• Title/Summary/Keyword: Co-immunoprecipitation

Search Result 83, Processing Time 0.022 seconds

Hepatitis B Virus DNA Polymerase Displays an Anti-Apoptotic Effect by Interacting with Elongation Factor-1 Alpha-2 in Hepatoma Cells

  • Niu, Xianli;Nong, Shirong;Gong, Junyuan;Zhang, Xin;Tang, Hui;Zhou, Tianhong;Li, Wei
    • Journal of Microbiology and Biotechnology
    • /
    • v.31 no.1
    • /
    • pp.16-24
    • /
    • 2021
  • Hepatitis B virus (HBV) genome P-encoded protein HBV DNA polymerase (Pol) has long been known as a reverse transcriptase during HBV replication. In this study, we investigated the impact of HBV Pol on host cellular processes, mainly apoptosis, and the underlying mechanisms. We showed a marked reduction in apoptotic rates in the HBV Pol-expressed HepG2 cells compared to controls. Moreover, a series of assays, i.e., yeast two-hybrid, GST pull-down, co-immunoprecipitation, and confocal laser scanning microscopy, identified the host factor eEF1A2 to be associated with HBV Pol. Furthermore, knockdown of eEF1A2 gene by siRNA abrogated the HBV Pol-mediated anti-apoptotic effect with apoptosis induced by endoplasmatic reticulum (ER) stress-inducer thapsigargin (TG), thus suggesting that the host factor eEF1A2 is essential for HBV Pol's anti-apoptosis properties. Our findings have revealed a novel role for HBV Pol in its modulation of apoptosis through integrating with eEF1A2.

Wnt-C59 inhibits proinflammatory cytokine expression by reducing the interaction between β-catenin and NF-κB in LPS-stimulated epithelial and macrophage cells

  • Jang, Jaewoong;Song, Jaewon;Sim, Inae;Yoon, Yoosik
    • The Korean Journal of Physiology and Pharmacology
    • /
    • v.25 no.4
    • /
    • pp.307-319
    • /
    • 2021
  • Dysregulation of the Wnt pathway causes various diseases including cancer, Parkinson's disease, Alzheimer's disease, schizophrenia, osteoporosis, obesity and chronic kidney diseases. The modulation of dysregulated Wnt pathway is absolutely necessary. In the present study, we evaluated the anti-inflammatory effect and the mechanism of action of Wnt-C59, a Wnt signaling inhibitor, in lipopolysaccharide (LPS)-stimulated epithelial cells and macrophage cells. Wnt-C59 showed a dose-dependent anti-inflammatory effect by suppressing the expression of proinflammatory cytokines including IL6, CCL2, IL1A, IL1B, and TNF in LPS-stimulated cells. The dysregulation of the Wnt/β-catenin pathway in LPS stimulated cells was suppressed by WntC59 treatment. The level of β-catenin, the executor protein of Wnt/β-catenin pathway, was elevated by LPS and suppressed by Wnt-C59. Overexpression of β-catenin rescued the suppressive effect of Wnt-C59 on proinflammatory cytokine expression and nuclear factor-kappa B (NF-κB) activity. We found that the interaction between β-catenin and NF-κB, measured by co-immunoprecipitation assay, was elevated by LPS and suppressed by Wnt-C59 treatment. Both NF-κB activity for its target DNA binding and the reporter activity of NF-κB-responsive promoter showed identical patterns with the interaction between β-catenin and NF-κB. Altogether, our findings suggest that the anti-inflammatory effect of Wnt-C59 is mediated by the reduction of the cellular level of β-catenin and the interaction between β-catenin and NF-κB, which results in the suppressions of the NF-κB activity and proinflammatory cytokine expression.

