398 results on '"Death-inducing signaling complex"'
Search Results
2. SOCS-1 ameliorates smoke inhalation-induced acute lung injury through inhibition of ASK-1 activity and DISC formation.
- Author
-
Zhang, Leifang, Xu, Chenming, Ma, Yating, Zhu, Kairui, Chen, Xiaoming, Shi, Qiwen, Su, Weike, and Zhao, Hang
- Subjects
- *
LUNG injuries , *SMOKE inhalation injuries , *CYTOKINES , *PATHOGENIC microorganisms , *EPITHELIAL cells - Abstract
Smoke inhalation leads to acute lung injury (ALI), a devastating clinical problem associated with high mortality. Suppressor of cytokine signaling-1 (SOCS-1) is a negative regulator of apoptosis and pro-inflammatory cytokine signaling, two major contributors to the pathogenesis of ALI. We have found that SOCS-1 protects lung epithelial cells from smoke-induced apoptosis through two mechanisms. One is that SOCS-1 enhances degradation of ASK-1 and diminishes cleavage of pro-caspase-3 to repress smoke-triggered apoptosis in lung epithelial cells. The other is that SOCS-1 represses smoke-triggered DISC formation through altering TRADD-caspase-8 interaction rather than TNFR-1-TRADD interaction or TNFR-1-TRAF-2 interaction. In conclusion, SOCS-1 relieves smoke inhalation-induced lung injury by repressing ASK-1 and DISC-mediated epithelium apoptosis. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
3. 23-Hydroxyursolic Acid Isolated from the Stem Bark of Cussonia bancoensis Induces Apoptosis through Fas/Caspase-8-Dependent Pathway in HL-60 Human Promyelocytic Leukemia Cells
- Author
-
Jong-Heon Won, Kyung-Sook Chung, Eun-Young Park, Jeong-Hun Lee, Jung-Hye Choi, Leon Azefack Tapondjou, Hee-Juhn Park, Masaaki Nomura, Ahmed H.E. Hassan, and Kyung-Tae Lee
- Subjects
23-hydroxyursolic acid ,apoptosis ,caspase ,Bcl-2 ,mitochondria ,death-inducing signaling complex ,Organic chemistry ,QD241-441 - Abstract
The natural product 23-hydroxyursolic acid (23-HUA) is a derivative of ursolic acid, which is known to induce cancer cell apoptosis. However, apoptotic effects and mechanisms of 23-HUA have not been well characterized yet. Herein, we investigated the molecular mechanisms of 23-HUA-induced apoptosis in HL-60 human promyelocytic leukemia cells. 23-HUA-treated HL-60 cells showed apoptotic features including internucleosomal DNA condensation and fragmentation as well as externalization of phosphatidylserine residues. 23-HUA induced a series of mitochondrial events including disruption of mitochondrial membrane potential (ΔΨm), cytochrome c and Smac/DIABLO release and loss of balance between pro-apoptotic and anti-apoptotic Bcl-2 proteins in HL-60 cells. In addition, 23-HUA activated caspase-8, caspase-9 and caspase-3. Pretreatment with a broad caspase inhibitor (z-VAD-fmk), a caspase-3 inhibitor (z-DEVD-fmk), and a caspase-8 inhibitor (z-IETD-fmk) significantly attenuated 23-HUA-induced DNA fragmentation. After 23-HUA-induced apoptosis, proteins expression levels of FasL, Fas and FADD constituting the death-inducing signaling complex (DISC) were upregulated in HL-60 cells. Moreover, transfection with Fas or FADD siRNA significantly blocked 23-HUA-induced DNA fragmentation and caspases activation. Taken together, these findings indicate that 23-HUA induces apoptosis in HL-60 human promyelocytic leukemia cells through formation of DISC and caspase-8 activation leading to loss of ΔΨm and caspase-3 activation.
- Published
- 2018
- Full Text
- View/download PDF
4. Efficient recruitment of c‐FLIP L to the death‐inducing signaling complex leads to Fas resistance in natural killer‐cell lymphoma
- Author
-
Azuchi Masuda, Mayumi Yoshimori, Koichi Sugimoto, Yasushi Isobe, Ayako Arai, and Norio Komatsu
- Subjects
0301 basic medicine ,Cancer Research ,Programmed cell death ,Chemistry ,General Medicine ,medicine.disease ,Caspase 8 ,Extranodal NK/T-cell lymphoma, nasal type ,Jurkat cells ,Fas ligand ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Oncology ,Apoptosis ,Cell culture ,030220 oncology & carcinogenesis ,Death-inducing signaling complex ,medicine ,Cancer research - Abstract
Activation-induced cell death (AICD) mediated by the Fas/Fas ligand (FasL) system plays a key role in regulating immune response. Although normal natural killer (NK) cells use this system for their homeostasis, malignant NK cells seem to disrupt the process. Extranodal NK/T-cell lymphoma, nasal type (ENKL) is a rare but fatal disease, for which novel therapeutic targets need to be identified. We confirmed that ENKL-derived NK cell lines NK-YS and Hank1, and primary lymphoma cells expressed procaspase-8/FADD-like interleukin-1β-converting enzyme (FLICE) modulator and cellular FLICE-inhibitory protein (c-FLIP), along with Fas and FasL. Compared with Fas-sensitive Jurkat cells, NK-YS and Hank1 showed resistance to Fas-mediated apoptosis in spite of the same expression levels of c-FLIP and the death-inducing signaling complex (DISC) formation. Unexpectedly, the long isoform of c-FLIP (c-FLIPL ) was coimmunoprecipitated with Fas predominantly in both ENKL-derived NK cell lines after Fas ligation. Indeed, c-FLIPL was more sufficiently recruited to the DISC in both ENKL-derived NK cell lines than in Jurkat cells after Fas ligation. Knockdown of c-FLIPL per se enhanced autonomous cell death and restored the sensitivity to Fas in both NK-YS and Hank1 cells. Although ENKL cells are primed for AICD, they constitutively express and efficiently utilize c-FLIPL , which prevents their Fas-mediated apoptosis. Our results show that c-FLIPL could be a promising therapeutic target against ENKL.
- Published
- 2020
5. Death-Inducing Signaling Complex
- Author
-
Vohr, Hans-Werner, editor
- Published
- 2016
- Full Text
- View/download PDF
6. Association of p38MAPK‐p53‐Fas aggregation in S‐allyl cysteine mediated regulation of hepatocarcinoma
- Author
-
Kaustav Dutta Chowdhury, Gobinda Chandra Sadhukhan, Sujan Chatterjee, Udipta Chakraborti, Pujita Ghosh, Anupam Basu, Dipanwita Sengupta, and Debajyoti Patra
- Subjects
Male ,Fas Ligand Protein ,Pyridines ,Health, Toxicology and Mutagenesis ,p38 mitogen-activated protein kinases ,Cell ,S-Allyl cysteine ,Antineoplastic Agents ,010501 environmental sciences ,Management, Monitoring, Policy and Law ,Toxicology ,p38 Mitogen-Activated Protein Kinases ,01 natural sciences ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,Liver Neoplasms, Experimental ,0302 clinical medicine ,medicine ,Animals ,Humans ,Cysteine ,fas Receptor ,Phosphorylation ,Protein Kinase Inhibitors ,Caspase ,0105 earth and related environmental sciences ,biology ,Chemistry ,Cell growth ,Imidazoles ,Hep G2 Cells ,General Medicine ,medicine.anatomical_structure ,Apoptosis ,Caspases ,030220 oncology & carcinogenesis ,Death-inducing signaling complex ,biology.protein ,Cancer research ,Tumor Suppressor Protein p53 - Abstract
Bioactive components of dietary phytochemicals have been reported to possess antitumor activities. Evidences suggested key role of stress responsive p38MAPK in the induction of nutraceuticals mediated apoptosis in hepatocellular carcinoma (HCC). Current study demonstrated detailed molecular bagatelle associated with p38 MAPK mediated effective suppression of cell growth both in HepG2 and chemically induced liver carcinoma after S-allyl cysteine (SAC) treatment. SAC promoted p38MAPK activity responsible for p53 phosphorylation, its stabilization followed by nuclear translocation leading to induction in expression and oligomerization of Fas protein. Distinctive p38MAPK-p53 axis dependent Fas-FasL-FADD mediated caspase activities along with perturbed cell cycling became normalized with continuation of SAC treatment for another month to diethylnitrosamine induced liver carcinoma. Co-treatment with SB203580, the p38MAPK inhibitor, prevented pro-apoptotic effect of SAC by altering p53 phosphorylation and death inducing signaling complex conformation in HepG2 and induced HCC. Collectively study suggested significant contribution of p38MAPK-p53-DISC-Caspase pathway in the regulation of anti-neoplastic activity of SAC against HCC.
- Published
- 2019
7. C-27-carboxylated oleanane triterpenoids up-regulate TRAIL DISC assembly via p38 MAPK and CHOP-mediated DR5 expression in human glioblastoma cells
- Author
-
Gang Min Hur, InWha Park, MinKyun Na, So-Ra Lee, Jin Bong Park, Wei Zhou, Xuezhe Piao, Kidong Kang, Taeg Kyu Kwon, and Hee Sun Byun
- Subjects
0301 basic medicine ,p38 mitogen-activated protein kinases ,Cell ,Caspase 8 ,p38 Mitogen-Activated Protein Kinases ,Biochemistry ,TNF-Related Apoptosis-Inducing Ligand ,03 medical and health sciences ,Transactivation ,Downregulation and upregulation ,medicine ,Humans ,Oleanolic Acid ,Pharmacology ,Plant Extracts ,Chemistry ,Up-Regulation ,Gene Expression Regulation, Neoplastic ,Receptors, TNF-Related Apoptosis-Inducing Ligand ,030104 developmental biology ,medicine.anatomical_structure ,Death-inducing signaling complex ,Cancer research ,Tumor necrosis factor alpha ,Glioblastoma ,Transcription Factor CHOP ,Intracellular - Abstract
Despite recent tremendous progress, targeting of TNF-related apoptosis-inducing ligand (TRAIL) as a cancer therapy has limited success in many clinical trials, in part due to inactivation of death inducing signaling complex (DISC)-mediated caspase-8 signaling cascade in highly malignant tumors such as glioblastoma. In this study, screening of constituents derived from Astilbe rivularis for TRAIL-sensitizing activity identified C-27-carboxylated oleanolic acid derivatives (C27OAs) including 3β-hydroxyolean-12-en-27-oic acid (C27OA-1), 3β,6β,7α-trihydroxyolean-12-en-27-oic acid (C27OA-2), and 3β-trans-p-coumaroyloxy-olean-12-en-27-oic acid (C27OA-3) as novel TRAIL sensitizers. Interestingly, these C27OAs did not affect apoptotic cell death induced by either ligation of other death receptor (DR) types, such as TNF and Fas or DNA damaging agents, which suggests that C27OAs effectively and selectively sensitize TRAIL-mediated caspase-8 activation. Mechanistically, C27OAs upregulate the expression of cell surface DR5 and DISC formation without affecting downstream intracellular apoptosis-related proteins. The upregulation of DR5 expression by C27OAs strictly depends on transactivation of C/EBP homology protein, which is regulated through the p38 MAPK pathway, rather than p53 and intracellular reactive oxygen species status. Taken together, our results identify the novel C27OAs as TRAIL sensitizers targeting the upstream DISC assembly of DR5, and provide a rationale for further development of C27OAs for facilitating TRAIL-based chemotherapy in glioblastoma patients.
- Published
- 2018
8. Nanoscale FasL Organization on DNA Origami to Decipher Apoptosis Signal Activation in Cells
- Author
-
Ricarda M L Berger, Cornelia Monzel, Tim Liedl, Oliver Hill, Simon M. Kempe, Amelie Heuer-Jungemann, Johann M. Weck, and Joachim O. Rädler
- Subjects
Cell signaling ,Cell ,Apoptosis ,02 engineering and technology ,010402 general chemistry ,01 natural sciences ,Fas ligand ,Biomaterials ,medicine ,DNA origami ,General Materials Science ,fas Receptor ,Receptor ,Chemistry ,General Chemistry ,DNA ,021001 nanoscience & nanotechnology ,0104 chemical sciences ,Cell biology ,Coupling (electronics) ,medicine.anatomical_structure ,Death-inducing signaling complex ,0210 nano-technology ,Carrier Proteins ,Biotechnology ,Signal Transduction - Abstract
Cell signaling is initiated by characteristic protein patterns in the plasma membrane, but tools to decipher their molecular organization and activation are hitherto lacking. Among the well-known signaling pattern is the death inducing signaling complex with a predicted hexagonal receptor architecture. To probe this architecture, DNA origami-based nanoagents with nanometer precise arrangements of the death receptor ligand FasL are introduced and presented to cells. Mimicking different receptor geometries, these nanoagents act as signaling platforms inducing fastest time-to-death kinetics for hexagonal FasL arrangements with 10 nm inter-molecular spacing. Compared to naturally occurring soluble FasL, this trigger is faster and 100x more efficient. Nanoagents with different spacing, lower FasL number or higher coupling flexibility impede signaling. The results present DNA origami as versatile signaling scaffolds exhibiting unprecedented control over molecular number and geometry. They define molecular benchmarks in apoptosis signal initiation and constitute a new strategy to drive particular cell responses.
