116 results on '"Death effector domain"'
Search Results
2. FasL-Independent Activation of Fas
- Author
-
Mollinedo, Faustino, Gajate, Consuelo, and Wajant, Harald
- Published
- 2006
- Full Text
- View/download PDF
3. Regulation of Fas Signaling by FLIP Proteins
- Author
-
Thome, Margot and Wajant, Harald
- Published
- 2006
- Full Text
- View/download PDF
4. Mammalian Cell Death Pathways : Intrinsic and Extrinsic
- Author
-
McDonald, E. Robert, III, El-Deiry, Wafik S., Teicher, Beverly A., editor, and El-Deiry, Wafik S., editor
- Published
- 2005
- Full Text
- View/download PDF
5. Death domain fold proteins in immune signaling and transcriptional regulation
- Author
-
Yu-San Huoh and Sun Hur
- Subjects
0301 basic medicine ,Inflammasomes ,Biology ,Biochemistry ,Pyrin domain ,Article ,03 medical and health sciences ,0302 clinical medicine ,Death Domain ,medicine ,Transcriptional regulation ,Receptor ,Molecular Biology ,Biological Phenomena ,Death domain ,Innate immune system ,Inflammasome ,Cell Biology ,Cell biology ,CARD Signaling Adaptor Proteins ,Cytoskeletal Proteins ,030104 developmental biology ,030220 oncology & carcinogenesis ,Death effector domain ,Signal transduction ,Signal Transduction ,medicine.drug - Abstract
Death domain fold (DDF) superfamily comprises of the death domain (DD), death effector domain (DED), caspase activation recruitment domain (CARD), and pyrin domain (PYD). By utilizing a conserved mode of interaction involving six distinct surfaces, a DDF serves as a building block that can densely pack into homomultimers or filaments. Studies of immune signaling components have revealed that DDF-mediated filament formation plays a central role in mediating signal transduction and amplification. The unique ability of DDFs to self-oligomerize upon external signals and induce oligomerization of partner molecules underlies key processes in many innate immune signaling pathways, as exemplified by RIG-I-like receptor signalosome and inflammasome assembly. Recent studies showed that DDFs are not only limited to immune signaling pathways, but also are involved with transcriptional regulation and other biological processes. Considering that DDF annotation still remains a challenge, the current list of DDFs and their functions may represent just the tip of the iceberg within the full spectrum of DDF biology. In this review, we discuss recent advances in our understanding of DDF functions, structures, and assembly architectures with a focus on CARD- and PYD-containing proteins. We also discuss areas of future research and the potential relationship of DDFs with biomolecular condensates formed by liquid-liquid phase separation (LLPS).
- Published
- 2021
6. Alternative Pre-mRNA Splicing and Regulation of Programmed Cell Death
- Author
-
Wu, J. Y., Tang, H., Havlioglu, N., Müller, W. E. G., editor, Jeanteur, Ph., editor, Kostovic, I., editor, Kuchino, Y., editor, Macieira-Coelho, A., editor, Rhoads, R. E., editor, and Jeanteur, Philippe, editor
- Published
- 2003
- Full Text
- View/download PDF
7. Apoptosis
- Author
-
Wong, Henry K., Tsokos, George C., editor, and Atkins, James L., editor
- Published
- 2003
- Full Text
- View/download PDF
8. The Death Receptor Family and the Extrinsic Pathway
- Author
-
Guicciardi, Maria Eugenia, Gores, Gregory J., Yin, Xiao-Ming, editor, and Dong, Zheng, editor
- Published
- 2003
- Full Text
- View/download PDF
9. The Death Receptors
- Author
-
Peter, Marcus E., Scaffidi, Carsten, Medema, Jan Paul, Kischkel, Frank, Krammer, Peter H., Hennig, W., editor, Nover, L., editor, Scheer, U., editor, and Kumar, Sharad, editor
- Published
- 1999
- Full Text
- View/download PDF
10. Molecular Mechanisms of TNF Receptor-Mediated Signaling
- Author
-
Malek, N. P., Pluempe, J., Kubicka, S., Manns, M. P., Trautwein, C., Schlag, P. M., editor, Senn, H.-J., editor, Diehl, V., editor, Parkin, D. M., editor, Rajewsky, M. F., editor, Rubens, R., editor, Wannenmacher, M., editor, Oldhafer, Karl J., editor, Lang, Hauke, editor, and Pichlmayr, Rudolf, editor
- Published
- 1998
- Full Text
- View/download PDF
11. Measuring Composition of CD95 Death-Inducing Signaling Complex and Processing of Procaspase-8 in this Complex
- Author
-
Laura K. Hillert-Richter and Inna N. Lavrik
- Subjects
