• 제목/요약/키워드: Lysosome pathway

검색결과 21건 처리시간 0.027초

Regulation of Protein Degradation by Proteasomes in Cancer

  • Jang, Ho Hee
    • Journal of Cancer Prevention
    • /
    • 제23권4호
    • /
    • pp.153-161
    • /
    • 2018
  • Imbalance of protein homeostasis (proteostasis) is known to cause cellular malfunction, cell death, and diseases. Elaborate regulation of protein synthesis and degradation is one of the important processes in maintaining normal cellular functions. Protein degradation pathways in eukaryotes are largely divided into proteasome-mediated degradation and lysosome-mediated degradation. Proteasome is a multisubunit complex that selectively degrades 80% to 90% of cellular proteins. Proteasome-mediated degradation can be divided into 26S proteasome (20S proteasome + 19S regulatory particle) and free 20S proteasome degradation. In 1980, it was discovered that during ubiquitination process, wherein ubiquitin binds to a substrate protein in an ATP-dependent manner, ubiquitin acts as a degrading signal to degrade the substrate protein via proteasome. Conversely, 20S proteasome degrades the substrate protein without using ATP or ubiquitin because it recognizes the oxidized and structurally modified hydrophobic patch of the substrate protein. To date, most studies have focused on protein degradation via 26S proteasome. This review describes the 26S/20S proteasomal pathway of protein degradation and discusses the potential of proteasome as therapeutic targets for cancer treatment as well as against diseases caused by abnormalities in the proteolytic system.

세포사멸 조절 단백질인 IEX-1의 새로운 결합단백질로서의 CATHEPSIN B의 발견 (Identification of CATHEPSIN B as a Novel Binding Protein of the Cell Death Regulating Protein IEX-1)

  • 유상미;이강석;배지현
    • 한국발생생물학회지:발생과생식
    • /
    • 제16권2호
    • /
    • pp.129-135
    • /
    • 2012
  • 선행연구에서 본 연구자는 IEX-1 단백질이 난소 암세포에서 세포사멸(apoptosis)을 유도하는 기능을 수행함을 확인하였으나, 세포사멸과 세포생존의 여러 단계에서 어떠한 신호전달 체계로 IEX-1이 작용하는지는 정확히 알지 못하고 있다. 따라서 IEX-1 단백질과 결합하는 새로운 단백질을 찾기 위해 yeast two-hybrid system을 이용하였다. 그 결과 IEX-1이 여러 다양한 인간 암 세포에서 세포사멸을 유도하는 CATHEPSIN B 단백질과 결합함을 밝혔다. 본 연구에서는 리소좀 프로테아제의 일원인 CATHEPSIN B 단백질과 IEX-1 단백질의 결합을 면역침강법과 western blot 분석으로 확인하였다. 그러므로 이 결과들을 통해서 IEX-1과 CATHEPSIN B는 세포 내에서 서로의 기능에 영향을 미칠 것으로 예상되며, 세포사멸을 유도하는 IEX-1 단백질이 lysosome-mediated apoptotic pathway에 관여할 가능성을 시사한다.

Synergetic effect of soluble whey protein hydrolysate and Panax ginseng berry extract on muscle atrophy in hindlimb-immobilized C57BL/6 mice

  • Han, Min Ji;Shin, Ji Eun;Park, Seok Jun;Choung, Se-Young
    • Journal of Ginseng Research
    • /
    • 제46권2호
    • /
    • pp.283-289
    • /
    • 2022
  • Background: Sarcopenia, defined as loss of muscle mass and strength with age, becomes a public health concern as the elderly population increases. This study aimed to determine whether the mixture of soluble whey protein hydrolysate (WPH) and Panax ginseng berry extract (GBE) has a synergetic effect on sarcopenia and, if so, to identify the relevant mechanisms and optimal mixing ratio. Methods: In the first experiment, C57BL/6 mice were hindlimb immobilized for one-week and then administered WPH 800 mg/kg, GBE 100 mg/kg, WPH 800 mg/kg+ GBE 100 mg/kg mixture, and Fructus Schisandrae extract (SFE) 200 mg/kg for two weeks. In the second experiment, experimental design was same, but mice were administered three different doses of WPH and GBE mixture (WPH 800 mg/kg+ GBE 100 mg/kg, WPH 800 mg/kg+ GBE 90 mg/kg, WPH 1000 mg/kg+ GBE 75 mg/kg). Results: In the first experiment, we confirmed the synergetic effect of WPH and GBE on muscle mass and identified that GBE was more effective on the protein synthesis side, and WPH tended to be slightly more effective for protein degradation. In the second experiment, among three different ratios, the WPH 800 mg/kg+ GBE 100 mg/kg was most effective for muscle mass and strength. The mixtures activated muscle protein synthesis via PI3K/Akt/mTORc1 pathway and inhibited muscle protein degradation via suppressing ubiquitin-proteasome system (UPS) and autophagy-lysosome system (ALS), and these effects were more GBE dose-dependent than WPH. Conclusion: The WPH and GBE mixture having a synergetic effect is a potential agent to prevent sarcopenia.