- Published
- 2021
9. Functional interaction between hMYH and hTRADD in the TNF-α-mediated survival and death pathways of HeLa cells.
- Author
-
Vy Tran, An Hue, Hahm, Soo-Hyun, Han, Se Hee, Chung, Ji Hyung, Park, Geon Tae, and Han, Ye Sun
- Subjects
- *
HELA cells , *TUMOR necrosis factors , *IMMUNE system , *APOPTOSIS , *CELLULAR signal transduction , *CHROMOSOMAL translocation , *RECEPTOR-interacting proteins - Abstract
The tumor necrosis factor (TNF) signaling pathway is a classical immune system pathway that plays a key role in regulating cell survival and apoptosis. The TNF receptor-associated death domain (TRADD) protein is recruited to the death domain of TNF receptor 1 (TNFR1), where it interacts with TNF receptor-associated factor 2 (TRAF2) and receptor-interacting protein (RIP) for the induction of apoptosis, necrosis, nuclear factor kappa-light-chain-enhancer of activated B cells (NFκB), and mitogen-activated protein (MAP) kinase activation. In this study, we found that the human MutY homolog (hMYH) interacted with human TRADD (hTRADD) via the C-terminal domain of hMYH. Moreover, under conditions promoting TNF-α-induced cell death or survival in HeLa cells, this interaction was weakened or enhanced, respectively. The interaction between hMYH and hTRADD was important for signaling pathways mediated by TNF-α. Our results also suggested that the hTRADD–hMYH association was involved in the nuclear translocation of NFκB and formation of the TNFR1–TRADD complex. Thus, this study identified a novel mechanism through which the hMYH–hTRADD interaction may affect the TNF-α signaling pathway. Implications In HeLa cells, the hTRADD–hMYH interaction functioned in both cell survival and apoptosis pathways following TNF-α stimulation. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
10. Death effecter domain for the assembly of death-inducing signaling complex.
- Author
-
Yang, Jin
- Abstract
Death-inducing signaling complex (DISC) is a platform for the activation of initiator caspase in extrinsic apoptosis. Assembly of DISC is accomplished by two different types of homotypic interaction: one is between death domains (DDs) of a death receptor and FADD, and the other is between death effecter domains (DEDs) of FADD, procaspase-8/-10 and cFLIP. Recent biochemical investigations on the stoichiometry of DISC have revealed that single-DED-containing FADD exists in DISC in a substantially lower abundance than the sum of tandem-DEDs-containing components that are procaspase-8 and cFLIP. In addition, the homology models of the tandem DEDs in procaspase-8 and cFLIP show that two different interaction faces, H1-H4 face and H2-H5 face, are exposed for possible inter-molecular DED-DED interactions. These recent findings led to a proposal of the DED chain model for the interactions between FADD, procaspase-8 and cFLIP in DISC. This emerging view provides new insights on the topology of DED-DED network in DISC and furthermore on how procaspase-8 and cFLIP cluster for dimerization and proteolytic activation. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
11. Reconstruction of the Fas-Based Death-Inducing Signaling Complex (DISC) Using a Protein–Protein Docking Meta-Approach
- Author
-
Melissa Thomas, Leif A. Eriksson, and Sayyed Jalil Mahdizadeh
- Subjects
Death Domain Receptor Signaling Adaptor Proteins ,Multiprotein complex ,Fas-Associated Death Domain Protein ,General Chemical Engineering ,CASP8 and FADD-Like Apoptosis Regulating Protein ,Apoptosis ,Library and Information Sciences ,Article ,03 medical and health sciences ,0302 clinical medicine ,Protein Interaction Mapping ,Macromolecular docking ,FADD ,030304 developmental biology ,Death domain ,Caspase 8 ,0303 health sciences ,biology ,Chemistry ,Cell Membrane ,General Chemistry ,Fas receptor ,Computer Science Applications ,Cell biology ,Docking (molecular) ,030220 oncology & carcinogenesis ,Death-inducing signaling complex ,biology.protein ,Death effector domain ,Signal Transduction - Abstract
The death-inducing signaling complex (DISC) is a fundamental multiprotein complex, which triggers the extrinsic apoptosis pathway through stimulation by death ligands. DISC consists of different death domain (DD) and death effector domain (DED) containing proteins such as the death receptor Fas (CD95) in complex with FADD, procaspase-8, and cFLIP. Despite many experimental and theoretical studies in this area, there is no global agreement neither on the DISC architecture nor on the mechanism of action of the involved species. In the current work, we have tried to reconstruct the DISC structure by identifying key protein interactions using a new protein–protein docking meta-approach. We combined the benefits of five of the most employed protein–protein docking engines, HADDOCK, ClusPro, HDOCK, GRAMM-X, and ZDOCK, in order to improve the accuracy of the predicted docking complexes. Free energy of binding and hot spot interacting residues were calculated and determined for each protein–protein interaction using molecular mechanics generalized Born surface area and alanine scanning techniques, respectively. In addition, a series of in-cellulo protein-fragment complementation assays were conducted to validate the protein–protein docking procedure. The results show that the DISC formation initiates by dimerization of adjacent FasDD trimers followed by recruitment of FADD through homotypic DD interactions with the oligomerized death receptor. Furthermore, the in-silico outcomes indicate that cFLIP cannot bind directly to FADD; instead, cFLIP recruitment to the DISC is a hierarchical and cooperative process where FADD initially recruits procaspase-8, which in turn recruits and heterodimerizes with cFLIP. Finally, a possible structure of the entire DISC is proposed based on the docking results.
- Published
- 2021
- Full Text
- View/download PDF
12. Cell-Penetrable Peptide-Conjugated FADD Induces Apoptosis and Regulates Inflammatory Signaling in Cancer Cells
- Author
-
Chandramani Pathak, Kishu Ranjan, Bhargav N. Waghela, and Foram U. Vaidya
- Subjects
Programmed cell death ,Fas-Associated Death Domain Protein ,Cell-Penetrating Peptides ,urologic and male genital diseases ,Catalysis ,Article ,NF-κB ,Inorganic Chemistry ,lcsh:Chemistry ,Transactivation ,Mice ,Neoplasms ,Animals ,Humans ,cancer ,FADD ,Physical and Theoretical Chemistry ,Molecular Biology ,lcsh:QH301-705.5 ,Spectroscopy ,Death domain ,biology ,Chemistry ,Organic Chemistry ,apoptosis ,General Medicine ,Hep G2 Cells ,HCT116 Cells ,Computer Science Applications ,Cell biology ,Neoplasm Proteins ,HEK293 Cells ,RAW 264.7 Cells ,Gene Expression Regulation ,lcsh:Biology (General) ,lcsh:QD1-999 ,Apoptosis ,inflammation ,Cancer cell ,Death-inducing signaling complex ,biology.protein ,MCF-7 Cells ,peptides ,Tumor necrosis factor alpha ,biological phenomena, cell phenomena, and immunity ,HeLa Cells - Abstract
Dysregulated expression of Fas-associated death domain (FADD) is associated with the impediment of various cellular pathways, including apoptosis and inflammation. The adequate cytosolic expression of FADD is critical to the regulation of cancer cell proliferation. Importantly, cancer cells devise mechanisms to suppress FADD expression and, in turn, escape from apoptosis signaling. Formulating strategies, for direct delivery of FADD proteins into cancer cells in a controlled manner, may represent a promising therapeutic approach in cancer therapy. We chemically conjugated purified FADD protein with cell permeable TAT (transactivator of transcription) peptide, to deliver in cancer cells. TAT-conjugated FADD protein internalized through the caveolar pathway of endocytosis and retained in the cytosol to augment cell death. Inside cancer cells, TAT-FADD rapidly constituted DISC (death inducing signaling complex) assembly, which in turn, instigate apoptosis signaling. The apoptotic competency of TAT-FADD showed comparable outcomes with the conventional apoptosis inducers. Notably, TAT-FADD mitigates constitutive NF-&kappa, B activation and associated downstream anti-apoptotic genes Bcl2, cFLIPL, RIP1, and cIAP2, independent of pro-cancerous TNF-&alpha, priming. In cancer cells, TAT-FADD suppresses the canonical NLRP3 inflammasome priming and restricts the processing and secretion of proinflammatory IL-1&beta, Our results demonstrate that TAT-mediated intracellular delivery of FADD protein can potentially recite apoptosis signaling with simultaneous regulation of anti-apoptotic and proinflammatory NF-&kappa, B signaling activation in cancer cells.
- Published
- 2020
13. An engineered construct of cFLIP provides insight into DED1 structure and interactions
- Author
-
Dale F. Mierke, Tamar Basiashvili, Alexandra E. Panaitiu, and Maria Pellegrini
- Subjects
Models, Molecular ,Magnetic Resonance Spectroscopy ,Calmodulin ,Fas-Associated Death Domain Protein ,CASP8 and FADD-Like Apoptosis Regulating Protein ,Context (language use) ,Protein Engineering ,Article ,Protein Structure, Secondary ,Protein Domains ,Structural Biology ,Protein Interaction Maps ,FADD ,Molecular Biology ,Death domain ,Binding Sites ,biology ,Effector ,Chemistry ,Circular Dichroism ,Cell biology ,Death-inducing signaling complex ,biology.protein ,Death effector domain ,Function (biology) ,Protein Binding - Abstract
Cellular FLICE-like inhibitory protein (cFLIP) is a member of the Death Domain superfamily with pivotal roles in many cellular processes and disease states, including cancer and autoimmune disorders. In the context of the death-inducing signaling complex (DISC), cFLIP isoforms regulate extrinsic apoptosis by controlling procaspase-8 activation. The function of cFLIP is mediated through a series of protein-protein interactions, engaging the two N-terminal death effector domains (DEDs). Here, we solve the structure of an engineered DED1 domain of cFLIP using solution nuclear magnetic resonance (NMR) and we define the interaction with FADD and calmodulin, protein-protein interactions that regulate the function of cFLIP in the DISC. cFLIP DED1 assumes a canonical DED fold characterized by six α helices and is able to bind calmodulin and FADD through two separate interfaces. Our results clearly demonstrate the role of DED1 in the cFLIP/FADD association and contribute to the understanding of the assembly of DISC filaments.