Caspase 8 ,Death Domain Receptor Signaling Adaptor Proteins ,General Immunology and Microbiology ,biology ,Chemistry ,Immunoprecipitation ,General Chemical Engineering ,General Neuroscience ,Signal transducing adaptor protein ,Apoptosis ,Fas receptor ,General Biochemistry, Genetics and Molecular Biology ,Cell biology ,CD95 death-inducing signaling complex ,biology.protein ,Death effector domain ,fas Receptor ,FADD ,Signal Transduction ,Death domain - Abstract
Extrinsic apoptosis is mediated by the activation of death receptors (DRs) such as CD95/Fas/APO-1 or tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-receptor 1/receptor 2 (TRAIL-R1/R2). Stimulation of these receptors with their cognate ligands leads to the assembly of the death-inducing signaling complex (DISC). DISC comprises DR, the adaptor protein Fas-associated protein with death domain (FADD), procaspases-8/-10, and cellular FADD-like interleukin (IL)-1β-converting enzyme-inhibitory proteins (c-FLIPs). The DISC serves as a platform for procaspase-8 processing and activation. The latter occurs via its dimerization/oligomerization in the death effector domain (DED) filaments assembled at the DISC. Activation of procaspase-8 is followed by its processing, which occurs in several steps. In this work, an established experimental workflow is described that allows the measurement of DISC formation and the processing of procaspase-8 in this complex. The workflow is based on immunoprecipitation techniques supported by western blot analysis. This workflow allows careful monitoring of different steps of procaspase-8 recruitment to the DISC and its processing and is highly relevant for investigating molecular mechanisms of extrinsic apoptosis.
- Published
- 2021
12. Heterologous Expression and Auto-Activation of Human Pro-Inflammatory Caspase-1 in Saccharomyces cerevisiae and Comparison to Caspase-8
- Author
-
Marta Valenti, María Molina, and Víctor J. Cid
- Subjects
0301 basic medicine ,Saccharomyces cerevisiae Proteins ,death domain ,Saccharomyces cerevisiae ,Immunology ,Caspase 1 ,caspase-1 ,yeast ,Caspase 8 ,Substrate Specificity ,03 medical and health sciences ,Galactokinase ,0302 clinical medicine ,Gene Expression Regulation, Fungal ,Humans ,Immunology and Allergy ,Caspase ,Death domain ,Original Research ,Microbial Viability ,biology ,Chemistry ,Microfilament Proteins ,Intracellular Signaling Peptides and Proteins ,heterologous expression ,Phosphate-Binding Proteins ,RC581-607 ,biology.organism_classification ,Actin cytoskeleton ,Cell biology ,Mitochondria ,Enzyme Activation ,Actin Cytoskeleton ,030104 developmental biology ,Apoptosis ,030220 oncology & carcinogenesis ,Enzyme Induction ,biology.protein ,Death effector domain ,humanized yeast models ,Immunologic diseases. Allergy - Abstract
Caspases are a family of cysteine proteases that play an essential role in inflammation, apoptosis, cell death, and development. Here we delve into the effects caused by heterologous expression of human caspase-1 in the yeast Saccharomyces cerevisiae and compare them to those of caspase-8. Overexpression of both caspases in the heterologous model led to their activation and caused mitochondrial hyperpolarization, damage to different organelles, and cell death. All these effects were dependent on their protease activity, and caspase-8 was more aggressive than caspase-1. Growth arrest could be at least partially explained by dysfunction of the actin cytoskeleton as a consequence of the processing of the yeast Bni1 formin, which we identify here as a likely direct substrate of both caspases. Through the modulation of the GAL1 promoter by using different galactose:glucose ratios in the culture medium, we have established a scenario in which caspase-1 is sufficiently expressed to become activated while yeast growth is not impaired. Finally, we used the yeast model to explore the role of death-fold domains (DD) of both caspases in their activity. Peculiarly, the DDs of either caspase showed an opposite involvement in its intrinsic activity, as the deletion of the caspase activation and recruitment domain (CARD) of caspase-1 enhanced its activity, whereas the deletion of the death effector domain (DED) of caspase-8 diminished it. We show that caspase-1 is able to efficiently process its target gasdermin D (GSDMD) when co-expressed in yeast. In sum, we propose that S. cerevisiae provides a manageable tool to explore caspase-1 activity and structure–function relationships.
