• 제목/요약/키워드: autophagy

검색결과 493건 처리시간 0.024초

LncRNA PART1 Attenuates Myocardial Ischemia-Reperfusion Injury by Regulating TFAP2C/DUSP5 Axis via miR-302a-3p

  • Min Zeng;Xin Wei;Jinchao Zhou;Siqi Luo
    • Korean Circulation Journal
    • /
    • 제54권5호
    • /
    • pp.233-252
    • /
    • 2024
  • Background and Objectives: Myocardial ischemia-reperfusion injury (MIRI) refers to the damage of cardiac function caused by restoration of blood flow perfusion in ischemic myocardium. However, long non-coding RNA prostate androgen regulated transcript 1 (PART1)'s role in MIRI remain unclear. Methods: Immunofluorescence detected LC3 expression. Intermolecular relationships were verified by dual luciferase reporter assay. 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide, flow cytometry and transferase-mediated dUTP nick-end labeling (TUNEL) assays analyzed cell viability and apoptosis. The release of lactate dehydrogenase was tested via enzyme-linked immunosorbent assay (ELISA). Left anterior descending coronary artery surgery induced a MIRI mouse model. Infarct area was detected by 2,3,5-triphenyltetrazolium chloride staining. Hematoxylin and eosin staining examined myocardial injury. ELISA evaluated myocardial marker (creatine kinase MB) level. Results: PART1 was decreased in hypoxia/reoxygenation (H/R) induced AC16 cells and MIRI mice. PART1 upregulation attenuated the increased levels of Bax, beclin-1 and the ratio of LC3II/I, and enhanced the decrease of Bcl-2 and p62 expression in H/R-treated cells. PART1 upregulation alleviated H/R-triggered autophagy and apoptosis via miR-302a-3p. Mechanically, PART1 targeted miR-302a-3p to upregulate transcription factor activating enhancer-binding protein 2C (TFAP2C). TFAP2C silencing reversed the protected effects of miR-302a-3p inhibitor on H/R treated AC16 cells. We further established TFAP2C combined to dual-specificity phosphatase 5 (DUSP5) promoter and activated DUSP5. TFAP2C upregulation suppressed H/R-stimulated autophagy and apoptosis through upregulating DUSP5. Overexpressed PART1 reduced myocardial infarction area and attenuated MIRI in mice. Conclusion: PART1 improved the autophagy and apoptosis in H/R-exposed AC16 cells through miR-302a-3p/TFAP2C/DUSP5 axis, which might provide novel targets for MIRI treatment.

Involvement of ROS in Curcumin-induced Autophagic Cell Death

  • Lee, Youn-Ju;Kim, Nam-Yi;Suh, Young-Ah;Lee, Chu-Hee
    • The Korean Journal of Physiology and Pharmacology
    • /
    • 제15권1호
    • /
    • pp.1-7
    • /
    • 2011
  • Many anticancer agents as well as ionizing radiation have been shown to induce autophagy which is originally described as a protein recycling process and recently reported to play a crucial role in various disorders. In HCT116 human colon cancer cells, we found that curcumin, a polyphenolic phytochemical extracted from the plant Curcuma longa, markedly induced the conversion of microtubule-associated protein 1 light chain 3 (LC3)-I to LC3-II and degradation of sequestome-1 (SQSTM1) which is a marker of autophagosome degradation. Moreover, we found that curcumin caused GFP-LC3 formation puncta, a marker of autophagosome, and decrease of GFP-LC3 and SQSTM1 protein level in GFP-LC3 expressing HCT116 cells. It was further confirmed that treatment of cells with hydrogen peroxide induced increase of LC3 conversion and decrease of GFP-LC3 and SQSTM1 levels, but these changes by curcumin were almost completely blocked in the presence of antioxidant, N-acetylcystein (NAC), indicating that curcumin leads to reactive oxygen species (ROS) production, which results in autophagosome development and autolysosomal degradation. In parallel with NAC, SQSTM1 degradation was also diminished by bafilomycin A, a potent inhibitor of autophagosome-lysosome fusion, and cell viability assay was further confirmed that cucurmin-induced cell death was partially blocked by bafilomycin A as well as NAC. We also observed that NAC abolished curcumin-induced activation of extracelluar signal-regulated kinases (ERK) 112 and p38 mitogen-activated protein kinases (MAPK), but not Jun N-terminal kinase (JNK). However, the activation of ERK1/2 and p38 MAPK seemed to have no effect on the curcumin-induced autophagy, since both the conversion of LC3 protein and SQSTM1 degradation by curcumin was not changed in the presence of NAC. Taken together, our data suggest that curcumin induced ROS production, which resulted in autophagic activation and concomitant cell death in HCT116 human colon cancer cell. However, ROS-dependent activation of ERK1/2 and p38 MAPK, but not JNK, might not be involved in the curcumin-induced autophagy.