- Published
- 2022
14. Biomol NMR Assign
- Author
-
Gavin W. Collie, Gilles Guichard, Cameron D. Mackereth, Antoine Baudin, Antoine Loquet, Stéphane Chaignepain, Anne Guichard, Benoit Odaert, Mélanie Berbon, Agnès Hocquellet, Sabrina Rousseau, Chimie et Biologie des Membranes et des Nanoobjets (CBMN), École Nationale d'Ingénieurs des Travaux Agricoles - Bordeaux (ENITAB)-Institut de Chimie du CNRS (INC)-Université de Bordeaux (UB)-Centre National de la Recherche Scientifique (CNRS), Santé publique et épidémiologie des déterminants professionnels et sociaux de la santé, Epidémiologie, sciences sociales, santé publique (IFR 69), Université Paris 1 Panthéon-Sorbonne (UP1)-Université Paris-Sud - Paris 11 (UP11)-École des hautes études en sciences sociales (EHESS)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université Paris 1 Panthéon-Sorbonne (UP1)-Université Paris-Sud - Paris 11 (UP11)-École des hautes études en sciences sociales (EHESS)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Versailles Saint-Quentin-en-Yvelines (UVSQ), Institut Européen de Chimie et Biologie, Biotechnologie des protéines recombinantes à visée santé, Université de Bordeaux (UB)-Institut Polytechnique de Bordeaux, Institut de biologie et chimie des protéines [Lyon] (IBCP), Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Centre National de la Recherche Scientifique (CNRS), Acides Nucléiques : Régulations Naturelle et Artificielle (ARNA), Université de Bordeaux (UB)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Institut de Chimie du CNRS (INC)-Centre National de la Recherche Scientifique (CNRS), Immunologie et chimie thérapeutiques (ICT), Cancéropôle du Grand Est-Centre National de la Recherche Scientifique (CNRS), ARNA - Acides Nucléiques : Régulations Naturelle et Artificielle, Ligue Contre le Cancer, Université de Bordeaux (UB)-École Nationale d'Ingénieurs des Travaux Agricoles - Bordeaux (ENITAB)-Institut de Chimie du CNRS (INC)-Centre National de la Recherche Scientifique (CNRS), and Université Paris 1 Panthéon-Sorbonne (UP1)-Université Paris-Sud - Paris 11 (UP11)-École des hautes études en sciences sociales (EHESS)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université Paris 1 Panthéon-Sorbonne (UP1)-Université Paris-Sud - Paris 11 (UP11)-École des hautes études en sciences sociales (EHESS)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université de Versailles Saint-Quentin-en-Yvelines (UVSQ)-Institut National de la Santé et de la Recherche Médicale (INSERM)
- Subjects
0301 basic medicine ,Programmed cell death ,TRAIL ,Apoptosis ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,Biochemistry ,03 medical and health sciences ,NMR spectroscopy ,0302 clinical medicine ,[SDV.SP.MED]Life Sciences [q-bio]/Pharmaceutical sciences/Medication ,Structural Biology ,Secondary structure ,DR5 ,Receptor ,Protein secondary structure ,ComputingMilieux_MISCELLANEOUS ,Cancer ,[SDV.BBM.BS]Life Sciences [q-bio]/Biochemistry, Molecular Biology/Structural Biology [q-bio.BM] ,Chemistry ,[CHIM.MATE]Chemical Sciences/Material chemistry ,Nuclear magnetic resonance spectroscopy ,Ligand (biochemistry) ,3. Good health ,Cell biology ,030104 developmental biology ,Resonance assignment ,030220 oncology & carcinogenesis ,Death-inducing signaling complex ,Cancer cell - Abstract
Death receptors (DR) selectively drive cancer cells to apoptosis upon binding to the Tumor necrosis factor-a-Related Apoptosis-Inducing Ligand (TRAIL). Complex formation induces the oligomerization of the death receptors DR4 (TRAIL-R1) and DR5 (TRAIL-R2) and transduces the apoptogenic signal to their respective death domains, leading to Death Inducing Signaling Complex (DISC) formation, caspase activation and ultimately cell death. Several crystal structures of the ExtraCellular Domain from Death Receptor 5 (DR5-ECD) have been reported in complex with the TRAIL ligand or anti-DR5 antibodies, but none for the isolated protein. In order to fill this gap and to perform binding experiments with TRAIL peptidomimetics, we have produced isotopically labelled DR5-ECD and started a conformational analysis by using high-field 3D NMR spectroscopy. Herein, we present the first resonance assignment of a TRAIL receptor in solution and the determination of its secondary structure from NMR chemical shifts.
- Published
- 2018
15. K6 linked polyubiquitylation of FADD by CHIP prevents death inducing signaling complex formation suppressing cell death
- Author
-
Jihye Shin, Young Woo Nam, Jin-Ho Seo, Eun-Woo Lee, Manhyung Jeong, Seon Hyeong Lee, Daehyeon Seong, Jaewhan Song, and Cheolju Lee
- Subjects
0301 basic medicine ,Cancer Research ,Programmed cell death ,Fas-Associated Death Domain Protein ,Ubiquitin-Protein Ligases ,Mice, Nude ,Apoptosis ,urologic and male genital diseases ,Caspase 8 ,Jurkat cells ,Fas ligand ,TNF-Related Apoptosis-Inducing Ligand ,Jurkat Cells ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,Genetics ,Animals ,Humans ,FADD ,Molecular Biology ,Death domain ,Mice, Inbred BALB C ,Cell Death ,biology ,Ubiquitination ,Signal transducing adaptor protein ,Cell biology ,HEK293 Cells ,030104 developmental biology ,030220 oncology & carcinogenesis ,Death-inducing signaling complex ,MCF-7 Cells ,biology.protein ,Female ,biological phenomena, cell phenomena, and immunity ,Apoptosis Regulatory Proteins ,HeLa Cells ,Signal Transduction - Abstract
Fas-associated death domain (FADD) is an adaptor protein recruiting complexes of caspase 8 to death ligand receptors to induce extrinsic apoptotic cell death in response to a TNF superfamily member. Although, formation of the complex of FADD and caspase 8 upon death stimuli has been studied in detail, posttranslational modifications fine-tuning these processes have yet to be identified. Here we revealed that K6-linked polyubiquitylation of FADD on lysines 149 and 153 mediated by C terminus HSC70-interacting protein (CHIP) plays an important role in preventing formation of the death inducing signaling complex (DISC), thus leading to the suppression of cell death. Cells depleted of CHIP showed higher sensitivity toward death ligands such as FasL and TRAIL, leading to upregulation of DISC formation composed of a death receptor, FADD, and caspase 8. CHIP was able to bind to FADD, induce K6-linked polyubiquitylation of FADD, and suppress DISC formation. By mass spectrometry, lysines 149 and 153 of FADD were found to be responsible for CHIP-mediated FADD ubiquitylation. FADD mutated at these sites was capable of more potent cell death induction as compared with the wild type and was no longer suppressed by CHIP. On the other hand, CHIP deficient in E3 ligase activity was not capable of suppressing FADD function and of FADD ubiquitylation. CHIP depletion in ME-180 cells induced significant sensitization of these cells toward TRAIL in xenograft analyses. These results imply that K6-linked ubiquitylation of FADD by CHIP is a crucial checkpoint in cytokine-dependent extrinsic apoptosis.
- Published
- 2018
16. FBXO7 triggers caspase 8-mediated proteolysis of the transcription factor FOXO4 and exacerbates neuronal cytotoxicity
- Author
-
Yun Ju Lee, Minju Hyun, Kwang Chul Chung, Sungyeon Jung, and Su Hyoun Lee
- Subjects
PEI, polyethylenimine ,Male ,HEK293, human embryonic kidney 293 ,Cell Cycle Proteins ,ECL, enhanced chemiluminescence ,Biochemistry ,DMEM, Dulbecco’s modified Eagle’s medium ,Mice ,caspase 8 ,DISC, death-inducing signaling complex ,FADD, FAS-associated death domain ,Mitophagy ,FADD ,Caspase ,Neurons ,6-OHDA, 6-hydroxydopamine ,biology ,Chemistry ,MPP+, 1-methyl-4-phenylpyridium ion ,FOXO4 ,HRP, horseradish peroxidase ,FOXO Family ,Forkhead Transcription Factors ,Parkinson Disease ,Cell biology ,Ubiquitin ligase ,Death-inducing signaling complex ,MCF-7 Cells ,AD, Alzheimer's disease ,Ubl, ubiquitin-like ,Research Article ,6-OHDA ,X-IAP, X-linked IAP ,Caspase 8 ,PRR, proline-rich region ,ROS, reactive oxygen species ,FBS, fetal bovine serum ,UPS, ubiquitin-proteasome system ,Animals ,Humans ,FBXO7 ,Molecular Biology ,PD, Parkinson’s disease ,SCF, SKP1-Cullin1-F-box ,NDDs, neurodegenerative diseases ,TRAIL, TNF-related apoptosis-inducing ligand ,neuronal cell death ,F-Box Proteins ,FBXO7, F-box only protein 7 ,Cell Biology ,HEK293 Cells ,Proteasome ,Proteolysis ,Parkinson’s disease ,biology.protein - Abstract
Parkinson's disease (PD) is characterized by the progressive loss of midbrain dopamine neurons in the substantia nigra. Mutations in the F-box only protein 7 gene (Fbxo7) have been reported to cause an autosomal recessive form of early-onset familial PD. FBXO7 is a part of the SKP1-Cullin1-F-box (SCF) E3 ubiquitin ligase complex which mediates ubiquitination of numerous substrates. FBXO7 has also been to regulate mitophagy, cell growth, and proteasome activity. A member of the FOXO family, the transcription factor FOXO4, is also known to modulate several cellular responses, including cell cycle progression and apoptosis; however, the relationship between FBXO7 and FOXO4 has not been investigated. In this study, we determined that FBXO7 binds to FOXO4 and negatively regulates intracellular FOXO4 levels. Interestingly, we also found that FBXO7-mediated degradation of FOXO4 did not occur through either of two major proteolysis systems, the ubiquitin-proteasome system or the lysosome-autophagy pathway, although it was blocked by a caspase 8-specific inhibitor and caspase-8-knockdown. Moreover, intracellular FOXO4 levels were greatly reduced in dopaminergic MN9D cells following treatment with neurotoxic 6-hydroxydopamine (6-OHDA), which was produced upon FBXO7-mediated and caspase 8-mediated proteolysis. Taken together, these results suggest that FOXO4 is negatively regulated in FBXO7-linked PD through caspase-8 activation, suppressing the cytoprotective effect of FOXO4 during 6-OHDA-induced neuronal cell death.
- Published
- 2021
17. Death-Inducing Signaling Complex
- Author
-
Schwab, Manfred, editor
- Published
- 2011
- Full Text
- View/download PDF
18. Structural insight for the roles of fas death domain binding to fadd and oligomerization degree of the fas-fadd complex in the death-inducing signaling complex formation: A computational study.
- Author
-
Yan, Qi, McDonald, Jay M., Zhou, Tong, and Song, Yuhua
- Abstract
Fas binding to Fas-associated death domain (FADD) activates FADD-caspase-8 binding to form death-inducing signaling complex (DISC) that triggers apoptosis. The Fas-Fas association exists primarily as dimer in the Fas-FADD complex, and the Fas-FADD tetramer complexes have the tendency to form higher order oligomer. The importance of the oligomerized Fas-FADD complex in DISC formation has been confirmed. This study sought to provide structural insight for the roles of Fas death domain (Fas DD) binding to FADD and the oligomerization of Fas DD-FADD complex in activating FADD-procaspase-8 binding. Results show Fas DD binding to FADD stabilized the FADD conformation, including the increased stability of the critical residues in FADD death effector domain (FADD DED) for FADD-procaspase-8 binding. Fas DD binding to FADD resulted in the decreased degree of both correlated and anticorrelated motion of the residues in FADD and caused the reversed correlated motion between FADD DED and FADD death domain (FADD DD). The exposure of procaspase-8 binding residues in FADD that allows FADD to interact with procaspase-8 was observed with Fas DD binding to FADD. We also observed different degrees of conformational and motion changes of FADD in the Fas DD-FADD complex with different degrees of oligomerization. The increased conformational stability and the decreased degree of correlated motion of the residues in FADD in Fas DD-FADD tetramer complex were observed compared to those in Fas DD-FADD dimer complex. This study provides structural evidence for the roles of Fas DD binding to FADD and the oligomerization degree of Fas DD-FADD complex in DISC formation to signal apoptosis. Proteins 2013. © 2012 Wiley Periodicals, Inc. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
19. Death-Inducing Signaling Complex
- Author
-
Schwab, Manfred, editor
- Published
- 2009
- Full Text
- View/download PDF
20. ER stress sensitizes cells to TRAIL through down-regulation of FLIP and Mcl-1 and PERK-dependent up-regulation of TRAIL-R2.
