR 방법에 의해 확인하였으며, caspase-3, -8 와 -9의 활성은 spectrophotometer로 reactive oxygen species (ROS)는 형광분광계에 의해 측정되었다. 미토콘드리아에서 세포질로 분비된 cytochrome c는 western blot으로 조사하였다. 산화질소 유리제인 sodium nitroprusside (SNP) 처리는 사람 섬유아세포의 생존률을 시간과 농도 의존적으로 감소시켰고, 세포용적축소, 염색사 용축, DNA 절편화를 일으켰다. 또한, SNP 처리로 미토콘드리아에서 세포질로 유리되는 cytochrome c 양이 증가되었고, caspase-9 과 caspase-3 의 활성이 증가되었다. 한편, SNP 처리에 의해 death receptor 구성요소인 Fas 발현이 증가되었고, caspase-8의 활성이 증가되었다. Bcl-2 family 에 대한 RT-PCR 분석결과, 세포고사를 억제하는 Bcl-2 발현은 감소되었으나 세포고사를 자극하는 Bax와 Bid의 발현은 증가되었다. Soluble guanylate cyclase 억제제인 ODQ는 SNP에 의한 세포 생존율 감소를 차단하지 못했다. 따라서, 본 실험의 결과들은 사람 섬유아세포에서 산화질소유도 세포고사에 Bcl-2 family나 ROS가 매개하는 미토콘드리아 의존 및 death receptor 의존 세포고사기전이 관여함을 시사하였다.

FUN14 Domain-Containing Protein 1 Is Involved in Amyloid Beta Peptide-Induced Mitochondrial Dysfunction and Cell Injury in HT-22 Neuronal Cells (HT-22 신경세포에서 아밀로이드 베타 펩티드에 의한 미토콘드리아와 세포 손상 기전에서 FUN14 도메인 함유 단백 1의 역할)

  • Jae Hoon Kang;Jae Suk Woo
    • Journal of Life Science
    • /
    • v.34 no.1
    • /
    • pp.37-47
    • /
    • 2024
  • FUN14 domain-containing protein 1 (FUNDC1), an outer mitochondrial membrane protein, contributes to removal of damaged mitochondria through mitophagy. In this study, to elucidate the role of the FUNDC1 in the amyloid beta peptide (Aβ)-induced neuropathy, changes in the degree of mitochondrial dysfunction and cell injury caused by Aβ treatment were examined in the HT-22 neuronal cells in which the FUNDC1 expression was transiently silenced or overexpressed. We found that Aβ treatment causes a time-dependent decrease of the FUNDC1 expression. In the Aβ-treated cells, there were a drop in MTT reduction ability, depletion of cellular ATP, disruption of mitochondrial membrane potential, stimulation of cellular ROS production, and increased mitochondrial Ca2+ load. Activation of caspase-3 and induction of apoptotic cell death were also observed. Transient silencing of the FUNDC1 expression by transfection with the FUNDC1 small interfering RNA per se caused mitochondrial dysfunction and apoptotic cell death like the effect of Aβ treatment. Conversely, in cells in which the FUNDC1 was transiently overexpressed by FUNDC1-Myc transfection, overexpression itself had no effect on the mitochondrial functional integrity and cell survival but showed a significant prevention effect against mitochondrial and cell injury caused by Aβ treatment. Overall, these results suggest that the FUNDC1 is importantly involved in the Aβ-induced mitochondrial dysfunction and cell injury in the HT-22 neuronal cells.

4-Hydroxynonenal Induces Endothelial Apoptosis through Mitochondrial Depolarization (4-Hydroxynonenal의 미토콘드리아 탈분극을 통한 혈관 내피 세포의 세포사 유도기전)

  • Kang, Dae-Yeon;Lee, Ji-Young;Kim, Min-Sun;Kim, Chul-Hong;Kim, Hyung-Keun;Lee, Sun-Mi;Kwon, Young-Mi;Lee, Jae-won;Baik, Hyung-Suk;Yu, Byung-Pal;Chung, Hae-Young
    • Journal of Life Science
    • /
    • v.18 no.11
    • /
    • pp.1513-1520
    • /
    • 2008
  • The 4-Hydroxynonenal (HNE) affects vascular dysfunctions probably through the interruption of the cellular redox balance. To better understand vascular abnormalities resulting from the accumulation of HNE, we delineated mechanism by which mitochondrial apoptosis occurs in the YPEN-1 endothelial cells. HNE treatment led to the loss of mitochondrial membrane potential (${\delta}{\Psi}_m$), resulting in the release of cytochrome c. Data showed decreased Bcl-2 and increased Bax protein levels in HNE-treated cells. NAC, a reactive oxygen species (ROS) scavenger, and penicillamine, the peroxynitrite scavenger, blocked HNE-mediated ROS generation, thereby thwarting the cytochrome c release and apoptosis. The treatment of the cells with zVAD-fmk, a broad range caspase inhibitor did not suppress HNE-induced apoptosis, suggesting that the apoptosis might be the possibility of caspase-independent process. Our findings delineate the underlying mechanism of the HNE induced endothelial apoptosis by triggering depolarization of mitochondria membrane potential that can lead to the deterioration of vasculature homeostasis and subsequent vascular dysfunction with aging.