비스테로이드소염제(Nonsteroidal Anti-inflammatory Drug, NSAID)에 의한 인간 암세포의 imatinib 및 TRAIL의 세포 독성 증강 기전 연구 (Potentiation of the Cytotoxic Effects of Imatinib and TRAIL by Nonsteroidal Anti-inflammatory Drugs on Human Cancer Cells)

  • 문현정;강치덕;김선희
    • 생명과학회지
    • /
    • 제30권8호
    • /
    • pp.661-671
    • /
    • 2020
  • 항암 요법의 실패의 주요 원인으로 암세포의 항암제에 대한 내성 획득이 잘 알려져 있다. 비스테로이드소염제(NSAID)는 항염증작용뿐만 아니라 항암제와의 병용요법으로 임상적인 암 치료 요법에 응용되고있다. 본 연구에서는 NSAIDs 인 celecoxib 및 이의 구조 유사체인 2,5-dimethyl celecoxib 그리고 ibuprofen의 인간 암세포에 대한 imatinib 및 TNF-related apoptosis inducing ligand (TRAIL) 세포 독성 변화에 미치는 영향을 조사하였다. NSAID는 TRAIL 및 imatinib에 각각 약제 내성을 나타내는 간암 세포와 백혈병 세포에서 이들 약물의 세포독성을 증강시키는 활성을 나타내었다. NSAID는 ATF4/CHOP의 발현 증강으로 소포체 스트레스 및 오토파지(Autophagy, 자가포식)를 유도하였다. 이로 인한 DR5 발현 증강과 함께 c-FLIP 발현 억제로 TRAIL의 세포독성을 증강시키는 기전을 나타내었다. NSAID로 유도되는 오토파지 활성은 imatinib-resistant CD44highK562 백혈병세포의 imatinib 감수성을 증강시켰으며, NSAID는 이 세포에서 높은 발현을 나타내는 다양한 stemness-related marker 단백질의 발현 감소를 촉진시키는 활성으로 세포사멸을 유도하는 것을 알 수 있었다. 이러한 결과는 NSAID의 오토파지 유도 활성이 TRAIL과 imatinib의 세포 독성을 증강시키는 것으로서, NSAID와 이들 약물과 병용 처리방법은 인간 암세포의 TRAIL 및 imatinib 내성을 극복 시킴과 동시에 암세포에 이들 약물의 독성 부작용을 감소시킬 수 있는 낮은 농도의 처리를 가능하게 할 것으로 사료된다.

생쥐 근육세포에서 코티졸이 세포질세망 스트레스, 자연 세포사멸과 자가포식에 미치는 영향 (Effects of Cortisol on Endoplasmic Reticulum-stress, Apoptosis, and Autophagy in Mouse Muscle C2C12 Cells)

  • 신동현;김경환;이지현;조병욱
    • 생명과학회지
    • /
    • 제28권10호
    • /
    • pp.1127-1131
    • /
    • 2018
  • 운동 후 분비되는 스트레스 호르몬인 cortisol을 통한 근육세포에 미치는 운동 스트레스의 재현과 coritisol 처리 농도에 따른 세포사멸, 세포질세망 스트레스 및 자가포식현상과의 관련성을 검증 하였다. 마우스 근육 세포주 C2C12를 배양하여 다양한 농도의 cortisol을 12시간 처리하여 세포의 형태 변화를 관찰하고, 세포사멸 마커인 IER3의 발현을 세포면역화학법을 이용하여 확인하였다. 또한 ER-stress와 자가포식 현상의 유도 여부를 확인하기 위하여 BiP와 LC3-I/LC3-II 항체를 이용하여 웨스턴 블랏법을 통해 검증 하였다. 그 결과 cortisol의 농도가 $50{\mu}g/ml$$100{\mu}g/ml$로 증가함에 따라 IER3와 BiP 및 LC3-II의 발현량도 유의적으로 증가함을 확인 할 수 있었다. 이러한 결과는 운동 스트레스 호르몬인 cortisol이 운동 후 근육세포의 세포사멸, 세포질세망 스트레스 및 자가포식에 영향을 미침을 보여준다. 본 연구결과는 호르몬과 근육세포 간의 관련성 연구에 기여할 것으로 기대된다.