- Author
-
Martín-Pérez, Rosa, Niwa, Maho, and López-Rivas, Abelardo
- Abstract
Despite recent evidences suggesting that agents inducing endoplasmic reticulum (ER) stress could be exploited as potential antitumor drugs in combination with tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), the mechanisms of this anticancer action are not fully understood. Moreover, the effects of ER stress and TRAIL in nontransformed cells remain to be investigated. In this study we report that ER stress-inducing agents sensitizes both transformed and nontransformed cells to TRAIL-induced apoptosis. In addition, glucose-regulated protein of 78 kDa (GRP78) knockdown by RNA interference induces ER stress and facilitates apoptosis by TRAIL. We demonstrate that TRAIL death-inducing signaling complex (DISC) formation and early signaling are enhanced in ER stressed cells. ER stress alters the cellular levels of different apoptosis-related proteins including a decline in the levels of FLIP and Mcl-1 and the up-regulation of TRAIL-R2. Up-regulation of TRAIL-R2 following ER stress is dependent on the expression of PKR-like ER kinase (PERK) and independent of CAAT/enhancer binding protein homologous protein (CHOP) and Ire1α. Silencing of TRAIL-R2 expression by siRNA blocks the ER stress-mediated sensitization to TRAIL-induced apoptosis. Furthermore, simultaneous silencing of cFLIP and Mcl-1 expression by RNA interference results in a marked sensitization to TRAIL-induced apoptosis. Finally, in FLIP-overexpressing cells ER stress-induced sensitization to TRAIL-activated apoptosis is markedly reduced. In summary, our data reveal a pleiotropic mechanism involving both apoptotic and anti-apoptotic proteins for the sensitizing effect of ER stress on the regulation of TRAIL receptor-mediated apoptosis in both transformed and nontransformed cells. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
21. Haplophytin-A induces caspase-8-mediated apoptosis via the formation of death-inducing signaling complex in human promyelocytic leukemia HL-60 cells
- Author
-
Won, Kyung-Jae, Chung, Kyung-Sook, Lee, Yong Sup, Alia, Muhammad Shaiq, Pervez, Muhammad Kashif, Fatima, Samreen, Choi, Jung-Hye, and Lee, Kyung-Tae
- Subjects
- *
HAPLOPHYTON , *LEUKEMIA , *APOPTOSIS , *MITOCHONDRIAL membranes , *PHOSPHATIDYLSERINES , *SMALL interfering RNA , *RIBOSE - Abstract
Abstract: Haplophytin-A (10-methoxy-2,2-dimethyl-2,6-dihydro-pyrano[3,2-c]quinolin-5-one), a novel quinoline alkaloid, was isolated from the Haplophyllum acutifolium. In this study, we investigated the effect of haplophytin-A on the apoptotic activity and the molecular mechanism of action in human promyelocytic leukemia HL-60 cells. Treatment with haplophytin-A (50μM) induced classical features of apoptosis, such as, DNA fragmentation, DNA ladder formation, and the externalization of annexin-V-targeted phosphatidylserine residues in HL-60 cells. In addition, haplophytin-A triggered the activations of caspase-8, -9, and -3, and the cleavage of poly (ADP-ribose) polymerase (PARP) in HL-60 cells. In addition, haplophytin-A caused the loss of mitochondrial membrane potential (ΔΨ m ) and the release of cytochrome c and Smac/DIABLO to the cytosol, and modulated the expression levels of Bcl-2 family proteins. We further demonstrated that knockdown of caspase-8 using its siRNA inhibited the mitochondrial translocation of tBid, the activations of caspase-9 and caspase-3, and subsequent DNA fragmentation by haplophytin-A. Furthermore, haplophytin-A-induced the formation of death-inducing signaling complex (DISC) and then activated caspase-8 in HL-60 cells. During haplophytin-A-induced apoptosis, caspase-8-stimulated tBid provide a link between the death receptor-mediated extrinsic pathway and the mitochondria- mediated intrinsic pathway. Taken together, these results suggest that the novel compound haplophytin-A play therapeutical role for leukemia via the potent apoptotic activity through the extrinsic pathway, involving the intrinsic pathway. [Copyright &y& Elsevier]
- Published
- 2010
- Full Text
- View/download PDF
22. Lipid rafts and clusters of apoptotic signaling molecule-enriched rafts in cancer therapy.
- Published
- 2010
- Full Text
- View/download PDF
23. Abstract 1932: Talazoparib interacts with oncolytic reovirus to enhance death-inducing signaling complex (DISC)-mediated apoptosis and immune response
- Author
-
Jyoti S. Choudhary, Dragomir B. Krastev, Harriet Whittock, Victoria Roulstone, Lu Yu, Alan Melcher, Richard Elliott, Malin Pedersen, Kevin J. Harrington, Tencho Tenev, James C. Wright, Grey A. Wilkinson, Stephen J. Pettitt, Christopher J. Lord, Martin McLaughlin, Pascal Meier, M. Coffey, Galabina Bozhanova, Joan N. Kyula, and Arnaud J. Legrand
- Subjects
Cancer Research ,Immune system ,Oncology ,Apoptosis ,Chemistry ,Death-inducing signaling complex ,Cancer research ,Oncolytic virus - Abstract
Reovirus (RT3D) is a naturally occurring double-stranded RNA oncolytic virus that has shown preclinical efficacy in a wide range of tumor types. Early phase clinical studies have shown that this agent has modest monotherapy efficacy and can safely be combined with cytotoxic chemotherapy regimens. In the current studies, we used a high-throughput drug screen approach of different targeted therapeutic agents with the aim of looking for potential viral sensitizers that could enhance RT3D tumor killing. BMN-673 (talazoparib), a clinically approved poly(ADP)-ribose polymerase 1 (PARP-1) inhibitor was identified as a top hit and found to sensitize profoundly to RT3D both in vitro and in vivo in human xenograft tumors in a nude mouse model. We found that RT3D activated cellular PARP1 and was associated with PARylation of cellular proteins, including components of the DISC-associated cell death machinery. Combined treatment with RT3D and talazoparib enhanced extrinsic apoptosis (amplified by autocrine/paracrine TNF-α and TRAIL signaling), NF-κB pathway activity and pro-inflammatory cytokine production (CCL5/RANTES, CXCL8/IL8, CXCL1/GRO and CXCL10/IP10). Signaling was shown to be dependent on nucleic acid sensing mechanisms mediated by RIG-I and TLR3. We also found anti-tumour efficacy in an immunocompetent mouse model and this correlated with an increase in an immune response following combination treatment of RT3D and talazoparib. Our data provide a strong rationale for the combination of oncolytic RT3D with PARP1 inhibitors to exploit immunogenic response in cancer treatment. Citation Format: Joan N. Kyula, Victoria Roulstone, Richard Elliott, Harriet Whittock, Galabina Bozhanova, Martin McLaughlin, Malin Pedersen, Dragomir Krastev, Stephen Pettitt, Arnaud Legrand, Tencho Tenev, James Wright, Lu Yu, Jyoti Choudhary, Pascal Meier, Christopher J. Lord, Alan Melcher, Grey Wilkinson, Matt Coffey, Kevin J. Harrington. Talazoparib interacts with oncolytic reovirus to enhance death-inducing signaling complex (DISC)-mediated apoptosis and immune response [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr 1932.
- Published
- 2021
24. Lipid raft connection between extrinsic and intrinsic apoptotic pathways
- Author
-
Gajate, Consuelo, Gonzalez-Camacho, Fernando, and Mollinedo, Faustino
- Subjects
- *
APOPTOSIS , *CELLULAR signal transduction , *CELL receptors , *ANTINEOPLASTIC agents , *PHARMACODYNAMICS , *CANCER cells , *ENZYME activation , *LEUKEMIA - Abstract
Abstract: Apoptosis in mammalian cells is modulated by extrinsic and intrinsic signaling pathways through the formation of death receptor-mediated death-inducing signaling complex (DISC) and mitochondrial-derived apoptosome, respectively. We found by ultrastructural approaches that the antitumor drug edelfosine induced aggregates of lipid rafts containing Fas/CD95 receptor and Fas-associated death domain-containing protein in leukemic cells. Death receptors together with DISC and apoptosome constituents were recruited in rafts during edelfosine treatment in multiple myeloma cells. This apoptotic response involved caspases-8/-9/-10 that were translocated to rafts. Lipid raft disruption by cholesterol depletion inhibited loss of mitochondrial transmembrane potential, caspase activation and apoptosis, whereas cholesterol replenishment restored these responses. Our data indicate that rafts act as scaffolds where extrinsic and intrinsic apoptotic signaling pathways concentrate, forming clusters of apoptotic signaling molecule-enriched rafts (CASMER), which function as novel supramolecular entities in the triggering of apoptosis, and play an important role in edelfosine-induced apoptosis in blood cancer cells. [Copyright &y& Elsevier]
- Published
- 2009
- Full Text
- View/download PDF
25. An engineered construct of cFLIP provides insight into DED1 structure and interactions.
- Author
-
Panaitiu, Alexandra E., Basiashvili, Tamar, Mierke, Dale F., and Pellegrini, Maria
- Subjects
- *
NUCLEAR magnetic resonance , *CALMODULIN , *PROTEIN folding , *PROTEIN-protein interactions , *AUTOIMMUNE diseases - Abstract
Cellular FLICE-like inhibitory protein (cFLIP) is a member of the Death Domain superfamily with pivotal roles in many cellular processes and disease states, including cancer and autoimmune disorders. In the context of the death-inducing signaling complex (DISC), cFLIP isoforms regulate extrinsic apoptosis by controlling procaspase-8 activation. The function of cFLIP is mediated through a series of protein-protein interactions, engaging the two N-terminal death effector domains (DEDs). Here, we solve the structure of an engineered DED1 domain of cFLIP using solution nuclear magnetic resonance (NMR) and we define the interaction with FADD and calmodulin, protein-protein interactions that regulate the function of cFLIP in the DISC. cFLIP DED1 assumes a canonical DED fold characterized by six α helices and is able to bind calmodulin and FADD through two separate interfaces. Our results clearly demonstrate the role of DED1 in the cFLIP/FADD association and contribute to the understanding of the assembly of DISC filaments. [Display omitted] • A soluble, well-folded DED1 of cFLIP is generated with a graft from FADD DED • DED1ch is a classical 7-helix bundle amenable to NMR studies • DED1ch binds FADD DED utilizing a typical hydrophilic interface of type IIb • DED1ch retains binding to calmodulin involving its sixth helix Panaitiu et al. report the solution structure of an engineered DED1 of cFLIP, obtained by borrowing a small stretch of residues from the single DED of FADD. The resulting protein produces NMR spectra that allow for structure determination and investigation of protein-protein interactions, including with DED of FADD and calmodulin. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
26. Regulation of hypoxic neuronal death signaling by neuroglobin.
- Author
-
Khan, Adil A., Xiao Ou Mao, Banwait, Surita, Dermardirossian, Celine M., Bokoch, Gary M., Kunlin Jin, and Grenberg, David A.
- Subjects
- *
CELLULAR signal transduction , *NEURONS , *CELL death , *GLOBIN , *HYPOXEMIA , *PROTEIN kinases - Abstract
The signal transduction pathways involved in neuronal death are not well understood. Neuroglobin (Ngb), a recently discovered vertebrate globin expressed predominantly in the brain, shows increased expression in neurons in response to oxygen deprivation and protects neurons from ischemic and hypoxic death. The mechanism of this neuroprotection is unclear. We examined the surface distribution of raft membrane microdomains in cortical neuron cultures during hypoxia using the raft marker cholera toxin B (CTx-B) subunit. Mechanistically, we demonstrate that hypoxia induces rapid polarization of somal membranes and aggregation of microdomains with the subjacent mitochondrial network. This signaling complex is formed well before neurons commit to die, consistent with an early role in death signal transduction Neurons from Ngb-overexpressing transgenic (Ngb-Tg) mice do not undergo microdomain polarization or mitochondrial aggregation in response to, and are resistant to death from hypoxia. We link the protective actions of Ngb to inhibition of Pak1 kinase activity and Rac1-GDP-dissociation inhibitor disassociation, and inhibition of actin assembly and death-signaling module polarization. [ABSTRACT FROM AUTHOR]
- Published
- 2008
- Full Text
- View/download PDF
27. Dissecting lipid raft facilitated cell signaling pathways in cancer
- Author
-
Patra, Samir Kumar
- Subjects
- *
CANCER , *CELL membranes , *CELL death , *ENDOCYTOSIS - Abstract
Abstract: Cancer is one of the most devastating disorders in our lives. Higher rate of proliferation than death of cells is one of the essential factors for development of cancer. The dynamicity of cell membrane plays some vital roles in cell survival and cell death, including protection, endocytosis, signaling, and increases in mechanical stability during cell division, as well as decrease of shear forces during separation of two cells after division, and cell separation from tissues for cancer metastasis. Within the membrane, there are specialized domains, known as lipid rafts. A raft can coordinate various signaling pathways. Recent data on the proteomics of lipid rafts/caveolae have highlighted the enigmatic role of various signaling proteins in cancer development. Analysis of these data of raft proteome from various tumors, cancer tissues, and cell lines cultured without and with therapeutic agents, as well as from model rafts revealed that there may be two subsets of raft assemblage in cell membrane. One subset of raft is enriched with cholesterol–sphingomyeline–ganglioside–cav-1/Src/EGFR (hereafter, “chol-raft”) that is involved in normal cell signaling, and when dysregulated promotes cell transformation and tumor progression; another subset of raft is enriched with ceramide–sphingomyeline–ganglioside–FAS/Ezrin (hereafter, “cer-raft”) that generally promotes apoptosis. In view of this, and to focus insight into the cancer cell physiology caused by the lipid rafts mediated signals and their receptors, and the downstream transmitters, either proliferative (for example, EGF and EGFR) or death-inducing (for example, FASL and FAS), and the precise roles of some therapeutic drugs and endogenous acid sphingomylenase in this scenario in in situ transformation of “chol-raft” into “cer-raft” are summarized and discussed in this contribution. [Copyright &y& Elsevier]
