• Title/Summary/Keyword: FADD

Search Result 39, Processing Time 0.034 seconds

Overexpression and Biological Characterization of the Death Domain Complex between TRADD and FADD

  • Hwang, Eun Young;Jeong, Mi Suk;Sung, Minkyung;Jang, Se Bok
    • Bulletin of the Korean Chemical Society
    • /
    • v.34 no.4
    • /
    • pp.1089-1095
    • /
    • 2013
  • The tumor necrosis factor-receptor 1 (TNFR1)-associated death domain protein (TRADD) contains an N-terminal TRAF binding domain and a C-terminal death domain. TRADD is known to interact directly with TNF receptor 2 (TNFR2) and the Fas-associated death domain protein (FADD), which are signal transducers that activate NF-${\kappa}B$ and induce apoptosis, respectively. To date, there has been no structural information on the TRADD and FADD death domain (DDs) complex. In this study, the death domains of TRADD and FADD were co-expressed and purified from Escherichia coli for structural characterization. We found that human TRADD (hTRADD) interacted strongly with mouse FADD (mFADD) via their DDs and interacted weakly with human FADD (hFADD)-DD. Moreover, the structures of the TRADD-DD:FADD-DD complexes were separately modeled from predicted structures in the protein data bank (PDB). The results of this study will have important applications in human diseases such as cancer, AIDS, degenerative and autoimmune diseases, and infectious diseases.

The roles of FADD in extrinsic apoptosis and necroptosis

  • Lee, Eun-Woo;Seo, Jin-Ho;Jeong, Man-Hyung;Lee, Sang-Sik;Song, Jae-Whan
    • BMB Reports
    • /
    • v.45 no.9
    • /
    • pp.496-508
    • /
    • 2012
  • Fas-associated protein with death domain (FADD), an adaptor that bridges death receptor signaling to the caspase cascade, is indispensible for the induction of extrinsic apoptotic cell death. Interest in the non-apoptotic function of FADD has greatly increased due to evidence that FADD-deficient mice or dominant-negative FADD transgenic mice result in embryonic lethality and an immune defect without showing apoptotic features. Numerous studies have suggested that FADD regulates cell cycle progression, proliferation, and autophagy, affecting these phenomena. Recently, programmed necrosis, also called necroptosis, was shown to be a key mechanism that induces embryonic lethality and an immune defect. Supporting these findings, FADD was shown to be involved in various necroptosis models. In this review, we summarize the mechanism of extrinsic apoptosis and necroptosis, and discuss the in vivo and in vitro roles of FADD in necroptosis induced by various stimuli.

Full-length Fas-associated Death Domain Protein Interacts with Short Form of Cellular FLICE Inhibitory Protein

  • Jeong, Mi-Suk;Jang, Se-Bok
    • Bulletin of the Korean Chemical Society
    • /
    • v.27 no.1
    • /
    • pp.87-92
    • /
    • 2006
  • Fas-associated death domain protein (FADD) recruits and activates procaspase-8 through interactions between the death effector domains of these two proteins. Cellular FLICE-inhibitory protein (c-FLIP) was identified as a molecule with sequence homology to caspase-8. It has been postulated that c-FLIP prevents formation of the competent death-inducing signaling complex in a ligand-dependent manner, through its interaction with FADD and/or caspase-8. However, the interaction of FADD and $c-FLIP_s$ (short form) in apoptosis signaling has been controversially discussed. We show the purification and the characterization of human full-length FADD and $c-FLIP_s$ expressed in Escherichia coli. The purified FADD and $c-FLIP_s$ are shown as homogeneity, respectively, in SDS-PAGE analysis and light-scattering measurements. The folding properties of the $\alpha$-helical structure of FADD and the super-secondary structure of $c-FLIP_s$ proteins were characterized by circular dichroism spectroscopy. Furthermore, we report here a series of biochemical and biophysical data for FADD-$c-FLIP_s$ binding in vitro. The binding of both FADD and $c-FLIP_s$ proteins was detected by BIAcore biosensor, fluorescence measurement, and size-exclusion column (SEC).