Effects of propofol-induced autophagy against oxidative stress in human osteoblasts

  • Kim, Eun-Jung;Choi, In-Seok;Yoon, Ji-Young;Park, Bong-Soo;Yoon, Ji-Uk;Kim, Cheul-Hong
    • Journal of Dental Anesthesia and Pain Medicine
    • /
    • 제16권1호
    • /
    • pp.39-47
    • /
    • 2016
  • Background: Oxidative stress occurs during the aging process and other conditions such as bone fracture, bone diseases, and osteoporosis, but the role of oxidative stress in bone remodeling is unknown. Propofol exerts antioxidant effects, but the mechanisms of propofol preconditioning on oxidative stress have not been fully explained. Therefore, the aim of this study was to evaluate the protective effects of propofol against $H_2O_2$-induced oxidative stress on a human fetal osteoblast (hFOB) cell line via activation of autophagy. Methods: Cells were randomly divided into the following groups: control cells were incubated in normoxia (5% $CO_2$, 21% $O_2$, and 74% $N_2$) without propofol. Hydrogen peroxide ($H_2O_2$) group cells were exposed to $H_2O_2\;(200{\mu}M)$ for 2 h, propofol preconditioning (PPC)/$H_2O_2$ group cells were pretreated with propofol then exposed to $H_2O_2$, 3-methyladenine (3-MA)/PPC/$H_2O_2$ cells were pretreated with 3-MA (1 mM) and propofol, then were exposed to $H_2O_2$. Cell viability and apoptosis were evaluated. Osteoblast maturation was determined by assaying bone nodular mineralization. Expression levels of bone related proteins were determined by western blot. Results: Cell viability and bone nodular mineralization were decreased significantly by $H_2O_2$, and this effect was rescued by propofol preconditioning. Propofol preconditioning effectively decreased $H_2O_2$-induced hFOB cell apoptosis. However, pretreatment with 3-MA inhibited the protective effect of propofol. In western blot analysis, propofol preconditioning increased protein levels of collagen type I, BMP-2, osterix, and TGF-${\beta}1$. Conclusions: This study suggests that propofol preconditioning has a protective effect on $H_2O_2$-induced hFOB cell death, which is mediated by autophagy activation.

20(S)-Ginsenoside Rh2 displays efficacy against T-cell acute lymphoblastic leukemia through the PI3K/Akt/mTOR signal pathway

  • Xia, Ting;Zhang, Jin;Zhou, Chuanxin;Li, Yu;Duan, Wenhui;Zhang, Bo;Wang, Min;Fang, Jianpei
    • Journal of Ginseng Research
    • /
    • 제44권5호
    • /
    • pp.725-737
    • /
    • 2020
  • Background: T-cell acute lymphoblastic leukemia (T-ALL) is a kind of aggressive hematological cancer, and the PI3K/Akt/mTOR signaling pathway is activated in most patients with T-ALL and responsible for poor prognosis. 20(S)-Ginsenoside Rh2 (20(S)-GRh2) is a major active compound extracted from ginseng, which exhibits anti-cancer effects. However, the underlying anticancer mechanisms of 20(S)-GRh2 targeting the PI3K/Akt/mTOR pathway in T-ALL have not been explored. Methods: Cell growth and cell cycle were determined to investigate the effect of 20(S)-GRh2 on ALL cells. PI3K/Akt/mTOR pathway-related proteins were detected in 20(S)-GRh2-treated Jurkat cells by immunoblotting. Antitumor effect of 20(S)-GRh2 against T-ALL was investigated in xenograft mice. The mechanisms of 20(S)-GRh2 against T-ALL were examined by cell proliferation, apoptosis, and autophagy. Results: In the present study, the results showed that 20(S)-GRh2 decreased cell growth and arrested cell cycle at the G1 phase in ALL cells. 20(S)-GRh2 induced apoptosis through enhancing reactive oxygen species generation and upregulating apoptosis-related proteins. 20(S)-GRh2 significantly elevated the levels of pEGFP-LC3 and autophagy-related proteins in Jurkat cells. Furthermore, the PI3K/Akt/mTOR signaling pathway was effectively blocked by 20(S)-GRh2. 20(S)-GRh2 suppressed cell proliferation and promoted apoptosis and autophagy by suppressing the PI3K/Akt/mTOR pathway in Jurkat cells. Finally, 20(S)-GRh2 alleviated symptoms of leukemia and reduced the number of white blood cells and CD3 staining in the spleen of xenograft mice, indicating antitumor effects against T-ALL in vivo. Conclusion: These findings indicate that 20(S)-GRh2 exhibits beneficial effects against T-ALL through the PI3K/Akt/mTOR pathway and could be a natural product of novel target for T-ALL therapy.