- Published
- 2008
- Full Text
- View/download PDF
28. FLIP as an Anti-Cancer Therapeutic Target.
- Author
-
Jin Kuk Yang
- Abstract
Suppression of apoptosis is one of the hallmarks of carcinogenesis. Tumor cells endure apoptotic pressure by overexpressing several antiapoptotic proteins, and FLICE inhibitory protein (FLIP) is one of the important antiapoptotic proteins that have been shown to be overexpressed in various primary tumor cells. FLIP has two death-effector domains in tandem, mimicking the prodomain of procaspase-8. It is recruited to Fadd in death-inducing signaling complex, thereby preventing the activation of procaspase-8. To date, three isoforms of human cytosolic FLIP (c-FLIP) and six viral homologs (v-FLIP) have been identified. Recently, the crystal structure of v-FLIP MC159 was determined for the first time as an atomic-detail FLIP structure, which revealed that two death effector domains are packed tightly against each other mainly through conserved hydrophobic interactions. The overexpression of c-FLIP in tumor cells has been shown to be the determinant of the tumor's resistance to death ligands such as FasL and TRAIL. It has also been shown that the downregulation of c-FLIP results in sensitizing resistant tumor cells. Therefore, the agents directly targeting c-FLIP at mRNA and protein levels are expected to be developed in near future and tested for the potential as a new class of anti-cancer drugs. [ABSTRACT FROM AUTHOR]
- Published
- 2008
- Full Text
- View/download PDF
29. Apoptosis signaling pathways and lymphocyte homeostasis.
- Author
-
Xu, Guangwu and Shi, Yufang
- Subjects
LYMPHOCYTES ,LEUCOCYTES ,APOPTOSIS ,HOMEOSTASIS ,PHYSIOLOGICAL control systems - Abstract
It has been almost three decades since the term “apoptosis” was first coined to describe a unique form of cell death that involves orderly, gene-dependent cell disintegration. It is now well accepted that apoptosis is an essential life process for metazoan animals and is critical for the formation and function of tissues and organs. In the adult mammalian body, apoptosis is especially important for proper functioning of the immune system. In recent years, along with the rapid advancement of molecular and cellular biology, great progress has been made in understanding the mechanisms leading to apoptosis. It is generally accepted that there are two major pathways of apoptotic cell death induction: extrinsic signaling through death receptors that leads to the formation of the death-inducing signaling complex (DISC), and intrinsic signaling mainly through mitochondria which leads to the formation of the apoptosome. Formation of the DISC or apoptosome, respectively, activates initiator and common effector caspases that execute the apoptosis process. In the immune system, both pathways operate; however, it is not known whether they are sufficient to maintain lymphocyte homeostasis. Recently, new apoptotic mechanisms including caspase-independent pathways and granzyme-initiated pathways have been shown to exist in lymphocytes. This review will summarize our understanding of the mechanisms that control the homeostasis of various lymphocyte populations.Cell Research (2007) 17:759–771. doi: 10.1038/cr.2007.52; published online 19 June 2007 [ABSTRACT FROM AUTHOR]
- Published
- 2007
- Full Text
- View/download PDF
30. Calmodulin Binding to Death Receptor 5-mediated Death-Inducing Signaling Complex in Breast Cancer Cells
- Author
-
Donald J. Buchsbaum, Kurt R. Zinn, Harrison Kim, Yuhua Song, Tong Zhou, and Romone M. Fancy
- Subjects
0301 basic medicine ,Calmodulin ,biology ,Cell ,Cell Biology ,Biochemistry ,Cell biology ,03 medical and health sciences ,EGTA ,chemistry.chemical_compound ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,chemistry ,Apoptosis ,030220 oncology & carcinogenesis ,Death-inducing signaling complex ,medicine ,biology.protein ,Viability assay ,Signal transduction ,Molecular Biology ,Caspase - Abstract
Activation of death receptor-5 (DR5) leads to the formation of death-inducing signaling complex (DISC) for apoptotic signaling. TRA-8, a DR5 specific agonistic antibody, has demonstrated significant cytotoxic activity in vitro and in vivo without inducing hepatotoxicity. Calmodulin (CaM) that is overexpressed in breast cancer plays a critical role in regulating DR5-mediated apoptosis. However, the mechanism of CaM in regulating DR5-mediated apoptotic signaling remains unknown. In this study, we characterized CaM binding to DR5-mediated DISC for apoptosis in TRA-8 sensitive breast cancer cell lines using co-immunoprecipitation, fluorescence microscopic imaging, caspase signaling analysis, and cell viability assay. Results show that upon DR5 activation, CaM was recruited into DR5-mediated DISC in a calcium dependent manner. CaM antagonist, trifluoperazine (TFP), inhibited CaM recruitment into the DISC and attenuated DISC formation. DR5 oligomerization is critical for DISC formation for apoptosis. TFP decreased TRA-8 activated DR5 oligomerization, which was consistent with TFP's effect on DR5-mediated DISC formation. TFP and Ca2+ chelator, EGTA, impeded TRA-8-activated caspase-dependent apoptotic signaling, and TFP decreased TRA-8-induced cell cytotoxicity. These results demonstrated CaM binding to DR5-mediated DISC in a calcium dependent manner and may identify CaM as a key regulator of DR5-mediated DISC formation for apoptosis in breast cancer. J. Cell. Biochem. 118: 2285-2294, 2017. © 2017 Wiley Periodicals, Inc.
- Published
- 2017
31. Formation of neurodegenerative aggresome and death-inducing signaling complex in maternal diabetes-induced neural tube defects
- Author
-
Lixue Cao, Zhiyong Zhao, and E. Albert Reece
- Subjects
0301 basic medicine ,Protein Folding ,congenital, hereditary, and neonatal diseases and abnormalities ,Programmed cell death ,Huntingtin ,Cell Survival ,Apoptosis ,Biology ,Protein aggregation ,Parkin ,Diabetes Mellitus, Experimental ,Mice ,03 medical and health sciences ,Pregnancy ,mental disorders ,medicine ,Animals ,Neural Tube Defects ,Caspase 8 ,Multidisciplinary ,Neural tube ,Gene Expression Regulation, Developmental ,Biological Sciences ,nervous system diseases ,Cell biology ,Enzyme Activation ,Mice, Inbred C57BL ,Diabetes, Gestational ,030104 developmental biology ,medicine.anatomical_structure ,Aggresome ,Biochemistry ,Death-inducing signaling complex ,Female ,Chemical chaperone - Abstract
Diabetes mellitus in early pregnancy increases the risk in infants of birth defects, such as neural tube defects (NTDs), known as diabetic embryopathy. NTDs are associated with hyperglycemia-induced protein misfolding and Caspase-8-induced programmed cell death. The present study shows that misfolded proteins are ubiquitinylated, suggesting that ubiquitin-proteasomal degradation is impaired. Misfolded proteins form aggregates containing ubiquitin-binding protein p62, suggesting that autophagic-lysosomal clearance is insufficient. Additionally, these aggregates contain the neurodegenerative disease-associated proteins α-Synuclein, Parkin, and Huntingtin (Htt). Aggregation of Htt may lead to formation of a death-inducing signaling complex of Hip1, Hippi, and Caspase-8. Treatment with chemical chaperones, such as sodium 4-phenylbutyrate (PBA), reduces protein aggregation in neural stem cells in vitro and in embryos in vivo. Furthermore, treatment with PBA in vivo decreases NTD rate in the embryos of diabetic mice, as well as Caspase-8 activation and cell death. Enhancing protein folding could be a potential interventional approach to preventing embryonic malformations in diabetic pregnancies.
- Published
- 2017
32. Caspase-10 Negatively Regulates Caspase-8-Mediated Cell Death, Switching the Response to CD95L in Favor of NF-κB Activation and Cell Survival
- Author
-
Ramon Schilling, Michelle A. Hughes, Pascal Meier, Sebastian Horn, Martin Leverkus, Michaela Ploesser, Marion MacFarlane, Carsten Sticht, Tencho Tenev, and Martin R. Sprick
- Subjects
0301 basic medicine ,Indoles ,Fas-Associated Death Domain Protein ,CASP8 and FADD-Like Apoptosis Regulating Protein ,caspase-10 ,Apoptosis ,caspase-8 ,NF-κB ,chemistry.chemical_compound ,NF-KappaB Inhibitor alpha ,Clustered Regularly Interspaced Short Palindromic Repeats ,FADD ,RNA, Small Interfering ,Caspase 10 ,lcsh:QH301-705.5 ,Caspase 8 ,biology ,Imidazoles ,NF-kappa B ,Fas receptor ,Cell biology ,cell death ,Death-inducing signaling complex ,CD95 ,RNA Interference ,Signal transduction ,Oligopeptides ,Signal Transduction ,Programmed cell death ,Fas Ligand Protein ,Cell Survival ,Article ,General Biochemistry, Genetics and Molecular Biology ,Cell Line ,03 medical and health sciences ,Humans ,RNA, Messenger ,fas Receptor ,030102 biochemistry & molecular biology ,cFLIP ,Interleukin-8 ,DISC ,030104 developmental biology ,lcsh:Biology (General) ,chemistry ,biology.protein ,HeLa Cells - Abstract
Summary Formation of the death-inducing signaling complex (DISC) initiates extrinsic apoptosis. Caspase-8 and its regulator cFLIP control death signaling by binding to death-receptor-bound FADD. By elucidating the function of the caspase-8 homolog, caspase-10, we discover that caspase-10 negatively regulates caspase-8-mediated cell death. Significantly, we reveal that caspase-10 reduces DISC association and activation of caspase-8. Furthermore, we extend our co-operative/hierarchical binding model of caspase-8/cFLIP and show that caspase-10 does not compete with caspase-8 for binding to FADD. Utilizing caspase-8-knockout cells, we demonstrate that caspase-8 is required upstream of both cFLIP and caspase-10 and that DISC formation critically depends on the scaffold function of caspase-8. We establish that caspase-10 rewires DISC signaling to NF-κB activation/cell survival and demonstrate that the catalytic activity of caspase-10, and caspase-8, is redundant in gene induction. Thus, our data are consistent with a model in which both caspase-10 and cFLIP coordinately regulate CD95L-mediated signaling for death or survival., Graphical Abstract, Highlights • Caspase-10 negatively regulates DISC-mediated caspase-8 activation and cell death • DISC formation and caspase-10 recruitment depend on caspase-8 scaffold function • Caspase-10 rewires DISC signaling to NF-κB-induced gene induction/cell survival • The catalytic activity of caspase-10 and caspase-8 are redundant in gene induction, It has been assumed that caspase-10, and its homolog caspase-8, have redundant functions in cell death signaling. Horn et al. now reveal a role for caspase-10 in switching CD95 signaling from caspase-8-induced cell death to NF-κB activation/cell survival. DISC recruitment of caspase-10 and NF-κB activation critically depend upon caspase-8 scaffold function.