FADD Phosphorylation Modulates Blood Glucose Levels by Decreasing the Expression of InsulinDegrading Enzyme

  • Lin, Yan;Liu, Jia;Chen, Jia;Yao, Chun;Yang, Yunwen;Wang, Jie;Zhuang, Hongqin;Hua, Zi-Chun
    • Molecules and Cells
    • /
    • v.43 no.4
    • /
    • pp.373-383
    • /
    • 2020
  • Our previous study revealed a novel role of Fas-associated death domain-containing protein (FADD) in islet development and insulin secretion. Insulin-degrading enzyme (IDE) is a zinc metalloprotease that selectively degrades biologically important substrates associated with type 2 diabetes (T2DM). The current study was designed to investigate the effect of FADD phosphorylation on IDE. We found that the mRNA and protein levels of IDE were significantly downregulated in FADD-D mouse livers compared with control mice. Quantitative real-time polymerase chain reaction analysis showed that FADD regulates the expression of IDE at the transcriptional level without affecting the stability of the mRNA in HepG2 cells. Following treatment with cycloheximide, the IDE protein degradation rate was found to be increased in both FADD-D primary hepatocytes and FADD-knockdown HepG2 cells. Additionally, IDE expression levels were reduced in insulin-stimulated primary hepatocytes from FADD-D mice compared to those from control mice. Moreover, FADD phosphorylation promotes nuclear translocation of FoxO1, thus inhibiting the transcriptional activity of the IDE promoter. Together, these findings imply a novel role of FADD in the reduction of protein stability and expression levels of IDE.

The Solution Structure of FADD Death Domain: Structural Basis of Death Domain Interactions of Fas and FADD

  • Jeong, Euj-Jun;SookHee, Bang;Kim, Key-Sun
    • Proceedings of the Korean Biophysical Society Conference
    • /
    • 1999.06a
    • /
    • pp.21-21
    • /
    • 1999
  • A signal of Fas-mediated apoptosis is transferred through an adaptor protein FADD by interactions between death domains of Fas and FADD. To understand the signal transduction mechanism of Fas-mediated apoptosis, we solved the solution structure of a murine FADD death domain.(omitted)

  • PDF

Evidence of complex formation between FADD and c-FLIP death effector domains for the death inducing signaling complex

  • Hwang, Eun Young;Jeong, Mi Suk;Park, So Young;Jang, Se Bok
    • BMB Reports
    • /
    • v.47 no.9
    • /
    • pp.488-493
    • /
    • 2014
  • Adaptor protein FADD forms the death inducing signaling complex (DISC) by recruiting the initiating caspases-8 and -10 through homotypic death effector domain (DED) interactions. Cellular FLICE-inhibitory protein (c-FLIP) is an inhibitor of death ligand-induced apoptosis downstream of death receptors, and FADD competes with procaspase-8/10 for recruitment for DISC. However, the mechanism of action of FADD and c-FLIP proteins remain poorly understood at the molecular level. In this study, we provide evidence indicating that the death effector domain (DED) of FADD interacts directly with the death effector domain of human c-FLIP. In addition, we use homology modeling to develop a molecular docking model of FADD and c-FLIP proteins. We also find that four structure-based mutants (E80A, L84A, K169A and Y171A) of c-FLIP DEDs disturb the interaction with FADD DED, and that these mutations lower the stability of the c-FLIP DED.

Biological Activity of Tumor Necrosis Factor-α Secreted from Smooth Muscle Cell Overexpressing FADD (FADD 과발현 평활근세포에서 분비하는 Turner Necrosis Factor-α의 작용)

  • Kim, Sun-Mi;Lee, Kyeong-Ah;Kim, Koan-Hoi
    • Journal of Life Science
    • /
    • v.17 no.1 s.81
    • /
    • pp.45-50
    • /
    • 2007
  • This study investigated biological activity of tumor necrosis factor $(TNF)-\alpha$ secreted from smooth muscle cell (SMC) destined for death by expressing Fas associated death domain containing protein (FADD) (FADD-SMC) when the cells are grown without tetracycline in culture medium. In the absence of tetracycline the FADD-SMC secreted approximately 1000 pg/ml $TNF-\alpha$, whereas hardly detectable amount of the cytokine existed in the presence of tetracycline. The culture medium collected from the FADD-SMC grown in the absence of tetracycline increased phosphorylated form of p38 MAPK and up-regulated nuclear factor kappa B (NF-kB). The medium collected without tetracycline also caused death of L929 cells. Depletion of $TNF-\alpha$ with the soluble TNF receptor (sTNFR) inhibited the phosphorylation of p38 MAPK, the up-regulation of NF-kB activity and the death activity of the medium collected from FADD-SMC in the absence of tetracycline. These results indicate that $TNF-\alpha$ secreted from SMC undergoing death is biologically active and can affect cellular function.