Cell Death Induction Mechanism of Non-small Cell Lung Cancer Cell Line, NCI-H1703 by Docetaxel (Docetaxel에 의한 비소세포폐암세포주 NCI-H1703의 세포사멸 유도기전)

  • Ha Hyeon-Cheol;Song Seung-Hwan;Park Chin-Su;Kim Jong-Won;Kim Yeong-Dae
    • Journal of Chest Surgery
    • /
    • v.39 no.9 s.266
    • /
    • pp.668-673
    • /
    • 2006
  • Background: Docetaxel has been effectively used as an anti-cancer chemotherapuetic agent for various tumor treatments including lung cancer. However, the cell death induction mechanism(s) involved with docetaxel treatment in lung cancer cells has not been known yet. Material and Method: In the present study, the cellular and biochemical changes of NCI-H1703 cells (non-small cell lung cancer cell line, p53-mutant) after docetaxel treatment have been monitored by flow cytometry, fluorescence microscopy and western blot. Result: Docetaxel treatment significantly resulted in decrease of S phase as well as increase of G2 phase, and consequently evoked an increase of cell death in NCI-H1703 cells. After docetaxel exposure the activations of caspase-3 and caspase-9 were detected. Conclusion: Take together, it is suggested that the docetaxel induces NCI-H1703 cell death by caspase-9 and caspase-3 dependent mitochondrial apoptotic pathway.

Combination Treatment with Arsenic Trioxide and Sulindac Induces Apoptosis of NCI-H157 Human Lung Carcinoma Cells via ROS Generation with Mitochondrial Dysfunction (NCI-H157 폐암 세포주에서 활성산소종의 생성과 미토콘드리아 기능변화를 한 Arsenic Trioxide와 Sulindac 병합요법의 세포고사효과)

  • Kim, Hak-Ryul;Yang, Sei-Hoon;Jeong, Eun-Taik
    • Tuberculosis and Respiratory Diseases
    • /
    • v.59 no.1
    • /
    • pp.30-38
    • /
    • 2005
  • Background : Arsenic trioxide ($As_2O_3$) has been used to treat acute promyelocytic leukemia, and it induces apoptosis in a variety of solid tumor cell lines including non-small cell lung cancer cells. However, nonsteroidal antiinflammatory drugs (NSAID) can enhance tumor response to chemotherapeutic drugs or radiation. It was previously demonstrated that a combination treatment with $As_2O_3$ and sulindac induces the apoptosis of NCI-H157 human lung carcinoma cells by activating the caspase cascade. This study aimed to determine if a combination treatment augmented its apoptotic potential through other pathways except for the activation of the caspase cascade. Material and Methods : The NCI-H157 cells were treated with $As_2O_3$, sulindac and antioxidants such as glutathione (GSH) and N-acetylcysteine (NAC). The cell viability was measured by a MTT assay, and the level of intracellular hydrogen peroxide ($H_2O_2$) generation was monitored fluorimetrically using a scopoletin-horse radish peroxidase (HRP) assay. Western blotting and mitochondrial membrane potential transition analysis were performed in order to define the mechanical basis of apoptosis. Results : The viability of the cells was decreased by a combination treatment of $As_2O_3$ and sulindac, and the cells were protected using antioxidants in a dose-dependent manner. The increased $H_2O_2$ generation by the combination treatment was inhibited by antioxidants. The combination treatment induced changes in the mitochondrial transmembrane potential as well as the expression of the Bcl-2 family proteins, and increased cytochrome c release into the cytosol. However, the antioxidants inhibited the effects of the combination treatment. Conclusion : Combination treatment with $As_2O_3$ and sulindac induces apoptosis in NCI-H157 human lung carcinoma cells via ROS generation with a mitochondrial dysfunction.

Microtubule-damaging Chemotherapeutic Agent-mediated Mitotic Arrest and Apoptosis Induction in Tumor Cells (미세소관-손상 항암제 처리에 의한 세포주기의 정지 및 에폽토시스 유도)