- Published
- 2017
33. Death-Inducing Signaling Complex
- Author
-
Assenmacher, Mario, Avraham, Hava Karsenty, Avraham, Shalom, Bala, Shukal, Barnett, John, Basketter, David, Ben-David, Yaacov, Berek, Claudia, Blümel, Jörg, Bolliger, Anne Provencher, Bolon, Brad, Bradley, S Gaylen, Brundage, Kathleen M, Brunner, Georg, Bugelski, Peter J, Burchiel, Scott W, Burns-Naas, Leigh Ann, Bussiere, Jeanine L., Cameron, Scott B, Carey, Michelle, Cederbrant, Karin, Chow, Anthony W, Cohen, Mitchell D., Colagiovanni, Dorothy, Contreras, Marcela, Cornacoff, Joel B, Corsini, Emanuela, Crevel, René, Cuff, Christopher, Czuprynski, Charles J, Damoiseaux, Jan G M C, Daniels, Geoff, Dayan, Anthony D, Dearman, Rebecca J, Dodson, Sarah V. M., Ebringer, Alan, Engel, Andrea, Esser, Charlotte, Fairley, Kimberly J, Fernandez-Botran, Rafael, Flaherty, Dennis K, Frings, Werner, Gad, Shayne Cox, Gardner, Donald E, Gardner, Susan C, Garssen, Johan, Gashev, Anatoliy A, Geffner, Jorge, Geginat, Gernot, Gemsa, Diethard, Gerberick, Frank, Germolec, Dori, Gilbert, Kathleen M., Giles-Komar, Jill, Gore, Elizabeth R, Griem, Peter, Hagelschuer, Ina, Haggerty, Helen G., Hall, Andrew, Hanneken, S., Hastings, Kenneth L, Havelaar, Arie H, Heisler, Eckhart, Helm, Ricki M, Henschler, Reinhard, Herrmann, Thomas, Herzyk, Danuta J, Higgins, Rachel R., Hitzfeld, Bettina, Holladay, Steven, Holsapple, Michael, House, Robert V, Hughes, Lucy, Jeong, Tae Cheon, Johnson, Victor J, de Jong, Wim H, de Jonge, Rob, Kamath, Arati, Kaminski, Norbert E, Kaminsky, Ronald, Karol, Meryl, Kashon, Michael L, Kerkvliet, Nancy I, Kimber, Ian, Knight, David M, Knulst, A C, Koren, Eugen, Kraal, Georg, Kretz-Rommel, Anke, Kuper, C Frieke, Ladics, Gregory, Laiosa, Michael, Landreth, Kenneth S., Lawrence, B Paige, Lawrence, David A, Lee, Byeong-Chel, Lee, William, Leino, Lasse, Lemke, Hilmar, Lewis, J G, Liebau, Jutta, Lollini, Pier-Luigi, van Loveren, Henk, Luebke, Bob, Luster, Michael I, Mage, Rose G, Maier, Curtis C., Martin, Michael U., Maurer, Thomas, McKarns, Susan C, Meade, B Jean, Moser, Bernhard, Nagata, Shigekazu, Nain, Marianne, Neumann, Norbert J., Novicki, Deborah L, Olsen, John L, Pauluhn, Jürgen, Pichler, Werner, Pieters, Raymond, Pollard, K Michael, Preissner, Klaus T, Pruett, Stephen B, Pumford, Neil R., Rashid, Taha, Ratajczak, Helen V, Redegeld, Frank A M, Regal, Jean F, Resch, Klaus, Rodgers, Kathleen, Roman, Danielle, Rose, Noel R, Rosenthal, Gary J., Sali, Tina, Samsom, Janneke N, Savelkoul, Huub F J, Schafer, Rosana, Schatz, Mark, Schild, Hansjoerg, Shepherd, David, Shiohara, Tetsuo, Silverstone, Allen, Simeonova, Petia P, Smialowicz, Ralph J, Smith, K G C, Soos, Jeanne M, Stittelaar, Koert J, Straube, Frank, Sulentic, Courtney E W, Swart, B, Takumi, Katsuhisa, Tarkowski, Maciej, Tervaert, Jan Willem Cohen, Thomas, Peter T, Tinkle, Sally S, Treacy, George, Trouba, Kevin, Tryphonas, Helen, Uguccioni, Mariagrazia, Ulrich, Peter, van der Heijden, Maurice W, Van Loveren, H, Vandebriel, Rob J, Vleminckx, Kris, Vohr, Hans-Werner, Weinbauer, Gerhard F, Weinstein, I Bernard, Weltzien, Hans Ulrich, Weston, Ainsley, White, Kimber L., Wilson, Clyde, Wing, Mark, Wolf, Anna Maria, Yaqoob, Parveenn, Yucesoy, Berran, Zawieja, David C, Zelikoff, Judith T, Zola, H, and van Zwieten, P A
- Published
- 2005
- Full Text
- View/download PDF
34. 23-Hydroxyursolic Acid Isolated from the Stem Bark of Cussonia bancoensis Induces Apoptosis through Fas/Caspase-8-Dependent Pathway in HL-60 Human Promyelocytic Leukemia Cells
- Author
-
Léon Azefack Tapondjou, Kyung-Tae Lee, Hee-Juhn Park, Jong-Heon Won, Kyung-Sook Chung, Masaaki Nomura, Ahmed H.E. Hassan, Eun-Young Park, Jung-Hye Choi, and Jeong-Hun Lee
- Subjects
0301 basic medicine ,caspase ,Pharmaceutical Science ,Caspase 8 ,Fas ligand ,Analytical Chemistry ,lcsh:QD241-441 ,03 medical and health sciences ,0302 clinical medicine ,lcsh:Organic chemistry ,Drug Discovery ,Bcl-2 ,FADD ,Physical and Theoretical Chemistry ,Fragmentation (cell biology) ,23-hydroxyursolic acid ,biology ,Chemistry ,Cytochrome c ,Organic Chemistry ,apoptosis ,death-inducing signaling complex ,Cell biology ,mitochondria ,030104 developmental biology ,Chemistry (miscellaneous) ,Apoptosis ,030220 oncology & carcinogenesis ,Death-inducing signaling complex ,biology.protein ,Molecular Medicine ,DNA fragmentation - Abstract
The natural product 23-hydroxyursolic acid (23-HUA) is a derivative of ursolic acid, which is known to induce cancer cell apoptosis. However, apoptotic effects and mechanisms of 23-HUA have not been well characterized yet. Herein, we investigated the molecular mechanisms of 23-HUA-induced apoptosis in HL-60 human promyelocytic leukemia cells. 23-HUA-treated HL-60 cells showed apoptotic features including internucleosomal DNA condensation and fragmentation as well as externalization of phosphatidylserine residues. 23-HUA induced a series of mitochondrial events including disruption of mitochondrial membrane potential (&Delta, &Psi, m), cytochrome c and Smac/DIABLO release and loss of balance between pro-apoptotic and anti-apoptotic Bcl-2 proteins in HL-60 cells. In addition, 23-HUA activated caspase-8, caspase-9 and caspase-3. Pretreatment with a broad caspase inhibitor (z-VAD-fmk), a caspase-3 inhibitor (z-DEVD-fmk), and a caspase-8 inhibitor (z-IETD-fmk) significantly attenuated 23-HUA-induced DNA fragmentation. After 23-HUA-induced apoptosis, proteins expression levels of FasL, Fas and FADD constituting the death-inducing signaling complex (DISC) were upregulated in HL-60 cells. Moreover, transfection with Fas or FADD siRNA significantly blocked 23-HUA-induced DNA fragmentation and caspases activation. Taken together, these findings indicate that 23-HUA induces apoptosis in HL-60 human promyelocytic leukemia cells through formation of DISC and caspase-8 activation leading to loss of &Delta, m and caspase-3 activation.
- Published
- 2018
- Full Text
- View/download PDF
35. Computer simulation of the spatial structures of MUC1 peptides capable of inhibiting apoptosis
- Author
-
Vladimir A. Ivanisenko, Inna N. Lavrik, and Nikita V. Ivanisenko
- Subjects
0301 basic medicine ,chemistry.chemical_classification ,Programmed cell death ,biology ,Signal transducing adaptor protein ,Peptide ,Caspase 8 ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,chemistry ,Biochemistry ,Death-inducing signaling complex ,Genetics ,biology.protein ,Animal Science and Zoology ,FADD ,Signal transduction ,Agronomy and Crop Science ,Caspase - Abstract
The identification of new effective apoptosis inhibitors plays an important role in the development of drugs for the treatment of various disorders, including neurogenerative diseases. Apoptosis is initiated via the formation of macromolecular protein complexes. These complexes exert the activation of caspases, which are key regulators and executors of apoptosis. The death inducing signaling complex, (DISC) plays a central role in the induction of the extrinsic apoptosis pathway. The adaptor protein FADD is the core component of the DISC that is essential for caspase activation at the DISC and subsequent apoptosis initiation. Therefore, inhibitors of FADD may serve as candidate drugs inhibiting apoptosis. Furthermore, the study of the mechanisms of action of these inhibitors is of great interest for understanding the signal transduction pathways of apoptosis. It has been reported that the mucin type 1 glycoprotein (MUC1) is a natural protein inhibitor of FADD. In particular, two fragments of the primary structure of the cytoplasmic domain of MUC1 (MUC1-CD) are capable of inhibiting the binding of procaspase 8 to FADD. However, the 3D structure of MUC1 has not been obtained yet. This significantly complicates the rational design of potential drugs based on the peptides derived from the MUC1 structure. The aim of the present study was in silico prediction of the 3D structures of MUC1-CD peptides corresponding to protein fragments 120 and 4672, as well as the analysis of their conformational properties. The special attention was placed on the MUC1-CD (46-72) peptide, which is able to bind to FADD. By using the method of molecular dynamics in implicit water it was shown that the structure of the peptide MUC1-CD (46-72) is similar to the three-dimensional structures of at least four fragments of caspase 8. These results indicate that the molecular mechanism of the inhibitory action of the peptide can be explained by the competitive binding of MUC1 to FADD due to the structural and conformational similarity to fragments of the caspase 8 DEDs.
- Published
- 2016
36. Expression of FADD and cFLIPL balances mitochondrial integrity and redox signaling to substantiate apoptotic cell death
- Author
-
Chandramani Pathak and Kishu Ranjan
- Subjects
0301 basic medicine ,Programmed cell death ,Cell signaling ,biology ,Clinical Biochemistry ,Autophagy ,Cell Biology ,General Medicine ,Mitochondrion ,urologic and male genital diseases ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,Apoptosis ,Death-inducing signaling complex ,biology.protein ,Cancer research ,Ectopic expression ,FADD ,biological phenomena, cell phenomena, and immunity ,Molecular Biology - Abstract
FADD and cFLIP both are pivotal components of death receptor signaling. The cellular signaling of apoptosis accomplished with death receptors and mitochondria follows independent pathways for cell death. FADD and cFLIP both have an important role in the regulation of apoptotic and non-apoptotic functions. Dysregulated expression of FADD and cFLIP is associated with resistance to apoptosis in cancer cells. Mitochondria are known to play critical role in maintaining cellular respiration and homeostasis in the cells as well as transduces various signals to determine the fate of cell death. However, involvement of FADD and cFLIP in regulation of mitochondrial integrity and programmed cell death signaling to define the fate of cells remains elusive. In the present study, we explored that, induced expression of FADD challenges the mitochondrial integrity and pulverizes the membrane potential by altering the expression of Bcl-2 and cytochrome c. In contrast, mutant of FADD was unable to affect the mitochondrial integrity. Interestingly, expression of FADD and cFLIP helps to balance redox potential by regulating the anti-oxidant levels. Further, we noticed that, knockdown of cFLIPL and induced expression of FADD rapidly accumulate intracellular ROS accompanied by JNK1 activation to substantiate apoptosis. Notably, the ectopic expression of cFLIPL resists the sensitivity of cancer cells against apoptosis inducers Etoposide and HA14-1. Altogether, our findings suggest that FADD and cFLIPL are important modulators of mitochondrial-associated apoptosis apart from the death receptor signaling.
- Published
- 2016
37. Characterization of the Interactions between Calmodulin and Death Receptor 5 in Triple-negative and Estrogen Receptor-positive Breast Cancer Cells
- Author
-
Romone M. Fancy, Hong Wang, Qinghua Zeng, Tong Zhou, Yuhua Song, Donald J. Buchsbaum, and Lingyun Wang
- Subjects
0301 basic medicine ,Calmodulin ,Estrogen receptor ,Cell Biology ,Triple Negative Breast Neoplasms ,Biology ,medicine.disease ,Biochemistry ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Breast cancer ,030220 oncology & carcinogenesis ,Death-inducing signaling complex ,biology.protein ,medicine ,FADD ,Signal transduction ,Molecular Biology ,Death domain - Abstract
Activation of death receptor-5 (DR5) leads to the formation of death inducing signaling complex (DISC) for apoptotic signaling. Targeting DR5 to induce breast cancer apoptosis is a promising strategy to circumvent drug resistance and present a target for breast cancer treatment. Calmodulin (CaM) has been shown to regulate DR5-mediated apoptotic signaling, however, its mechanism remains unknown. In this study, we characterized CaM and DR5 interactions in breast cancer cells with integrated experimental and computational approaches. Results show that CaM directly binds to DR5 in a calcium dependent manner in breast cancer cells. The direct interaction of CaM with DR5 is localized at DR5 death domain. We have predicted and verified the CaM-binding site in DR5 being (354)WEPLMRKLGL(363) that is located at the α2 helix and the loop between α2 helix and α3 helix of DR5 DD. The residues of Trp-354, Arg-359, Glu-355, Leu-363, and Glu-367 in DR5 death domain that are important for DR5 recruitment of FADD and caspase-8 for DISC formation to signal apoptosis also play an important role for CaM-DR5 binding. The changed electrostatic potential distribution in the CaM-binding site in DR5 DD by the point mutations of W354A, E355K, R359A, L363N, or E367K in DR5 DD could directly contribute to the experimentally observed decreased CaM-DR5 binding by the point mutations of the key residues in DR5 DD. Results from this study provide a key step for the further investigation of the role of CaM-DR5 binding in DR5-mediated DISC formation for apoptosis in breast cancer cells.
