• 제목/요약/키워드: microtubule associated protein light chain 3

검색결과 40건 처리시간 0.021초

Regulatory Role of Autophagy in Globular Adiponectin-Induced Apoptosis in Cancer Cells

  • Nepal, Saroj;Park, Pil-Hoon
    • Biomolecules & Therapeutics
    • /
    • 제22권5호
    • /
    • pp.384-389
    • /
    • 2014
  • Adiponectin, an adipokine predominantly secreted from adipose tissue, exhibits diverse biological responses, including metabolism of glucose and lipid, and apoptosis in cancer cells. Recently, adiponectin has been shown to modulate autophagy as well. While emerging evidence has demonstrated that autophagy plays a role in the modulation of proliferation and apoptosis of cancer cells, the role of autophagy in apoptosis of cancer cell caused by adiponectin has not been explored. In the present study, we demonstrated that globular adiponectin (gAcrp) induces both apoptosis and autophagy in human hepatoma cell line (HepG2 cells) and breast cancer cells (MCF-7), as evidenced by increase in caspase-3 activity, Bax, microtubule-associated protein light chain 3-II (LC3 II) protein levels, and autophagosome formation. Interestingly, gene silencing of LC3B, an autophagy marker, significantly enhanced gAcrp-induced apoptosis in both HepG2 and MCF-7 cell lines, whereas induction of autophagy by rapamycin, an mTOR inhibitor, significantly prevented gAcrp-induced apoptosis in hepatoma cells HepG2. Furthermore, modulation of autophagy produced similar effects on gAcrp-induced Bax expression in HepG2 cells. These results implicate that induction of autophagy plays a regulatory role in adiponectin-induced apoptosis of cancer cells, and thus inhibition of autophagy would be a novel promising target to enhance the efficiency of cancer cell apoptosis by adiponectin.

포공영(蒲公英)이 유방암 세포의 사멸에 미치는 영향 (Effects of Taraxaci Herba on Cell Death in Breast Cancer Cells)

  • 서국장;박경미;조성희;양승정
    • 대한한방부인과학회지
    • /
    • 제28권2호
    • /
    • pp.1-14
    • /
    • 2015
  • Objectives : This study was designed to investigate the effects of Taraxaci Herba (TH) on cell death in breast cancer cells. Methods : In this experiment, the effects of TH on proliferation rates, cell morphology and growth pattern, intracellular reactive oxygen species (ROS) production. In addition, the effects on nuclear condensation, fragmentation and formation of acidic vesicular organelles (AVO) in MCF-7 cells were also investigated. Finally, autophagy related with protein was observed by using western blot method. Results : TH inhibited proliferation of MCF-7 cells, TH elevated intracellular ROS levels significantly. Treatment with TH did not affect nuclear morphologies such as condensation or fragmentation. On the other hand, TH treatment effectively induced AVO. Finally, one of autophagy related with protein, Microtubule-associated proteins 1A/1B light chain 3A (MAP1LC3A, LC3) level was elevated by treatment with TH. Conclusions : These data indicate that TH is able to be used for patient with breast cancer and mechanisms are involved in autophagy through ROS generation.

Heterotrimeric Kinesin 2 모터 단백질의 Carboxyl-말단과 β2-tubulin의 결합 (The Carboxyl-terminal Tail of a Heterotrimeric Kinesin 2 Motor Subunit Directly Binds to β2-tubulin)