Amino-terminal arginylation as a degradation signal for selective autophagy

  • Cha-Molstad, Hyunjoo;Kwon, Yong Tae;Kim, Bo Yeon
    • BMB Reports
    • /
    • 제48권9호
    • /
    • pp.487-488
    • /
    • 2015
  • The ubiquitin-proteasome system and the autophagy lysosome system are the two major protein degradation machineries in eukaryotic cells. These two systems coordinate the removal of unwanted intracellular materials, but the mechanism by which they achieve this synchronization is largely unknown. The ubiquitination of substrates serves as a universal degradation signal for both systems. Our study revealed that the amino-terminal Arg, a canonical N-degron in the ubiquitin-proteasome system, also acts as a degradation signal in autophagy. We showed that many ER residents, such as BiP, contain evolutionally conserved arginylation permissive pro-N-degrons, and that certain inducers like dsDNA or proteasome inhibitors cause their translocation into the cytoplasm where they bind misfolded proteins and undergo amino-terminal arginylation by arginyl transferase 1 (ATE1). The amino-terminal Arg of BiP binds p62, which triggers p62 oligomerization and enhances p62-LC3 interaction, thereby stimulating autophagic delivery and degradation of misfolded proteins, promoting cell survival. This study reveals a novel ubiquitin-independent mechanism for the selective autophagy pathway, and provides an insight into how these two major protein degradation pathways communicate in cells to dispose the unwanted proteins. [BMB Reports 2015; 48(9): 487-488]

Human Intersectin 2 (ITSN2) binds to Eps8 protein and enhances its degradation

  • Ding, Xiaofeng;Yang, Zijian;Zhou, Fangliang;Hu, Xiang;Zhou, Chang;Luo, Chang;He, Zhicheng;Liu, Qian;Li, Hong;Yan, Feng;Wang, Fangmei;Xiang, Shuanglin;Zhang, Jian
    • BMB Reports
    • /
    • 제45권3호
    • /
    • pp.183-188
    • /
    • 2012
  • Participates in actin remodeling through Rac and receptor endocytosis via Rab5. Here, we used yeast two-hybrid system with Eps8 as bait to screen a human brain cDNA library. ITSN2 was identified as the novel binding factor of Eps8. The interaction between ITSN2 and Eps8 was demonstrated by the in vivo co-immunoprecipitation and colocalization assays and the in vitro GST pull-down assays. Furthermore, we mapped the interaction domains to the region between amino acids 260-306 of Eps8 and the coiled-coil domain of ITSN2. In addition, protein stability assays and immunofluorescence analysis showed ITSN2 overexpression induced the degradation of Eps8 proteins, which was markedly alleviated with the lysosome inhibitor NH4Cl treatment. Taken together, our results suggested ITSN2 interacts with Eps8 and stimulates the degradation of Eps8 proteins.

암 치료 표적으로써 cathepsin S (Cathepsin S as a Cancer Therapeutic Target)

  • 우선민;권택규
    • 생명과학회지
    • /
    • 제28권6호
    • /
    • pp.753-763
    • /
    • 2018
  • Cathepsin은 리소좀에 존재하는 효소로, 엔도좀-리소좀 경로를 통하여 손상된 단백질의 분해를 유도한다. 이러한 cathepsin은 활성화되는 촉매 잔기 위치(catalytic residue site)에 따라 cysteine, aspartate, serine cathepsin으로 나뉜다. 다양한 암세포에서 cysteine cathepsin은 유전자 증폭이나 전사, 번역, 전사 후 단계를 통해 높은 발현을 유지한다. 특히 cathepsin S의 경우, 다른 cysteine cathepsin과 달리 중성 pH 환경에서도 안정적으로 활성도를 유지하며 특정 조직에서 발현이 제한적이라는 특징을 가지고 있기 때문에 질병의 미세환경에서 중요한 역할을 한다. 면역세포에서의 cathepsin S는 불변 사슬(invariant chain)을 분해하여 항원 제시에 중요한 MHC class II의 활성화를 통해 면역 반응을 조절하며, 암세포에서의 cathepsin S는 전이와 혈관신생을 조절함으로써 암세포의 성장을 증가시키는 역할을 한다. Cathepsin S의 발현 조절에 문제가 생기면 암, 관절염, 심혈관 질환을 포함한 많은 병리학적 현상에 영향을 미친다. 특히 암세포에서 cathepsin S의 발현 억제와 결함은 혈관신생을 억제시킬 뿐만아니라, 암세포의 성장과 전이를 감소시키고 세포사멸을 유도한다. 따라서, cathepsin S는 암 치료에 있어 좋은 표적이 될 수 있다.