Annexin A2 gene interacting with viral matrix protein to promote bovine ephemeral fever virus release

  • Chen, Lihui;Li, Xingyu;Wang, Hongmei;Hou, Peili;He, Hongbin
    • Journal of Veterinary Science
    • /
    • v.21 no.2
    • /
    • pp.33.1-33.15
    • /
    • 2020
  • Bovine ephemeral fever virus (BEFV) causes bovine ephemeral fever, which can produce considerable economic damage to the cattle industry. However, there is limited experimental evidence regarding the underlying mechanisms of BEFV. Annexin A2 (AnxA2) is a calcium and lipid-conjugated protein that binds phospholipids and the cytoskeleton in a Ca2+-dependent manner, and it participates in various cellular functions, including vesicular trafficking, organization of membrane domains, and virus proliferation. The role of the AnxA2 gene during virus infection has not yet been reported. In this study, we observed that AnxA2 gene expression was up-regulated in BHK-21 cells infected with the virus. Additionally, overexpression of the AnxA2 gene promoted the release of mature virus particles, whereas BEFV replication was remarkably inhibited after reducing AnxA2 gene expression by using the small interfering RNA (siRNA). For viral proteins, overexpression of the Matrix (M) gene promotes the release of mature virus particles. Moreover, the AnxA2 protein interaction with the M protein of BEFV was confirmed by GST pull-down and co-immunoprecipitation assays. Experimental results indicate that the C-terminal domain (268-334 aa) of AxnA2 contributes to this interaction. An additional mechanistic study showed that AnxA2 protein interacts with M protein and mediates the localization of the M protein at the plasma membrane. Furthermore, the absence of the AnxA2-V domain could attenuate the effect of AnxA2 on BEFV replication. These findings can contribute to elucidating the regulation of BEFV replication and may have implications for antiviral strategy development.

Ginsenoside 20(S)-Rg3 reduces KIF20A expression and promotes CDC25A proteasomal degradation in epithelial ovarian cancer

  • Rong Zhang;Lei Li;Huihui Li;Hansong Bai;Yuping Suo;Ju Cui;Yingmei Wang
    • Journal of Ginseng Research
    • /
    • v.48 no.1
    • /
    • pp.40-51
    • /
    • 2024
  • Background: Ginsenoside 20(S)-Rg3 shows promising tumor-suppressive effects in ovarian cancer via inhibiting NF-kB signaling. This study aimed to explore the downstream tumor suppressive mechanisms of ginsenoside Rg3 via this signaling pathway. Materials and methods: A systematical screening was applied to examine the expression profile of 41 kinesin family member genes in ovarian cancer. The regulatory effect of ginsenoside Rg3 on KIF20A expression was studied. In addition, we explored interacting proteins of KIF20A and their molecular regulations in ovarian cancer. RNA-seq data from The Cancer Genome Atlas (TCGA) was used for bioinformatic analysis. Epithelial ovarian cancer cell lines SKOV3 and A2780 were used as in vitro and in vivo cell models. Commercial human ovarian cancer tissue arrays were used for immunohistochemistry staining. Results: KIF20A is a biomarker of poor prognosis among the kinesin genes. It promotes ovarian cancer cell growth in vitro and in vivo. Ginsenoside Rg3 can suppress the transcription of KIF20A. GST pull-down and co-immunoprecipitation (IP) assays confirmed that KIF20A physically interacts with BTRC (β-TrCP1), a substrate recognition subunit for SCFβ-TrCP E3 ubiquitin ligase. In vitro ubiquitination and cycloheximide (CHX) chase assays showed that via interacting with BTRC, KIF20A reduces BTRC-mediated CDC25A poly-ubiquitination and enhances its stability. Ginsenoside Rg3 treatment partly abrogates KIF20A overexpression-induced CDC25A upregulation. Conclusion: This study revealed a novel anti-tumor mechanism of ginsenoside Rg3. It can inhibit KIF20A transcription and promote CDC25A proteasomal degradation in epithelial ovarian cancer.