  • 정영주;박성우;김상진;이원희;김무성;엄상화;석대현
    • 생명과학회지
    • /
    • 제29권3호
    • /
    • pp.369-375
    • /
    • 2019
  • 미세소관은 알파- 와 베타-tubulin의 이량체가 종합되어 형성되며, 또한 세포내에서 tubulin 수송은 섬모나 편모와 같은 세포 부착 기관의 성장에 중요한 역할을 한다. Kinesin 2는 kinesin superfamily (KIF)의 분자 모터 단백질의 한 종류로 미세소관를 따라 다양한 운반체를 운반하며, 2종류의 다른 모터 단백질(KIF3A, KIF3B)과 kinesin-associated protein 3 (KAP3)로 구성되어 있다. Kinesin 2는 KIF3A의 cargo binding domain을 통하여 다양한 단백질과의 결합이 알려져 있지만, 아직 결합단백질의 다수는 아직 밝혀지지 않았다. 본 연구에서 KIF3A와 결합하는 단백질을 분리하기 위하여 효모 two-hybrid system을 사용하여 탐색한 결과 미세소관의 단위체의 한 종류인 ${\beta}2-tubulin$ type (Tubb2)을 분리하였다. Tubb2는 KIF3A와 결합하지만, KIF3B, KIF5B와 kinesin light chain 1 (KLC1)과는 결합하지 않았다. Tubb2의 C-말단은 KIF3A와의 결합에 필요하며, 다른 KIF3A는 Tubb의 isoforms인 Tubb1, Tubb2, Tubb3, Tubb4, Tubb5와도 결합하였다. 그러나 Tuba1은 KIF3A와 결합하지 않았다. 생쥐의 뇌 파쇄액을 KIF3A 항체로 면역침강한 결과 Tubb2는 heterotrimeric kinesin 2의 구성단백질들과 같이 침강하였다. 이러한 결과들은 heterotrimeric kinesin 2는 tubulin과 결합하여 세포 내에서 tubulins을 운반하는 것을 시사한다.

Anti-inflammatory effects of N-cyclooctyl-5-methylthiazol-2-amine hydrobromide on lipopolysaccharide-induced inflammatory response through attenuation of NLRP3 activation in microglial cells

  • Kim, Eun-A;Hwang, Kyouk;Kim, Ji-Eun;Ahn, Jee-Yin;Choi, Soo Young;Yang, Seung-Ju;Cho, Sung-Woo
    • BMB Reports
    • /
    • 제54권11호
    • /
    • pp.557-562
    • /
    • 2021
  • Microglial activation is closely associated with neuroinflammatory pathologies. The nucleotide-binding and oligomerization domain-like receptor containing a pyrin domain 3 (NLRP3) inflammasomes are highly organized intracellular sensors of neuronal alarm signaling. NLRP3 inflammasomes activate nuclear factor kappa-B (NF-κB) and reactive oxygen species (ROS), which induce inflammatory responses. Moreover, NLRP3 dysfunction is a common feature of chronic inflammatory diseases. The present study investigated the effect of a novel thiazol derivative, N-cyclooctyl-5-methylthiazol-2-amine hydrobromide (KHG26700), on inflammatory responses in lipopolysaccharide (LPS)-treated BV-2 microglial cells. KHG26700 significantly attenuated the expression of several pro-inflammatory cytokines, including tumor necrosis factor-α, interleukin-1β, and interleukin-6, in these cells, as well as the LPS-induced increases in NLRP3, NF-κB, and phospho-IkBα levels. KHG26700 also suppressed the LPS-induced increases in protein levels of autophagy protein 5 (ATG5), microtubule-associated protein 1 light chain 3 (LC3), and beclin-1, as well as downregulating the LPS-enhanced levels of ROS, lipid peroxidation, and nitric oxide. These results suggest that the anti-inflammatory effects of KHG26700 may be due, at least in part, to the regulation of the NLRP3-mediated signaling pathway during microglial activation.