Structural and Functional Roles of Caspase-8 in Extrinsic Apoptosis (Apoptosis의 외인성 경로에서 caspase-8의 구조적 및 기능적 역할)

  • Ha, Min Seon;Jeong, Mi Suk;Jang, Se Bok
    • Journal of Life Science
    • /
    • v.31 no.10
    • /
    • pp.954-959
    • /
    • 2021
  • Apoptosis is an important mechanism that regulates cellular populations to maintain homeostasis, and the caspases, a family of cysteine proteases, are key mediators of the apoptosis pathway. Caspase-8 is an initiator caspase of the extrinsic apoptotic pathway, which is initiated by extracellular stimuli. Caspase-8 have two conserved domains, N-terminal tandem death effector domains (DED) and C-terminal two catalytic domain, which are important for this extrinsic apoptosis pathway. In extrinsic apoptosis pathway, death receptors which members of TNF superfamily are activated by binding of death receptor specific ligands from cell outside. After the activated death receptors recruit adaptor protein Fas-associated death domain protein (FADD), death domains (DD) of death receptor and FADD bind to each other and FADD combined with death receptor recruits procaspase-8, a precursor form of caspase-8. The DED of FADD and procaspase-8 bind to one another and FADD-bound procaspase-8 is activated by cleavage of the prodomain. This death receptor-FADD-caspase-8 complex called death inducing signaling complex (DISC). Cellular FLICE-inhibitory proteins (c-FLIPs) regulate caspase-8 activation by acting both anti- and pro-apoptotically, and caspase-8 activation initiates the activation of executioner caspases such as caspase-3. Finally activated executioner caspases complete the apoptosis by acting critically DNA degradation, nuclear condensation, plasma membrane blebbing, and the proteolysis of certain caspase substrates.

Immunohistochemical Analysis of Fas-associated Death Domain Protein Expression in Stomach Cancers (위암의 Fas-associated Death Domain Protein 단백질의 발현)

  • Lee, Sug-Hyung;Lee, Jong-Woo;Park, Won-Sang;Lee, Jung-Young;Yoo, Nam-Jin
    • Journal of Gastric Cancer
    • /
    • v.3 no.2
    • /
    • pp.80-83
    • /
    • 2003
  • Purpose: Evidence exists that dysregulation of apoptosis is involved in the pathogenesis of cancer development. Fasassociated death domain (FADD) protein, an adaptor protein of death receptors, is a critical regulatory component of the extrinsic cell- death pathway that exerts its pro-apoptotic effect upon binding with death receptors. Expression of the FADD protein has not been reported in stomach cancer. The aim of this study was to explore the expression status of the FADD protein in stomach cancers. Materials and Methods: In the current study, we analyzed the expression of the FADD protein in 60 advanced stomach cancer by using immunohistochemistry and a tissue microarray approach. Results: Immunopositivity (defined as $\geq\30\%$) was observed for the FADD protein in 23 ($38\%$) of the 60 cancers. Normal gastric mucosal cells showed expression of the FADD protein. Conclusion: Taken together, these results indicate that decreased expression of the FADD protein is a frequent event in stomach cancers and suggest that to avoid apoptosis, stomach cancer cells in vivo may need loss of FADD expression, which might contribute to tumor development.

  • PDF

DED Interaction of FADD and Caspase-8 in the Induction of Apoptotic Cell Death

  • Park, Young-Hoon;Han, Chang Woo;Jeong, Mi Suk;Jang, Se Bok
    • Journal of Microbiology and Biotechnology
    • /
    • v.32 no.8
    • /
    • pp.1034-1040
    • /
    • 2022
  • Fas-associated death domain (FADD) is an adapter molecule that bridges the interaction between receptor-interacting protein 1 (RIP1) and aspartate-specific cysteine protease-8 (caspase-8). As the primary mediator of apoptotic cell death, caspase-8 has two N-terminal death-effector domains (DEDs) and it interacts with other proteins in the DED subfamily through several conserved residues. In the tumor necrosis receptor-1 (TNFR-1)-dependent signaling pathway, apoptosis is triggered by the caspase-8/FADD complex by stimulating receptor internalization. However, the molecular mechanism of complex formation by the DED proteins remains poorly understood. Here, we found that direct DED-DED interaction between FADD and caspase-8 and the structure-based mutations (Y8D/I128A, E12A/I128A, E12R/I128A, K39A/I128A, K39D/I128A, F122A/I128A, and L123A/I128A) of caspase-8 disrupted formation of the stable DED complex with FADD. Moreover, the monomeric crystal structure of the caspase-8 DEDs (F122A/I128A) was solved at 1.7 Å. This study will provide new insight into the interaction mechanism and structural characteristics between FADD and caspase-8 DED subfamily proteins.