- Published
- 2016
38. Co-operative and Hierarchical Binding of c-FLIP and Caspase-8: A Unified Model Defines How c-FLIP Isoforms Differentially Control Cell Fate
- Author
-
Martin Leverkus, Marion MacFarlane, Michelle A. Hughes, Rebekah Jukes-Jones, Sebastian Horn, Maria Feoktistova, Kelvin Cain, John W.R. Schwabe, Ian R. Powley, and Louise Fairall
- Subjects
0301 basic medicine ,Gene isoform ,Fas-Associated Death Domain Protein ,CASP8 and FADD-Like Apoptosis Regulating Protein ,Apoptosis ,Plasma protein binding ,Caspase 8 ,Bioinformatics ,Article ,03 medical and health sciences ,0302 clinical medicine ,Tandem Mass Spectrometry ,Humans ,Protein Isoforms ,Cell Lineage ,FADD ,Molecular Biology ,biology ,Activator (genetics) ,Cell Biology ,Cell biology ,030104 developmental biology ,Mutagenesis ,Flip ,030220 oncology & carcinogenesis ,Death-inducing signaling complex ,biology.protein ,Protein Binding - Abstract
Summary The death-inducing signaling complex (DISC) initiates death receptor-induced apoptosis. DISC assembly and activation are controlled by c-FLIP isoforms, which function as pro-apoptotic (c-FLIPL only) or anti-apoptotic (c-FLIPL/c-FLIPS) regulators of procaspase-8 activation. Current models assume that c-FLIP directly competes with procaspase-8 for recruitment to FADD. Using a functional reconstituted DISC, structure-guided mutagenesis, and quantitative LC-MS/MS, we show that c-FLIPL/S binding to the DISC is instead a co-operative procaspase-8-dependent process. FADD initially recruits procaspase-8, which in turn recruits and heterodimerizes with c-FLIPL/S via a hierarchical binding mechanism. Procaspase-8 activation is regulated by the ratio of unbound c-FLIPL/S to procaspase-8, which determines composition of the procaspase-8:c-FLIPL/S heterodimer. Thus, procaspase-8:c-FLIPL exhibits localized enzymatic activity and is preferentially an activator, promoting DED-mediated procaspase-8 oligomer assembly, whereas procaspase-8:c-FLIPS lacks activity and potently blocks procaspase-8 activation. This co-operative hierarchical binding model explains the dual role of c-FLIPL and crucially defines how c-FLIP isoforms differentially control cell fate., Graphical Abstract, Highlights • c-FLIP isoforms (L/S) do not directly compete with caspase-8 for binding to FADD • c-FLIP binds to the DISC via a co-operative hierarchical caspase-8-dependent process • Co-operative and hierarchical binding crucially explains the dual function of c-FLIPL • Our unified model defines how c-FLIP isoforms differentially direct cell fate, It is unclear how c-FLIP isoforms can differentially regulate caspase-8 activation to direct cell fate. Hughes et al. show that c-FLIPL/S recruitment to FADD is indirect and requires caspase-8. This co-operative hierarchical binding process explains the conundrum of the dual role of c-FLIPL and defines how c-FLIPL/S differentially control cell fate.
- Published
- 2016
- Full Text
- View/download PDF
39. Partial equilibrium approximations in apoptosis. II. The death-inducing signaling complex subsystem
- Author
-
Liu Hong, Ya-Jing Huang, and Wen-An Yong
- Subjects
Statistics and Probability ,Death Domain Receptor Signaling Adaptor Proteins ,Apoptosis ,computer.software_genre ,Models, Biological ,General Biochemistry, Genetics and Molecular Biology ,Humans ,Biochemical reactions ,Signaling process ,Upstream (networking) ,Mathematics ,General Immunology and Microbiology ,Applied Mathematics ,Partial equilibrium ,Mathematical Concepts ,General Medicine ,Human tumor ,Kinetics ,Modeling and Simulation ,Death-inducing signaling complex ,Data mining ,General Agricultural and Biological Sciences ,Biological system ,computer ,Signal Transduction - Abstract
This paper is a continuation of our previous work (Huang and Yong, 2013) for simplifying the Fas signaling-induced apoptotic pathway identified by Hua et al. (2005) for human tumor T cells. The previous paper studied the downstream intracelluar-signaling subsystem, while the present one is concerned with the upstream death-inducing signaling complex (DISC) subsystem. Under the assumption that the bind of Fas-associated death domains and FLICE-inhibitory proteins to the DISC is much faster than that of the initiator procaspases, we greatly simplify the upstream subsystem from 35 reactions with 26 species to 6 reactions with 9 species by adopting the classical and recently justified partial equilibrium approximation method. Numerical simulations show that the simplified model is in an excellent agreement with the original model. Most importantly, the simplified model clearly reveals the key reactants and dominated pathways in the Fas signaling process, and thus provides new insights into the apoptosis.
- Published
- 2015
40. Hypo-Expression of Flice-Inhibitory Protein and Activation of the Caspase-8 Apoptotic Pathways in the Death-Inducing Signaling Complex Due to Ischemia Induced by the Compression of the Asphyxiogenic Tool on the Skin in Hanging Cases
- Author
-
Alessandra De Matteis, Paola Frati, Giorgio Bolino, Vittorio Fineschi, Emanuela Turillazzi, and Aniello Maiese
- Subjects
0301 basic medicine ,c-FLIP ,Clinical Biochemistry ,hanging ,immunohistochemistry ,suicide ,vitality ,Caspase 8 ,Inhibitor of apoptosis ,Article ,03 medical and health sciences ,0302 clinical medicine ,Medicine ,030216 legal & forensic medicine ,Receptor ,lcsh:R5-920 ,business.industry ,Fas receptor ,030104 developmental biology ,Flip ,Apoptosis ,Death-inducing signaling complex ,Cancer research ,Tumor necrosis factor alpha ,lcsh:Medicine (General) ,business - Abstract
The FLICE-inhibitory protein (c-FLIPL) (55 kDa) is expressed in numerous tissues and most abundantly in the kidney, skeletal muscles and heart. The c-FLIPL has a region of homology with caspase-8 at the carboxy-terminal end which allows the molecule to assume a tertiary structure similar to that of caspases-8 and -10. Consequently, c-FLIPL acts as a negative inhibitor of caspase-8, preventing the processing and subsequent release of the pro-apoptotic molecule active form. The c-FLIP plays as an inhibitor of apoptosis induced by a variety of agents, such as tumor necrosis factor (TNF), T cell receptor (TCR), TNF-related apoptosis inducing ligand (TRAIL), Fas and death receptor (DR). Increased expression of c-FLIP has been found in many human malignancies and shown to be involved in resistance to CD95/Fas and TRAIL receptor-induced apoptosis. We wanted to verify an investigative protocol using FLIP to make a differential diagnosis between skin sulcus with vitality or non-vital skin sulcus in hanged subjects and those undergoing simulated hanging (suspension of the victim after murder). The study group consisted of 21 cases who died from suicidal hanging. The control group consisted of traumatic or natural deaths, while a third group consisted of simulated hanging cases. The reactions to the Anti-FLIP Antibody (Abcam clone-8421) was scored for each section with a semi-quantitative method by means of microscopic observation carried out with confocal microscopy and three-dimensional reconstruction. The results obtained allow us to state that the skin reaction to the FLIP is extremely clear and precise, allowing a diagnosis of unequivocal vitality and a very objective differentiation with the post-mortal skin sulcus.
- Published
- 2020
41. Cell Death
- Author
-
Nika N. Danial and David M. Hockenbery
- Subjects
Programmed cell death ,Cytochrome ,biology ,Apoptosis ,Death-inducing signaling complex ,biology.protein ,Death Receptors ,Mitochondrion ,Caspase ,Cell biology - Published
- 2018
42. Detection of RIPK1 in the FADD-Containing Death Inducing Signaling Complex (DISC) During Necroptosis
- Author
-
Rosalind L. Ang and Adrian T. Ting
- Subjects
0301 basic medicine ,Cell type ,biology ,Immunoprecipitation ,Chemistry ,Necroptosis ,urologic and male genital diseases ,Cell biology ,03 medical and health sciences ,RIPK1 ,030104 developmental biology ,0302 clinical medicine ,Apoptosis ,030220 oncology & carcinogenesis ,Death-inducing signaling complex ,biology.protein ,FADD ,biological phenomena, cell phenomena, and immunity ,Death domain - Abstract
FAS-associated protein with death domain (FADD) is a signaling molecule required by members of the TNF receptor superfamily (TNFRSF) such as FAS and TNFR1 to induce apoptosis. FADD is a small adapter molecule that functions as a scaffold to recruit procaspase-8 and other regulators. The FADD-containing signaling complex that initiates the apoptotic cascade has been termed the death inducing signaling complex (DISC). In the absence of FADD, death receptors cannot induce apoptosis and in appropriate cell types, these death receptors then induce necroptosis. Necroptosis can also be induced by death receptors in FADD-sufficient cells when caspase-8 is inhibited, usually accomplished by the addition of caspase inhibitors. Under such necroptotic conditions, the immunoprecipitation of FADD to isolate the DISC can be utilized to examine components of this complex. Here, we describe the immunoprecipitation of FADD and subsequent western-blotting to identify RIPK1 in this complex during necroptosis.
- Published
- 2018
43. The structure of the death receptor 4–TNF-related apoptosis-inducing ligand (DR4–TRAIL) complex
- Author
-
Michael L. Doyle, Steven Sheriff, Lin Cheng, Melissa Murdock, Wei Yong, Eric J. Lawrence, Vidhyashankar Ramamurthy, Aaron P. Yamniuk, Martin J. Corbett, and Lumelle A. Schneeweis
- Subjects
Models, Molecular ,musculoskeletal diseases ,Stereochemistry ,Molecular Sequence Data ,Biophysics ,TNF-Related Apoptosis-Inducing Ligand ,Plasma protein binding ,Calorimetry ,Biology ,Crystallography, X-Ray ,Biochemistry ,Research Communications ,immune system diseases ,Structural Biology ,Genetics ,Humans ,Amino Acid Sequence ,skin and connective tissue diseases ,Receptor ,Peptide sequence ,Condensed Matter Physics ,Fas receptor ,Ligand (biochemistry) ,Cell biology ,Receptors, TNF-Related Apoptosis-Inducing Ligand ,Apoptosis ,Death-inducing signaling complex ,Thermodynamics ,Crystallization ,Protein Binding - Abstract
The structure of death receptor 4 (DR4) in complex with TNF-related apoptosis-inducing ligand (TRAIL) has been determined at 3 Å resolution and compared with those of previously determined DR5–TRAIL complexes. Consistent with the high sequence similarity between DR4 and DR5, the overall arrangement of the DR4–TRAIL complex does not differ substantially from that of the DR5–TRAIL complex. However, subtle differences are apparent. In addition, solution interaction studies were carried out that show differences in the thermodynamics of binding DR4 or DR5 with TRAIL.
- Published
- 2015
44. Functional interaction between hMYH and hTRADD in the TNF-α-mediated survival and death pathways of HeLa cells
- Author
-
Soo-Hyun Hahm, Geon Tae Park, Ye Sun Han, Ji Hyung Chung, Se Hee Han, and An Hue Vy Tran
- Subjects
TRAF2 ,Programmed cell death ,Tumor Necrosis Factor-alpha ,Health, Toxicology and Mutagenesis ,NF-kappa B ,Apoptosis ,Biology ,TRADD ,TNF Receptor-Associated Death Domain Protein ,DNA Glycosylases ,Cell biology ,Necrosis ,Receptors, Tumor Necrosis Factor, Type I ,Death-inducing signaling complex ,Genetics ,Humans ,Protein Interaction Domains and Motifs ,Tumor necrosis factor alpha ,Signal transduction ,Molecular Biology ,HeLa Cells ,Signal Transduction ,Death domain - Abstract
The tumor necrosis factor (TNF) signaling pathway is a classical immune system pathway that plays a key role in regulating cell survival and apoptosis. The TNF receptor-associated death domain (TRADD) protein is recruited to the death domain of TNF receptor 1 (TNFR1), where it interacts with TNF receptor-associated factor 2 (TRAF2) and receptor-interacting protein (RIP) for the induction of apoptosis, necrosis, nuclear factor kappa-light-chain-enhancer of activated B cells (NFκB), and mitogen-activated protein (MAP) kinase activation. In this study, we found that the human MutY homolog (hMYH) interacted with human TRADD (hTRADD) via the C-terminal domain of hMYH. Moreover, under conditions promoting TNF-α-induced cell death or survival in HeLa cells, this interaction was weakened or enhanced, respectively. The interaction between hMYH and hTRADD was important for signaling pathways mediated by TNF-α. Our results also suggested that the hTRADD–hMYH association was involved in the nuclear translocation of NFκB and formation of the TNFR1–TRADD complex. Thus, this study identified a novel mechanism through which the hMYH–hTRADD interaction may affect the TNF-α signaling pathway. Implications In HeLa cells, the hTRADD–hMYH interaction functioned in both cell survival and apoptosis pathways following TNF-α stimulation.