Neuroprotective Effect of Chebulagic Acid via Autophagy Induction in SH-SY5Y Cells

  • Kim, Hee Ju;Kim, Joonki;Kang, Ki Sung;Lee, Keun Taik;Yang, Hyun Ok
    • Biomolecules & Therapeutics
    • /
    • 제22권4호
    • /
    • pp.275-281
    • /
    • 2014
  • Autophagy is a series of catabolic process mediating the bulk degradation of intracellular proteins and organelles through formation of a double-membrane vesicle, known as an autophagosome, and fusing with lysosome. Autophagy plays an important role of death-survival decisions in neuronal cells, which may influence to several neurodegenerative disorders including Parkinson's disease. Chebulagic acid, the major constituent of Terminalia chebula and Phyllanthus emblica, is a benzopyran tannin compound with various kinds of beneficial effects. This study was performed to investigate the autophagy enhancing effect of chebulagic acid on human neuroblastoma SH-SY5Y cell lines. We determined the effect of chebulagic acid on expression levels of autophagosome marker proteins such as, DOR/TP53INP2, Golgi-associated ATPase Enhancer of 16 kDa (GATE 16) and Light chain 3 II (LC3 II), as well as those of its upstream pathway proteins, AMP-activated protein kinase (AMPK), mammalian target of rapamycin (mTOR) and Beclin-1. All of those proteins were modulated by chebulagic acid treatment in a way of enhancing the autophagy. Additionally in our study, chebulagic acid also showed a protective effect against 1-methyl-4-phenylpyridinium ($MPP^+$) - induced cytotoxicity which mimics the pathological symptom of Parkinson's disease. This effect seems partially mediated by enhanced autophagy which increased the degradation of aggregated or misfolded proteins from cells. This study suggests that chebulagic acid is an attractive candidate as an autophagy-enhancing agent and therefore, it may provide a promising strategy to prevent or cure the diseases caused by accumulation of abnormal proteins including Parkinson's disease.

Gintonin stimulates autophagic flux in primary cortical astrocytes

  • Rahman, Md. Ataur;Hwang, Hongik;Nah, Seung-Yeol;Rhim, Hyewhon
    • Journal of Ginseng Research
    • /
    • 제44권1호
    • /
    • pp.67-78
    • /
    • 2020
  • Background: Gintonin (GT), a novel ginseng-derived exogenous ligand of lysophosphatidic acid (LPA) receptors, has been shown to induce cell proliferation and migration in the hippocampus, regulate calcium-dependent ion channels in the astrocytes, and reduce β-amyloid plaque in the brain. However, whether GT influences autophagy in cortical astrocytes is not yet investigated. Methods: We examined the effect of GT on autophagy in primary cortical astrocytes using immunoblot and immunocytochemistry assays. Suppression of specific proteins was performed via siRNA. LC3 puncta was determined using confocal microscopy. Results: GT strongly upregulated autophagy marker LC3 by a concentration- as well as time-dependent manner via G protein-coupled LPA receptors. GT-induced autophagy was further confirmed by the formation of LC3 puncta. Interestingly, on pretreatment with an mammalian target of rapamycin (mTOR) inhibitor, rapamycin, GT further enhanced LC3-II and LC3 puncta expression. However, GT-induced autophagy was significantly attenuated by inhibition of autophagy by 3-methyladenine and knockdown Beclin-1, Atg5, and Atg7 gene expression. Importantly, when pretreated with a lysosomotropic agent, E-64d/peps A or bafilomycin A1, GT significantly increased the levels of LC3-II along with the formation of LC3 puncta. In addition, GT treatment enhanced autophagic flux, which led to an increase in lysosome-associated membrane protein 1 and degradation of ubiquitinated p62/SQSTM1. Conclusion: GT induces autophagy via mTOR-mediated pathway and elevates autophagic flux. This study demonstrates that GT can be used as an autophagy-inducing agent in cortical astrocytes.