A Nucleotide Exchange Factor, BAP, dissociated Protein-Molecular Chaperone Complex in vitro (In vitro에서 핵산치환인자 BAP이 단백질-분자 샤페론 복합체 해리에 미치는 영향)

  • Lee Myoung-Joo;Kim Dong-Eun;Lee Tae-Ho;Jeong Yong-Kee;Kim Young-Hee;Chung Kyung-Tae
    • Journal of Life Science
    • /
    • v.16 no.3 s.76
    • /
    • pp.409-414
    • /
    • 2006
  • Molecular chaperones and folding enzymes in the endoplasmic reticulum (ER) associate with the newly synthesized proteins to prevent their aggregation and help them fold and assemble correctly. Chaperone function of BiP, which is a Hsp70 homologue in ER, is controlled by the N-terminal ATPase domain. The ATPase activity of the ATPase domain is affected by regulatory factors. BAP was identified as a nucleotide exchange factor of BiP (Grp78), which exchanges ADP with ATP in the ATPase domain of BiP This study presents whether BAP can influence folding of a protein, immunoglobulin heavy chain that is bound to BiP tightly. We first examined which nucleotide of ADP and ATP affects on BAP binding to BiP The data showed that endogenous BAP of HEK293 cells prefers ADP for binding to BiP in vitro, suggesting that BAP first releases ADP from the ATPase domain in order to exchange with ATP. Immunoglobulin heavy chain, an unfolded protein substrate, was released from BiP in the presence of BAP but not in the presence of ERdj3, which is another regulatory factor for BiP accelerating the rate of ATP hydrolysis of BiP The ADP-releasing function of BAP was, therefore, believed to be responsible for immunoglobulin heavy chain release from BiP. Grp170, another Hsp70 homologue in ER, did not co-precipited with BAP from $[^{35}S]$-metabolic labeled HEK293 lysate containing both overexpressed Grp170 and BAP. These data suggested that BAP has no specificity to Grp170 although the ATPase domains of Grp170 and BiP are homologous each other.

The GABAB receptor associates with regulators of G-protein signaling 4 protein in the mouse prefrontal cortex and hypothalamus

  • Kim, Gyeongwha;Jung, Soonwoong;Son, Hyeonwi;Kim, Sujeong;Choi, Jungil;Lee, Dong Hoon;Roh, Gu Seob;Kang, Sang Soo;Cho, Gyeong Jae;Choi, Wan Sung;Kim, Hyun Joon
    • BMB Reports
    • /
    • v.47 no.6
    • /
    • pp.324-329
    • /
    • 2014
  • Regulators of G-protein signaling (RGS) proteins regulate certain G-protein-coupled receptor (GPCR)-mediated signaling pathways. The GABAB receptor ($GABA_BR$) is a GPCR that plays a role in the stress response. Previous studies indicate that acute immobilization stress (AIS) decreases RGS4 in the prefrontal cortex (PFC) and hypothalamus (HY) and suggest the possibility of a signal complex composed of RGS4 and $GABA_BR$. Therefore, in the present study, we tested whether RGS4 associates with $GABA_BR$ in these brain regions. We found the co-localization of RGS4 and $GABA_BR$ subtypes in the PFC and HY using double immunohistochemistry and confirmed a direct association between $GABA_{B2}R$ and RGS4 proteins using co-immunoprecipitation. Furthermore, we found that AIS decreased the amount of RGS4 bound to $GABA_{B2}R$ and the number of double-positive cells. These results indicate that $GABA_BR$ forms a signal complex with RGS4 and suggests that RGS4 is a regulator of $GABA_BR$.

Kinesin Superfamily-associated Protein 3 (KAP3) Mediates the Interaction between Kinesin-II Motor Subunits and HS-1-associated Protein X-1 (HAX-1) through Direct Binding (Kinesin superfamily-associated protein 3 (KAP3)를 통한 HS-1-associated protein X-1 (HAX-1)과 Kinesin-II의 결합)