Kinesin Light Chain 1 (KLC1)의 Tetratricopeptide Repeat (TPR) 도메인과 Rab effector, EHBP1L1의 결합 (Rab Effector EHBP1L1 Associates with the Tetratricopeptide Repeat Domain of Kinesin Light Chain 1)

  • 정영주;박성우;김상진;김무성;엄상화;이정구;석대현
    • 생명과학회지
    • /
    • 제30권1호
    • /
    • pp.10-17
    • /
    • 2020
  • Kinesin 1은 미세소관을 따라 plus말단으로 이동하는 모터단백질로 세포내 물질 수송에 관여한다. Kinesin 1은 경쇄단위체(light chain subunit)를 통하여 운반체들인, 세포내 소기관, 다양한 소포체, 신경전달물질 수용체 단백질, 세포신호전달 단백질과 여러 단백질 복합체들과 결합하여 운반하는 kinesin superfamily protein (KIFs)의 한 종류이다. Kinesin light chains 1 (KLC1)은 모터 기능이 없는 단위체로서 kinesin heavy chain (KHC)과 결합한다. KLC1은 다양한 매개단백질들과 결합하지만 아직 결합하는 매개단백질이 충분히 밝혀지지 않았다. 본 연구에서는 KLC1의 tetratricopeptide repeat (TPR) 영역과 결합하는 단백질을 분리하기 위하여 효모 two-hybrid 탐색한 결과 EH domain-binding protein 1-like 1 (EHBP1L1) 을 분리하였다. EHBP1L1은 KLC1의 TPR 영역을 포함한 부위와 결합하지만 KIF5B (kinesin 1의 모터 단백질)과 KIF3A (kinesin 2의 모터 단백질)와는 결합하지 않았다. 또한 KLC1은 EHBP1L1의 C-말단에 존재하는 coiled-coil 도메인과 결합하였으며, 다른 EHBP1L1의 isoform인 EHBP1과는 결합하지 않았다. KLC1은 GST와는 결합하지 않지만 GST-EHBP1L1과 GST-EHBP1L1-coiled-coil domain과는 결합하였다. HEK-293T세포에 EHBP1L1과 KLC1을 동시에 발현시켰을 때 두 단백질은 세포 내에서 같은 부위에 존재하며, EHBP1L1을 면역침강한 결과 KLC1뿐만 아니라 KIF5B와도 같이 침강함을 확인하였다. 이러한 결과들은 kinesin 1은 EHBP1L1이 결합한 운반체를 수송함을 시사한다.

Localization of Autophagosome in Porcine Follicular Cumulus-oocyte Complex

  • Lee, Seunghoon;Kim, Dong-Hoon;Im, Gi-Sun;Ock, Sun-A;Ullah, Imran;Hur, Tai-Young
    • 한국수정란이식학회지
    • /
    • 제32권3호
    • /
    • pp.105-109
    • /
    • 2017
  • Autophagy is an intracellular degradation and recycling system. Oocyte maturation is dynamic process, in which various proteins should be synthesized and degraded. In our previous study, we reported the loci of autophagosome and dynamics of autophagic activity in porcine oocytes during in vitro maturation. In this study, we verified loci of autophagosome in porcine follicular cumulus-oocyte complex by detection of microtubule-associated protein 1A/1B-light chain 3 (LC3) which is the reliable marker of autophagosome. Porcine ovary including various sizes of follicles was fixed within 1 hour after collection from slaughterhouse. After fixation, immunohistochemistry was conducted on sliced ovary tissue containing various sizes of follicles by using LC3 antibody. As a result, LC3 signal was clearly detected in both cumulus and oocytes of various sizes of follicles. We also found ring shaped signal which represent autophagosome near oocyte membrane. Most of the signals in oocytes were localized nearby cellular membrane while evenly dispersed in cumulus cells. Therefore, this result suggests that autophagy occurs in porcine COCs (cumulus-oocyte complexes) at follicular stage.