  • Jun, Do Youn;Kim, Young Ho
    • Journal of Life Science
    • /
    • v.26 no.3
    • /
    • pp.376-386
    • /
    • 2016
  • Apoptosis induction has been proposed as an efficient mechanism by which malignant tumor cells can be removed following chemotherapy. The intrinsic mitochondria-dependent apoptotic pathway is frequently implicated in chemotherapy-induced tumor cell apoptosis. Since DNA-damaging agent (DDA)-induced apoptosis is mainly regulated by the tumor suppressor protein p53, and since more than half of clinical cancers possess inactive p53 mutants, microtubule-damaging agents (MDAs), of which apoptotic effect is mainly exerted via p53-independent routes, can be promising choice for cancer chemotherapy. Recently, we found that the apoptotic signaling pathway induced by MDAs (nocodazole, 17α-estradiol, or 2-methoxyestradiol) commonly proceeded through mitotic spindle defect-mediated prometaphase arrest, prolonged Cdk1 activation, and subsequent phosphorylation of Bcl-2, Mcl-1, and Bim in human acute leukemia Jurkat T cells. These microtubule damage-mediated alterations could render the cellular context susceptible to the onset of mitochondria-dependent apoptosis by triggering Bak activation, Δψm loss, and resultant caspase cascade activation. In contrast, when the MDA-induced Bak activation was inhibited by overexpression of anti-apoptotic Bcl-2 family proteins (Bcl-2 or Bcl-xL), the cells in prometaphase arrest failed to induce apoptosis, and instead underwent mitotic slippage and endoreduplication cycle, leading to formation of populations with 8N and 16N DNA content. These data indicate that cellular apoptogenic mechanism is critical for preventing polyploid formation following MDA treatment. Since the formation of polyploid cells, which are genetically unstable, may cause acquisition of therapy resistance and disease relapse, there is a growing interest in developing new combination chemotherapies to prevent polyploidization in tumors after MDA treatment.

Mechanisms Underlying the Inhibitory Effect of GS 283 in Various Smooth Muscles (GS 283의 평활근 억제 작용기전)

  • Kim, Si-Hwan;Lee, Young-Soo;Chong, Won-Seog;Chang, Ki-Churl
    • The Korean Journal of Pharmacology
    • /
    • v.30 no.1
    • /
    • pp.101-109
    • /
    • 1994
  • Pharmacological characterization of GS 283, a tetrahydroisoquinoline derivative has been elucidated using rat thoracic aorta, guinea pig tenea coli and rabbit mesentery artery in vitro. GS 283 showed calcium antagonistic action in vascular smooth muscle, since high $K^+-induced$ contraction was concentration dependently inhibited. GS 283 also inhibited the contraction induced by ${\alpha}_1$ receptor activation. Vasodilating action of GS 283 was not modified by the propranolol, indicating that GS 283 has no ${\beta}$ receptor stimulatory action. Simultaneous measurement of intracellular calcium change and muscle tension indicated that the inhibitory effect of GS 283 was accompanied by the increase in tissue fluorescence. This increment was not due to fura 2 fluorescence but to endogenous pyridine nucleotide, suggesting that GS 283 has an effect to inhibit mitochondrial function. GS 283 had an inhibitory action on cyclic AMP and GMP-dependent phosphodiesterases from rat brain with Ki values of 2.5 and 6.7 mM. From these findings we concluded that GS 283 has multiple action such as the inhibition of cyclic nucleotide-dependent phosphodiesterases, blocking of calcium channel as well as inhibition of mitochondrial function which are responsible for vasodilatation.

  • PDF

Resveratrol Induces Apoptosis in Primary Human Prostate Cancer Cells (Primary 인체 전립선 암세포에서 Resveratrol의 Apoptosis 유도 효과)

  • Kang, Hye-In;Kim, Jae-Yong;Cho, Hyun-Dong;Park, Kyung-Wuk;Kang, Jum-Soon;Seo, Kwon-Il
    • Journal of the Korean Society of Food Science and Nutrition
    • /
    • v.39 no.8
    • /
    • pp.1119-1125
    • /
    • 2010
  • To evaluate resveratrol as a prostate cancer preventive material, we investigated its anti-proliferative and apoptotic effects in RC-58T/h/SA#4 primary human prostate cancer cells. Resveratrol significantly decreased the number of viable RC-58T/h/SA#4 cells in a dose- and time-dependent manner. Resveratrol showed cytotoxicity against RC-58T/h/SA#4, LNCaP, PC-3 human prostate cancer cells with $IC_{50}$ values of 245, 320 and $340\;{\mu}M$, respectively. However the cytotoxic potential of resveratrol against normal RWPE-1 cells was lower ($IC_{50}=982\;{\mu}M$). Resveratrol induced cell death as evidenced by the increased formation of apoptotic bodies, nuclear condensation, sub-G1 phase, and DNA fragmentation. Resveratrol activated initiator caspases 8, and 9 as well as effector caspase 3 in a dose-dependent manner. Furthermore, the general caspase inhibitor z-VAD-fmk significantly inhibited resveratrol-induced apoptosis compared to cells without treatment. These results clearly indicate that resveratrol-induced apoptosis was dependent on caspase activation. Further, resveratrol modulated the down regulation of Bcl-2 (anti-apoptotic), and Bid. However, the level of Bax (pro-apoptotic) remained unchanged. These results suggest that resveratrol induced apoptosis in RC-58T/h/SA#4 cells via a mitochondrial-mediated caspase-dependent pathway, suggesting therapeutic potential against prostate cancer.