  • Jang, Won Hee;Seog, Dae-Hyun
    • Journal of Life Science
    • /
    • v.23 no.8
    • /
    • pp.978-983
    • /
    • 2013
  • Kinesin-II, a molecular motor, consists of two different motor subunits, KIF3A and KIF3B, and one large kinesin superfamily-associated protein 3 (KAP3), forming a heterotrimeric complex. KAP3 is associated with the tail domains of motor subunits. However, its exact role remains unclear. Here, we demonstrated KAP3 binding to the carboxyl (C)-terminal tail region of HS-associated protein X-1 (HAX-1). HAX-1 bound to the C-terminal region of KAP3, but not to KIFs (KIF3A, KIF3B, and KIF5B) and the kinesin light chain (KLC) in the yeast two-hybrid assays. The interaction was further confirmed in the glutathione S-transferase (GST) pull-down assay and by co-immunoprecipitation. Anti- HAX-1 antibody as well as anti-KIF3A antibody co-immunoprecipitated KIF3B and KAP3 from mouse brain extracts. These results suggest that KAP3 could mediate the interaction between Kinesin-II and HAX-1.

APP Tail 1 (PAT1) Interacts with Kinesin Light Chains (KLCs) through the Tetratricopeptide Repeat (TPR) Domain (APP tail 1 (PAT1)과 kinesin light chains (KLCs)의 tetratricopeptide repeat (TPR) domain을 통한 결합)

  • Jang, Won Hee;Kim, Sang-Jin;Jeong, Young Joo;Jun, Hee Jae;Moon, Il Soo;Seog, Dae-Hyun
    • Journal of Life Science
    • /
    • v.22 no.12
    • /
    • pp.1608-1613
    • /
    • 2012
  • A conventional kinesin, KIF5/Kinesin-I, transports various cargoes along the microtubule through interaction between its light chain subunit and the cargoes. Kinesin light chains (KLCs) interact with many different cargoes using their tetratricopeptide repeat (TPR) domain, but the mechanism underlying recognition and binding of a specific cargo has not yet been completely elucidated. We used the yeast two-hybrid assay to identify proteins that interact with the TPR domain of KLC1. We found an interaction between the TPR domain of KLC1 and an amyloid precursor protein (APP)-binding protein PAT1 (protein interacting with APP tail 1). The yeast two-hybrid assay demonstrated that the TPR domain-containing region of KLC1 mediated binding to the C-terminal tail region of PAT1. PAT1 also bound to KLC2 but not to kinesin heavy chains (KIF5A, KIF5B, and KIF5C) in the yeast two-hybrid assay. These protein-protein interactions were also observed in the glutathione S-transferase (GST) pull-down assay and by co-immunoprecipitation. Anti-PAT1 antibody as well as anti-APP anti-body co-immunoprecipitated KLC and KHCs associated with PAT1 from mouse brain extracts. These results suggest that PAT1 could mediate interactions between Kinesin-I and APP containing vesicles.

Interaction of GAT1 with Ubiquitin-Specific Protease Usp14 in Synaptic Terminal (GAT1과 ubiquitin-specific protease Usp14의 결합)

  • Seog, Dae-Hyun;Kim, Sang-Jin;Joung, Young-Ju;Yea, Sung-Su;Park, Yeong-Hong;Kim, Moo-Seong;Moon, Il-Soo;Jang, Won-Hee
    • Journal of Life Science
    • /
    • v.20 no.7
    • /
    • pp.1005-1011
    • /
    • 2010
  • $\gamma$-aminobutyric acid (GABA) is the major inhibitory neurotransmitter in the central nervous system. GABA transporters (GATs) control extracellular GABA levels by reuptake of released GABA from the synaptic cleft. However, how GATs are regulated has not yet been elucidated. Here, we used the yeast two-hybrid system to identify the specific binding protein(s) that interacts with the carboxyl (C)-terminal region of GAT1, the major isoform in the brain and find a specific interaction with the ubiquitin-specific protease 14 (Usp14), a deubiquitinating enzyme. Usp14 protein bound to the tail region of GAT1 and GAT2 but not to other GAT members in the yeast two-hybrid assay. The C-terminal region of Usp14 is essential for interaction with GAT1. In addition, these proteins showed specific interactions in the glutathione S-transferase (GST) pull-down assay. An antibody to GAT1 specifically co-immunoprecipitated Usp14 from mouse brain extracts. These results suggest that Usp14 may regulate the number of GAT1 at the cell surface.

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
    • /
    • v.45 no.3
    • /
    • 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.