- Published
- 2021
- Full Text
- View/download PDF
13. The first cloned sea cucumber FADD from Holothuria leucospilota: Molecular characterization, inducible expression and involvement of apoptosis
- Author
-
Xiao Jiang, Chunhua Ren, Wen Huang, Ting Chen, Xiaofen Wu, Lin Zhao, and Hongyan Sun
- Subjects
Lipopolysaccharides ,0301 basic medicine ,Untranslated region ,Fas-Associated Death Domain Protein ,Apoptosis ,Aquatic Science ,Biology ,03 medical and health sciences ,Complementary DNA ,Animals ,Holothuria ,Humans ,Environmental Chemistry ,Amino Acid Sequence ,FADD ,Phylogeny ,Death domain ,Messenger RNA ,Base Sequence ,Gene Expression Profiling ,Holothuria leucospilota ,04 agricultural and veterinary sciences ,General Medicine ,biology.organism_classification ,Immunity, Innate ,Up-Regulation ,Cell biology ,Open reading frame ,HEK293 Cells ,Poly I-C ,030104 developmental biology ,Gene Expression Regulation ,040102 fisheries ,biology.protein ,0401 agriculture, forestry, and fisheries ,Death effector domain ,Sequence Alignment - Abstract
In this study, a sea cucumber Fas-associated death domain (FADD) named HLFADD was first cloned from Holothuria leucospilota. The full-length cDNA of HLFADD is 2137 bp in size, containing a 116-bp 5′-untranslated region (UTR), a 1334-bp 3′-UTR and a 687-bp open reading frame (ORF) encoding a protein of 228 amino acids with a deduced molecular weight of 26.42 kDa. HLFADD protein contains a conserved death effector domain at its N-terminal and a conserved death domain at its C-terminal, structurally similar to its counterparts in vertebrates. The over-expressed HLFADD protein could induce apoptosis in HEK293 cells, suggesting a possible death receptor-mediated apoptosis pathway in echinoderms adapted with FADD. Moreover, HLFADD mRNA is ubiquitously expressed in all examined tissues, with the highest transcript level in the coelomocytes, followed by intestine. In vitro experiments performed in the H. leucospilota coelomocytes, the expression of HLFADD mRNA was significantly up-regulated by lipopolysaccharides (LPS) or polyriboinosinic-polyribocytidylic acid [poly (I:C)] challenge, suggesting that HLFADD might play important roles in the innate immune defense of sea cucumber against the invasion of bacteria and viruses.
- Published
- 2019
14. Platelets express adaptor proteins of the extrinsic apoptosis pathway and can activate caspase-8
- Author
-
Oliver Speer, Julia J.M. Eekels, Nadine Goelz, Christoph T. Kamber, Markus Schmugge, Milica Pantic, Francesca D. Franzoso, University of Zurich, Lebedeva, Irina V, and Schmugge, Markus
- Subjects
Male ,Confocal Microscopy ,Physiology ,Fas-Associated Death Domain Protein ,Protein Expression ,Protein Deglycase DJ-1 ,Apoptosis ,Fas ligand ,Spectrum Analysis Techniques ,Animal Cells ,Medicine and Health Sciences ,FADD ,Scanning Confocal Microscopy ,Child ,Microscopy ,Caspase 8 ,Multidisciplinary ,Cell Death ,biology ,Chemistry ,Light Microscopy ,Signal transducing adaptor protein ,Hematology ,General Medicine ,Flow Cytometry ,Body Fluids ,Cell biology ,Protein Transport ,Blood ,Cell Processes ,Spectrophotometry ,Medicine ,Female ,Death effector domain ,Cytophotometry ,Anatomy ,Cellular Types ,General Agricultural and Biological Sciences ,Research Article ,Platelets ,Blood Platelets ,Science ,Immunology ,610 Medicine & health ,Genetics and Molecular Biology ,Research and Analysis Methods ,Blood Plasma ,Autoimmune Diseases ,Gene Expression and Vector Techniques ,Humans ,Platelet activation ,Molecular Biology Techniques ,Blood Coagulation ,Molecular Biology ,Death domain ,Molecular Biology Assays and Analysis Techniques ,1000 Multidisciplinary ,Blood Cells ,Idiopathic Thrombocytopenic Purpura ,Intrinsic apoptosis ,Biology and Life Sciences ,Cell Biology ,Platelet Activation ,Thrombocytopenia ,TRADD ,Enzyme Activation ,Gene Expression Regulation ,10036 Medical Clinic ,General Biochemistry ,biology.protein ,Clinical Immunology ,Clinical Medicine - Abstract