α, γ-Mangostins Induce Autophagy and Show Synergistic Effect with Gemcitabine in Pancreatic Cancer Cell Lines

  • Kim, Myoungjae;Chin, Young-Won;Lee, Eun Joo
    • Biomolecules & Therapeutics
    • /
    • 제25권6호
    • /
    • pp.609-617
    • /
    • 2017
  • Pancreatic cancer is one of the most lethal and aggressive cancers in the world. However, no effective treatment is currently available for pancreatic cancer. The objective of this study was to determine the anti-pancreatic cancer effect of ${\alpha}$-mangostin (${\alpha}M$) and ${\gamma}$-mangostin (${\gamma}M$) extracted from the pericarp of Garcinia mangostana L.. Both ${\alpha}$M and ${\gamma}M$ reduced the viability of pancreatic cancer cells MIA PaCa-2 and PANC-1 in a dose-dependent manner. These compounds induced apoptosis by increasing c-PARP and c-Caspase 3 levels. They also induced autophagy by increasing levels of microtubule-associated protein 1A/1B light chain 3B (LC3II) in both cell lines while decreasing sequestosome 1 (p62) in MIA PaCa-2. Both ${\alpha}$M and ${\gamma}M$ induced autophagy through increasing phosphorylation levels of AMP-activated protein kinase (p-AMPK) and p38-mitogen activated protein kinase (p-p38) while decreasing phosphorylation level of mammalian target of rapamycin complex 1 (p-mTOR). Of various microRNAs (miRNA), miR-18a was found to be a putative regulatory miRNA for autophagy induced by ${\alpha}$M or ${\gamma}M$. In combination with gemcitabine, a compound frequently used in pancreatic cancer treatment, ${\alpha}$M and ${\gamma}M$ showed synergistic anti-cancer effects in MIA PaCa-2. Collectively, these results suggest that ${\alpha}$M and ${\gamma}M$ can induce apoptosis and autophagy in pancreatic cancer cells and that their anti-cancer effect is likely to be associated with miR-18a. In conclusion, ${\alpha}$M and ${\gamma}M$ might be used as a potential new therapy for pancreatic cancer.

Induction of Apoptosis and Autophagy by Resveratrol via Activation of AMPK Pathway in 5-Fluorouracil-Resistant Colon Cancer Cells

  • Lee, Seung Young;Jung, Ji Young;Choi, Hee Won;Choi, Kyung Min;Jeong, Jin-Woo
    • 한국자원식물학회:학술대회논문집
    • /
    • 한국자원식물학회 2018년도 추계학술대회
    • /
    • pp.101-101
    • /
    • 2018
  • Resveratrol is a polyphenolic compound, which is a naturally occurring phytochemical and is found in a variety of plants, including food such as grapes, berries and peanuts. Although several studies have demonstrated that resveratrol possesses anti-cancer activity against various types of human cancer, the molecular mechanisms of resveratrol-mediated overcome drug resistance potential are unclear. In this study, we determined whether resveratrol attenuates drug resistance responses in 5-fluorouracil-resistant colon cancer (SNUC5/5-FUR) cells. Treatment with resveratrol significantly enhanced apoptosis in a concentration-dependent manner, which was associated with the modulation of anti- and/or pro-apoptotic protein expression, activation of caspases and activation of AMP-activated protein kinase. Resveratrol treatment also increased the induction of autophagy through up-regulation of autophagy-related genes such as Microtubule-associated protein 1A/1B-light chain 3, P62 and beclin-1. However, blocking of autophagy by bafilomycin A1 reduced apoptotic cell death, suggesting that resveratrol-induced autophagy functions as a cell death mechanism in SNUC5/5-FU cells. Although the further studies are needed, these findings suggest that resveratrol may have therapeutic potential to overcome drug resistance in colon cancer patients.