ATG5 Expression Induced by MDMA (Ecstasy), Interferes with Neuronal Differentiation of Neuroblastoma Cells

  • Chae, Myounghee;Rhee, Gyu-Seek;Jang, Ik-Soon;Kim, Kwangsoo;Lee, Ji-Hae;Lee, Seung-Yeul;Kim, Minjung;Yang, Junyoung;Park, Junsoo;Lee, Seung-Hoon
    • Molecules and Cells
    • /
    • 제27권5호
    • /
    • pp.571-575
    • /
    • 2009
  • The amphetamine derivative 3, 4-methylenedioxymethamphetamine (MDMA) has become a popular recreational drug, and has also been shown to cause serotonergic neurotoxicity. This report shows that MDMA impairs brain development in a whole mouse embryo culture. The results of quantitative real-time PCR analysis showed that autophagy-related protein 5 (Atg5) expression is elevated in mouse embryo and neuroblastoma cells after MDMA treatment. This elevated Atg5 expression interferes with the neuronal differentiation of neuroblastoma cells such as SH-SY5Y and PC12 cells. Thus, our results suggest that the use of MDMA during pregnancy may impair neuronal development via an induction of Atg5 expression.

Fyn Kinase: A Potential Therapeutic Target in Acute Kidney Injury

  • Uddin, Md Jamal;Dorotea, Debra;Pak, Eun Seon;Ha, Hunjoo
    • Biomolecules & Therapeutics
    • /
    • 제28권3호
    • /
    • pp.213-221
    • /
    • 2020
  • Acute kidney injury (AKI) is a common disease with a complex pathophysiology which significantly contributes to the development of chronic kidney disease and end stage kidney failure. Preventing AKI can consequently reduce mortality, morbidity, and healthcare burden. However, there are no effective drugs in use for either prevention or treatment of AKI. Developing therapeutic agents with pleiotropic effects covering multiple pathophysiological pathways are likely to be more effective in attenuating AKI. Fyn, a non-receptor tyrosine kinase, has been acknowledged to integrate multiple injurious stimuli in the kidney. Limited studies have shown increased Fyn transcription level and activation under experimental AKI. Activated Fyn kinase propagates various downstream signaling pathways associated to the progression of AKI, such as oxidative stress, inflammation, endoplasmic reticulum stress, as well as autophagy dysfunction. The versatility of Fyn kinase in mediating various pathophysiological pathways suggests that its inhibition can be a potential strategy in attenuating AKI.

Recently Emerging Signaling Landscape of Ataxia-Telangiectasia Mutated (ATM) Kinase

  • Farooqi, Ammad Ahmad;Attar, Rukset;Arslan, Belkis Atasever;Romero, Mirna Azalea;ul Haq, Muhammad Fahim;Qadir, Muhammad Imran
    • Asian Pacific Journal of Cancer Prevention
    • /
    • 제15권16호
    • /
    • pp.6485-6488
    • /
    • 2014
  • Research over the years has progressively and sequentially provided near complete resolution of regulators of the DNA repair pathways which are so important for cancer prevention. Ataxia-telangiectasia mutated kinase (ATM), a high-molecular-weight PI3K-family kinase has emerged as a master regulator of DNA damage signaling and extensive cross-talk between ATM and downstream proteins forms an interlaced signaling network. There is rapidly growing scientific evidence emphasizing newly emerging paradigms in ATM biology. In this review, we provide latest information regarding how oxidative stress induced activation of ATM can be utilized as a therapeutic target in different cancer cell lines and in xenografted mice. Moreover, crosstalk between autophagy and ATM is also discussed with focus on how autophagy inhibition induces apoptosis in cancer cells.

Terpinen-4-ol Induces Autophagic and Apoptotic Cell Death in Human Leukemic HL-60 Cells

  • Banjerdpongchai, Ratana;Khaw-on, Patompong
    • Asian Pacific Journal of Cancer Prevention
    • /
    • 제14권12호
    • /
    • pp.7537-7542
    • /
    • 2013
  • Background: Terpinen-4-ol, a monoterpene, is found as the main component of essential oil extracts from many plants. In this study apoptotic and autophagic types of cell death induced by terpinen-4-ol and associated mechanisms were investigated in human leukemic HL-60 cells. Materials and Methods: The cytotoxicity of human leukemic U937 and HL-60 cells was determined by MTT assay. Cytochrome c release, expression of Bax, Bcl-2, Bcl-xl and cleaved Bid were determined by Western blotting. Cell morphology was examined under a transmission electron microscope. LC3-I/II, ATG5 and Beclin-1 levels were detected by immunoblotting. Results: Terpinen-4-ol exhibited cytotoxicity to human leukemic HL-60 but not U937 cells. The apoptotic response to terpinen-4-ol in HL-60 cells was due to induction of cytochrome c release from mitochondria and cleavage of Bid protein after the stimulation of caspase-8. There was a slightly decrease of Bcl-xl protein level. The characteristic cell morphology of autophagic cell death was demonstrated with multiple autophagosomes in the cytoplasm. At the molecular level, the results from Western blot analysis showed that terpinen-4-ol significantly induced accumulation of LC3-I/II, ATG5 and Beclin-1, regulatory proteins required for autophagy in mammalian cells. Conclusions: Terpinen-4-ol induced-human leukemic HL-60 cell death was via both autophagy and apoptosis.