Background Apoptotic pathways in platelets are important for their survival and function. Platelet apoptosis may be involved in the pathogenesis of immune thrombocytopenia (ITP), an autoimmune-mediated disease. In contrast to the intrinsic apoptosis pathway, not much is known about the extrinsic pathway mechanisms in platelets. Objectives To investigate the expression of proteins involved in the extrinsic apoptosis pathway, including the death receptors, adaptor and regulator proteins in human platelets. To determine a possible trigger of the extrinsic apoptosis pathway in platelets. Methods To investigate the expression of key markers of the extrinsic pathway we used targeted immunofluorescence and flow cytometry assays. To study their expression and interaction we performed Western blotting and co-immunoprecipitation. Treated platelets with different apoptosis triggers were subjected to flow cytometry. Results We could identify the protein expression of the pro-apoptotic proteins TRADD (Tumor Necrosis Factor Receptor type 1- Associated DEATH Domain protein), TRAF2/5, (TNF Associated Factor) and DEDAF (Death Effector Domain- Associated Factor), FADD (Fas-Associated protein with death domain) as well as the anti-apoptotic proteins DJ-1 (Deglycase 1) and c-FLIP in human platelets. ABT-737 treatment induced a disruption in the co-localization of DJ-1 with FADD. Platelets treated with ABT-737 showed an activation in caspase-3 and -8. The exposure to TNF (Tumor Necrosis Factor), FasL (Fas ligand), and TWEAK or to plasma derived from ITP patients, did not lead to changes in caspase-3 and -8 activation in platelets. Conclusions Human platelets express some proteins of the extrinsic apoptosis pathway which can be modulated only by ABT-737 treatment. However so far, no other apoptosis trigger or interaction with an external receptor have been yet identified.
- Published
- 2021
15. 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
16. 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
17. Structural insights of homotypic interaction domains in the ligand-receptor signal transduction of tumor necrosis factor (TNF)
- Author
-
Young-Hoon Park, Mi Suk Jeong, and Se Bok Jang
- Subjects
0301 basic medicine ,Protein domain ,TNF ,Apoptosis ,DD ,Ligands ,Biochemistry ,Pyrin domain ,Receptors, Tumor Necrosis Factor ,03 medical and health sciences ,0302 clinical medicine ,Protein Domains ,Animals ,Humans ,Molecular Biology ,Caspase ,DED ,Death domain ,biology ,Contributed Mini Review ,General Medicine ,Cell biology ,030104 developmental biology ,030220 oncology & carcinogenesis ,Multigene Family ,Tumor Necrosis Factors ,biology.protein ,Immunoglobulin superfamily ,Death effector domain ,Tumor necrosis factor alpha ,Signal transduction ,Signal Transduction - Abstract
Several members of tumor necrosis factor receptor (TNFR) superfamily that these members activate caspase-8 from death-inducing signaling complex (DISC) in TNF ligand-receptor signal transduction have been identified. In the extrinsic pathway, apoptotic signal transduction is induced in death domain (DD) superfamily; it consists of a hexahelical bundle that contains 80 amino acids. The DD superfamily includes about 100 members that belong to four subfamilies: death domain (DD), caspase recruitment domain (CARD), pyrin domain (PYD), and death effector domain (DED). This superfamily contains key building blocks: with these blocks, multimeric complexes are formed through homotypic interactions. Furthermore, each DD-binding event occurs exclusively. The DD superfamily regulates the balance between death and survival of cells. In this study, the structures, functions, and unique features of DD superfamily members are compared with their complexes. By elucidating structural insights of DD superfamily members, we investigate the interaction mechanisms of DD domains; these domains are involved in TNF ligand-receptor signaling. These DD superfamily members play a pivotal role in the development of more specific treatments of cancer. [BMB Reports 2016; 49(3): 159-166].
- Published
- 2016
18. Fuzzy support vector machine model to predict human death domain protein–protein interactions
- Author
-
Nemade, Prakash A. and Pardasani, Kamal R.