  • PDF

29-kDa FN-f inhibited autophagy through modulating localization of HMGB1 in human articular chondrocytes

  • Hwang, Hyun Sook;Choi, Min Ha;Kim, Hyun Ah
    • BMB Reports
    • /
    • 제51권10호
    • /
    • pp.508-513
    • /
    • 2018
  • Fibronectin fragments found in the synovial fluid of patients with osteoarthritis (OA) induce the catabolic responses in cartilage. Nuclear high-mobility group protein Box 1 (HMGB1), a damage-associated molecular pattern, is responsible for the regulation of signaling pathways related to cell death and survival in response to various stimuli. In this study, we investigated whether changes induced by 29-kDa amino-terminal fibronectin fragment (29-kDa FN-f) in HMGB1 expression influences the pathogenesis of OA via an HMGB1-modulated autophagy signaling pathway. Human articular chondrocytes were enzymatically isolated from articular cartilage. The level of mRNA was measured by quantitative real-time PCR. The expression of proteins was examined by western blot analysis, immnunofluorescence assay, and enzyme-linked immunosorbent assay. Interaction of proteins was evaluated by immunoprecipitation. The HMGB1 level was significantly lower in human OA cartilage than in normal cartilage. Although 29-kDa FN-f significantly reduced the HMGB1 expression at the mRNA and protein levels 6 h after treatment, the cytoplasmic level of HMGB1 was increased in chondrocytes treated with 29-kDa FN-f, which significantly inhibited the interaction of HMGB1 with Beclin-1, increased the interaction of Bcl-2 with Beclin-1, and decreased the levels of Beclin-1 and phosphorylated Bcl-2. In addition, the level of microtubule-associated protein 1 light chain 3-II, an autophagy marker, was down-regulated in chondrocytes treated with 29-kDa FN-f, whereas the effect was antagonized by mTOR knockdown. Furthermore, prolonged treatment with 29-kDa FN-f significantly increased the release of HMGB1 into the culture medium. These results demonstrated that 29-kDa FN-f inhibits chondrocyte autophagy by modulating the HMGB1 signaling pathway.

Neuroprotective Effect of Astersaponin I against Parkinson's Disease through Autophagy Induction

  • Zhang, Lijun;Park, Jeoung Yun;Zhao, Dong;Kwon, Hak Cheol;Yang, Hyun Ok
    • Biomolecules & Therapeutics
    • /
    • 제29권6호
    • /
    • pp.615-629
    • /
    • 2021
  • An active compound, triterpene saponin, astersaponin I (AKNS-2) was isolated from Aster koraiensis Nakai (AKNS) and the autophagy activation and neuroprotective effect was investigated on in vitro and in vivo Parkinson's disease (PD) models. The autophagy-regulating effect of AKNS-2 was monitored by analyzing the expression of autophagy-related protein markers in SH-SY5Y cells using Western blot and fluorescent protein quenching assays. The neuroprotection of AKNS-2 was tested by using a 1-methyl-4-phenyl-2,3-dihydropyridium ion (MPP+)-induced in vitro PD model in SH-SY5Y cells and an MPTP-induced in vivo PD model in mice. The compound-treated SH-SY5Y cells not only showed enhanced microtubule-associated protein 1A/1B-light chain 3-II (LC3-II) and decreased sequestosome 1 (p62) expression but also showed increased phosphorylated extracellular signal-regulated kinases (p-Erk), phosphorylated AMP-activated protein kinase (p-AMPK) and phosphorylated unc-51-like kinase (p-ULK) and decreased phosphorylated mammalian target of rapamycin (p-mTOR) expression. AKNS-2-activated autophagy could be inhibited by the Erk inhibitor U0126 and by AMPK siRNA. In the MPP+-induced in vitro PD model, AKNS-2 reversed the reduced cell viability and tyrosine hydroxylase (TH) levels and reduced the induced α-synuclein level. In an MPTP-induced in vivo PD model, AKNS-2 improved mice behavioral performance, and it restored dopamine synthesis and TH and α-synuclein expression in mouse brain tissues. Consistently, AKNS-2 also modulated the expressions of autophagy related markers in mouse brain tissue. Thus, AKNS-2 upregulates autophagy by activating the Erk/mTOR and AMPK/mTOR pathways. AKNS-2 exerts its neuroprotective effect through autophagy activation and may serve as a potential candidate for PD therapy.