- Published
- 2015
45. A RIPK3–Caspase 8 Complex Mediates Atypical Pro–IL-1β Processing
- Author
-
Kenta Moriwaki, John Bertin, Francis Ka-Ming Chan, and Peter J. Gough
- Subjects
Necroptosis ,Blotting, Western ,Interleukin-1beta ,Immunology ,Bone Marrow Cells ,Enzyme-Linked Immunosorbent Assay ,Transfection ,Caspase 8 ,Article ,Mice ,RIPK1 ,Animals ,Immunoprecipitation ,Immunology and Allergy ,Gene Knock-In Techniques ,FADD ,Caspase ,Inflammation ,Mice, Knockout ,biology ,NLRP1 ,Chemistry ,Dendritic Cells ,Cell biology ,Enzyme Activation ,Receptor-Interacting Protein Serine-Threonine Kinases ,Death-inducing signaling complex ,biology.protein ,Caspase 10 - Abstract
Caspase 8, the initiator caspase for death receptor–induced apoptosis, functions as a negative regulator of receptor interacting protein kinase 3 (RIPK3), an essential factor for TNF-, TLR3-, and TLR4-induced necroptosis. In certain situations, caspase 8 can also participate in pro–IL-1β processing. However, the biochemical complex that mediates caspase 8–mediated processing is not defined. In this study, we show that RIPK3 is crucial for caspase 1– and caspase 8–mediated pro–IL-1β and pro–IL-18 processing in bone marrow–derived dendritic cells (BMDCs) in response to LPS stimulation. Caspase 8–mediated pro–IL-1β processing requires intact RIPK1, RIPK3, TRIF, and FADD. In response to LPS, a complex that contains RIPK1, RIPK3, FADD, and caspase 8 is formed. Surprisingly, RIPK3-specific kinase inhibitors strongly enhanced caspase 8 activation and pro–IL-1β processing in LPS-stimulated BMDCs. However, studies in BMDCs expressing the kinase-inactive RIPK3-K51A mutant or RIPK1-K45A mutant showed that the kinase activity of neither RIPK1 nor RIPK3 is required for LPS-induced caspase 8 activation and IL-1β secretion. Hence, RIPK3 is an unexpected positive regulator of caspase 8 activity that promotes IL-1β maturation in BMDCs.
- Published
- 2015
46. RIP1 modulates death receptor mediated apoptosis and autophagy in macrophages
- Author
-
Juan Shi, Hui Guo, Zhenyu Yao, Peng Zhang, Shilian Liu, Dexian Zheng, and Yanxin Liu
- Subjects
Cancer Research ,Programmed cell death ,Apoptosis ,Biology ,Caspase 8 ,Models, Biological ,TNF-Related Apoptosis-Inducing Ligand ,Mice ,Autophagy ,Genetics ,Animals ,Humans ,Macrophage ,Research Articles ,Innate immune system ,Macrophages ,NF-kappa B ,Ubiquitination ,Membrane Proteins ,RNA-Binding Proteins ,Receptors, Death Domain ,General Medicine ,Cell biology ,Nuclear Pore Complex Proteins ,RAW 264.7 Cells ,Oncology ,Caspases ,Death-inducing signaling complex ,Molecular Medicine ,Beclin-1 ,Tumor necrosis factor alpha ,Apoptosis Regulatory Proteins - Abstract
Macrophages are responsible for defending against diverse pathogens and play a crucial role in the innate immune system. Macrophage's lifespan is determined by homeostatic balance between survival and apoptosis. Here we report that tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) triggers both apoptosis and autophagy in human U937 cells. Inhibition of autophagy facilitates TRAIL-induced apoptosis, suggesting that autophagy of macrophages protects against TRAIL-induced apoptosis. TRAIL treatment influences the expression of death receptors, indicating that TRAIL-induced apoptosis and autophagy are mediated by death receptors. RIP1 ubiquitination and expression regulate apoptosis and autophagy. Furthermore, expression and bioactivity of the p43/41-caspase-8 variant are critical to TRAIL-induced autophagy and apoptosis. Knockdown of RIP1 suppresses autophagy in macrophage. These data demonstrate that RIP1 is essential for the regulation of death receptor mediated autophagy and apoptosis. The results in this study contribute to understanding the regulation of autophagy and apoptosis in macrophages, and shed lights on death receptor-targeted therapy for cancer, inflammation and autoimmune diseases.
- Published
- 2014
47. Death effecter domain for the assembly of death-inducing signaling complex
- Author
-
Jin Kuk Yang
- Subjects
Inflammation ,Pharmacology ,Death Domain Receptor Signaling Adaptor Proteins ,Cancer Research ,Chain model ,biology ,Biochemistry (medical) ,Clinical Biochemistry ,Pharmaceutical Science ,Apoptosis ,Cell Biology ,Cell biology ,Death-inducing signaling complex ,biology.protein ,Animals ,Humans ,Protein Interaction Domains and Motifs ,FADD ,Protein Multimerization ,Signal transduction ,Receptor ,Extrinsic apoptosis ,Caspase ,Signal Transduction - Abstract
Death-inducing signaling complex (DISC) is a platform for the activation of initiator caspase in extrinsic apoptosis. Assembly of DISC is accomplished by two different types of homotypic interaction: one is between death domains (DDs) of a death receptor and FADD, and the other is between death effecter domains (DEDs) of FADD, procaspase-8/-10 and cFLIP. Recent biochemical investigations on the stoichiometry of DISC have revealed that single-DED-containing FADD exists in DISC in a substantially lower abundance than the sum of tandem-DEDs-containing components that are procaspase-8 and cFLIP. In addition, the homology models of the tandem DEDs in procaspase-8 and cFLIP show that two different interaction faces, H1-H4 face and H2-H5 face, are exposed for possible inter-molecular DED-DED interactions. These recent findings led to a proposal of the DED chain model for the interactions between FADD, procaspase-8 and cFLIP in DISC. This emerging view provides new insights on the topology of DED-DED network in DISC and furthermore on how procaspase-8 and cFLIP cluster for dimerization and proteolytic activation.
- Published
- 2014
48. Principles and mechanisms of CD95 activation
- Author
-
Harald Wajant
- Subjects
Programmed cell death ,Protein Conformation ,media_common.quotation_subject ,Clinical Biochemistry ,Apoptosis ,chemical and pharmacologic phenomena ,Caspase 8 ,Biochemistry ,Membrane Microdomains ,hemic and lymphatic diseases ,Animals ,Humans ,fas Receptor ,Internalization ,Molecular Biology ,Lipid raft ,Caspase ,media_common ,biology ,hemic and immune systems ,Fas receptor ,biological factors ,Cell biology ,Actin Cytoskeleton ,Death-inducing signaling complex ,biology.protein ,Protein Multimerization ,biological phenomena, cell phenomena, and immunity ,Signal Transduction - Abstract
CD95 (Apo1/Fas) has been originally identified as the target of cell death-inducing antibodies. The recognition of CD95 as an apoptosis-triggering receptor represents one of the early milestones in the apoptosis field. Moreover, the research on CD95-induced cell death fostered various other discoveries of broad and general relevance in cell biology, for example, the identification of caspase 8 as the initiator caspase of the extrinsic apoptosis pathway. Activation of CD95-associated intracellular signaling pathways is not a simple consequence of ligand binding but is the fine-tuned result of a complex interplay of various molecular mechanisms that eventually determine the strength and quality of the CD95 response. There is growing evidence that different forms of CD95 stimulation trigger the assembly of CD95 signaling complexes of distinct composition. Moreover, the formation of signaling competent CD95 complexes is a multistep process and the subject of regulation by various cellular cues. This review addresses the relevance of the molecular nature of the CD95-stimulating agonist for the quality of the CD95 response and discusses the importance of modification, clustering, internalization, and lipid raft and actin association of CD95 for CD95 activity.
- Published
- 2014
49. SOCS-1 ameliorates smoke inhalation-induced acute lung injury through inhibition of ASK-1 activity and DISC formation
- Author
-
Yating Ma, Hang Zhao, Weike Su, Xiaoming Chen, Qiwen Shi, Chenming Xu, Leifang Zhang, and Kairui Zhu
- Subjects
0301 basic medicine ,ARDS ,Death Domain Receptor Signaling Adaptor Proteins ,Smoke inhalation ,medicine.medical_treatment ,Immunology ,Acute Lung Injury ,Apoptosis ,Lung injury ,Biology ,MAP Kinase Kinase Kinase 5 ,03 medical and health sciences ,Suppressor of Cytokine Signaling 1 Protein ,medicine ,Immunology and Allergy ,Humans ,Lung ,Cells, Cultured ,Caspase 8 ,Suppressor of cytokine signaling 1 ,Smoke Inhalation Injury ,medicine.disease ,TNF Receptor-Associated Factor 2 ,TNF Receptor-Associated Death Domain Protein ,030104 developmental biology ,Cytokine ,Death-inducing signaling complex ,Cancer research ,Tumor necrosis factor alpha - Abstract
Smoke inhalation leads to acute lung injury (ALI), a devastating clinical problem associated with high mortality. Suppressor of cytokine signaling-1 (SOCS-1) is a negative regulator of apoptosis and pro-inflammatory cytokine signaling, two major contributors to the pathogenesis of ALI. We have found that SOCS-1 protects lung epithelial cells from smoke-induced apoptosis through two mechanisms. One is that SOCS-1 enhances degradation of ASK-1 and diminishes cleavage of pro-caspase-3 to repress smoke-triggered apoptosis in lung epithelial cells. The other is that SOCS-1 represses smoke-triggered DISC formation through altering TRADD-caspase-8 interaction rather than TNFR-1-TRADD interaction or TNFR-1-TRAF-2 interaction. In conclusion, SOCS-1 relieves smoke inhalation-induced lung injury by repressing ASK-1 and DISC-mediated epithelium apoptosis.
- Published
- 2017
50. STAT1 mediates transmembrane TNF-alpha-induced formation of death-inducing signaling complex and apoptotic signaling via TNFR1
- Author
-
Hongping Ba, Lu Han, Xuena Hu, Zunyue Zhang, Xiang-Ping Yang, Min Yu, Jing Wang, Bingjiao Yin, Yaping Jiang, Kun Yang, and Zhuoya Li
- Subjects
0301 basic medicine ,Death Domain Receptor Signaling Adaptor Proteins ,Fas-Associated Death Domain Protein ,Apoptosis ,Caspase 8 ,Amino Acid Chloromethyl Ketones ,Cell Line ,03 medical and health sciences ,Mice ,Cadaverine ,Animals ,Humans ,FADD ,Phosphorylation ,Molecular Biology ,Death domain ,Original Paper ,biology ,Tumor Necrosis Factor-alpha ,NF-kappa B ,Cell Biology ,TRADD ,TNF Receptor-Associated Death Domain Protein ,Cell biology ,030104 developmental biology ,HEK293 Cells ,STAT1 Transcription Factor ,Receptors, Tumor Necrosis Factor, Type I ,Death-inducing signaling complex ,STAT protein ,Cancer research ,biology.protein ,NIH 3T3 Cells ,Tumor necrosis factor alpha ,Signal transduction ,Protein Binding ,Signal Transduction - Abstract
Tumor necrosis factor-alpha (TNF-α) exists in two forms: secretory TNF-α (sTNF-α) and transmembrane TNF-α (tmTNF-α). Although both forms of TNF-α induce tumor cell apoptosis, tmTNF-α is able to kill tumor cells that are resistant to sTNF-α-mediated cytotoxicity, indicating their differences in signal transduction. Here, we demonstrate that internalization of TNFR1 is crucial for sTNF-α- but not for tmTNF-α-induced apoptosis. sTNF-α induces binding of tumor necrosis factor receptor type 1-associated death domain protein (TRADD) to the death domain (DD) of TNFR1 and subsequent activation of nuclear factor kappa B (NF-κB), and the formation of death-inducing signaling complexes (DISCs) in the cytoplasm after internalization. In contrast, tmTNF-α induces DISC formation on the membrane in a DD-independent manner. It leads to the binding of signal transducer and activator of transcription 1 (STAT1) to a region spanning amino acids 319-337 of TNFR1 and induces phosphorylation of serine at 727 of STAT1. The phosphorylation of STAT1 promotes its binding to TRADD, and thus recruits Fas-associated protein with DD (FADD) and caspase 8 to form DISC complexes. This STAT1-dependent signaling results in apoptosis but not NF-κB activation. STAT1-deficiency in U3A cells counteracts tmTNF-α-induced DISC formation and apoptosis. Conversely, reconstitution of STAT1 expression restores tmTNF-α-induced apoptotic signaling in the cell line. Consistently, tmTNF-α suppresses the growth of STAT1-containing HT1080 tumors, but not of STAT1-deficient U3A tumors in vivo. Our data reveal an unappreciated molecular mechanism of tmTNF-α-induced apoptosis and may provide a new clue for cancer therapy.
- Published
- 2017
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.