중성지방에 의한 대식세포 사멸 과정에서 Cathepsin B의 영향 (Cathepsin B Is Implicated in Triglyceride (TG)-Induced Cell Death of Macrophage)

  • 정병출;임재원;김성훈;김윤석
    • 대한임상검사과학회지
    • /
    • 제52권3호
    • /
    • pp.245-252
    • /
    • 2020
  • 대식세포사멸은 죽상판 형성에 영향을 미쳐 죽상동맥경화증 발병에 관여하는 것으로 알려져 있다. 중성지방 역시 죽상동맥경화 발병에 기여한다고 알려져 있는데 최근 본 연구팀에서는 중성지방이 대식세포사멸을 유발한다는 결과를 확인하였다. 본 연구에서는 cathepsin B가 중성지방에 의해 유발되는 대식세포사멸 과정에 관여하는지 확인하고자 연구를 진행하였다. THP-1 대식세포에 중성지방 처리 시 cathepsin B의 발현량에는 변화가 없고 리소좀에 있던 cathepsin B가 세포질로 방출되어 세포질의 cathepsin B가 증가한 것을 확인하였다. 다음으로 cathepsin B 억제제인 CA-074 Me를 처리 시 중성지방에 의해 유도되는 대식세포사멸이 일부 회복되는 것을 확인하였다. 본 연구팀의 이전 연구에서 중성지방에 의한 대식세포사멸이 caspase-1, -2 및 apoptotic caspase 활성화를 매개로 일어남을 확인하였기 때문에 본 연구에서는 이러한 caspase 활성 경로와 cathepsin B와의 연관성에 대해 연구하였다. cathepsin B 억제시 caspase-7, -8 및 -1의 활성은 억제되었으나, caspase-3, -9 및 -2는 활성에는 영향을 미치지 않음을 알 수 있었다. 정리하면, 중성지방에 의해 세포질로 방출된 cathepsin B는 caspase-1 활성화에 기여하고, 활성화된 caspase-1은 외인성 apoptotic caspase 경로를 활성화하여 THP-1 대식세포 사멸을 유발한다는 것을 알 수 있다.

Melatonin protects endothelial progenitor cells against AGE-induced apoptosis via autophagy flux stimulation and promotes wound healing in diabetic mice

  • Jin, Haiming;Zhang, Zengjie;Wang, Chengui;Tang, Qian;Wang, Jianle;Bai, Xueqin;Wang, Qingqing;Nisar, Majid;Tian, Naifeng;Wang, Quan;Mao, Cong;Zhang, Xiaolei;Wang, Xiangyang
    • Experimental and Molecular Medicine
    • /
    • 제50권11호
    • /
    • pp.13.1-13.15
    • /
    • 2018
  • Wound healing is delayed in diabetic patients. Increased apoptosis and endothelial progenitor cell (EPC) dysfunction are implicated in delayed diabetic wound healing. Melatonin, a major secretory product of the pineal gland, promotes diabetic wound healing; however, its mechanism of action remains unclear. Here, EPCs were isolated from the bone marrow of mice. Treatment of EPCs with melatonin alleviated advanced glycation end product (AGE)-induced apoptosis and cellular dysfunction. We further examined autophagy flux after melatonin treatment and found increased light chain 3 (LC3) and p62 protein levels in AGE-treated EPCs. However, lysosome-associated membrane protein 2 expression was decreased, indicating that autophagy flux was impaired in EPCs treated with AGEs. We then evaluated autophagy flux after melatonin treatment and found that melatonin increased the LC3 levels, but attenuated the accumulation of p62, suggesting a stimulatory effect of melatonin on autophagy flux. Blockage of autophagy flux by chloroquine partially abolished the protective effects of melatonin, indicating that autophagy flux is involved in the protective effects of melatonin. Furthermore, we found that the AMPK/mTOR signaling pathway is involved in autophagy flux stimulation by melatonin. An in vivo study also illustrated that melatonin treatment ameliorated impaired wound healing in a streptozotocin-induced diabetic wound healing model. Thus, our study shows that melatonin protects EPCs against apoptosis and dysfunction via autophagy flux stimulation and ameliorates impaired wound healing in vivo, providing insight into its mechanism of action in diabetic wound healing.