- Published
- 2015
- Full Text
- View/download PDF
19. The Role of Death Domains Superfamily in Multiple Sclerosis Pathogenesis
- Author
-
Abbas Mirshafiey and Kabir Magaji Hamid
- Subjects
biology ,Multiple sclerosis ,Experimental autoimmune encephalomyelitis ,medicine.disease ,Pyrin domain ,Oligodendrocyte ,Cell biology ,Myelin ,medicine.anatomical_structure ,medicine ,biology.protein ,Death effector domain ,Neuroscience ,Caspase ,Death domain - Abstract
Multiple sclerosis (MS) and its animal model, experimental autoimmune encephalomyelitis (EAE), are inflammatory diseases of the central nervous system (CNS), mediated by several immune cells. Oligodendrocytes are responsible for the formation and maintenance of myelin around multiple axons. In MS oligodendrocytes are the targets of inflammatory and immune attacks. Thus, the destruction of a single oligodendrocyte, possibly by apoptosis, results in the loss of myelin around several axons and the loss of many oligodendrocytes limiting the ability to repair or regenerate demyelinated areas. Apoptosis is mediated by an aggregation of various protein components, specifically death domains (DD) superfamily. This superfamily is composed of the death domain (DD), the death effector domain (DED), the caspase recruitment domain (CARD) and the pyrin domain (PYD) subfamilies. Within each subfamily, members form homotypic interactions and facilitate the assembly of oligomeric signaling complexes. Members of the death domain superfamily are critical components of apoptotic and inflammatory signaling. We summarize the structure and functions of the DD superfamily, and describe the role of the DD proteins in oligodendrocytes death and proinflammatory activation in MS pathogenesis.
- Published
- 2015
20. DDIAS suppresses TRAIL-mediated apoptosis by inhibiting DISC formation and destabilizing caspase-8 in cancer cells
- Author
-
Seung-Ho Park, Kyeong Eun Jung, Hyun Seung Ban, Misun Won, Bokyung Kim, Ji-Young Lee, and Joo-Young Im
- Subjects
0301 basic medicine ,Cancer Research ,Death Domain Receptor Signaling Adaptor Proteins ,Carcinoma, Hepatocellular ,Lung Neoplasms ,Apoptosis ,Cell Cycle Proteins ,Caspase 8 ,TNF-Related Apoptosis-Inducing Ligand ,03 medical and health sciences ,Carcinoma, Non-Small-Cell Lung ,Genetics ,Biomarkers, Tumor ,Tumor Cells, Cultured ,Humans ,FADD ,Protein kinase A ,Molecular Biology ,Death domain ,Cell Proliferation ,biology ,Cell growth ,Liver Neoplasms ,030104 developmental biology ,Cancer cell ,Cancer research ,biology.protein ,Death effector domain ,Apoptosis Regulatory Proteins - Abstract
DNA damage-induced apoptosis suppressor (DDIAS) has an anti-apoptotic function during DNA damage in lung cancer. However, the anti-apoptotic mechanism of DDIAS in cancer cells under other conditions has not been reported. We report here that DDIAS protects cancer cells from tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-induced apoptosis by two distinct mechanisms in non-small cell lung cancer (NSCLC) and hepatocellular carcinoma (HCC) cells. DDIAS depletion sensitized NSCLC and HCC cells to TRAIL-mediated apoptosis, an effect that was abrogated by pharmacological or genetic inhibition of caspase-8 and was independent of caspase-9, p53, or mitogen-activated protein kinase signaling. Interestingly, we found that the N terminus of DDIAS interacted with the death effector domain of Fas-associated protein death domain (FADD) and prevented its recruitment to the death-inducing signaling complex (DISC), thereby blocking caspase-8 activation. DDIAS knockdown also suppressed epidermal growth factor-induced phosphorylation of p90 ribosomal S6 kinase (RSK) 2 and stabilized caspase-8 by preventing its ubiquitination and proteasomal degradation. This effect was abolished by RSK2 overexpression. Taken together, DDIAS has dual functions in inhibiting DISC formation as well as in destabilizing caspase-8, thereby suppressing TRAIL-mediated apoptosis of cancer cells. Thus, we suggest that DDIAS can serve as an effective therapeutic target in the treatment of NSCLC and HCC.
- Published
- 2017
21. Phosphoprotein enriched in astrocytes (PEA)-15: A potential therapeutic target in multiple disease states
- Author
-
Graeme Nixon and Fiona H. Greig
- Subjects
TRAIL, tumour necrosis factor-related apoptosis-inducing ligand ,Protein Conformation ,Proliferation ,MAP, mitogen-activated protein ,Apoptosis ,Bioinformatics ,PDGF, platelet-derived growth factor ,CaMKII, calcium/calmodulin-dependent protein kinase II ,Neoplasms ,PEA-15, phosphoprotein enriched in astrocytes-15 ,RSK, ribosomal s6 kinase ,Medicine ,Pharmacology (medical) ,FADD ,DED, death effector domain ,Cancer ,Brain Diseases ,ERK1/2, extracellular signal-regulated kinases 1/2 ,ERK1/2 ,biology ,Kinase ,Intracellular Signaling Peptides and Proteins ,food and beverages ,Type 2 diabetes ,PEA-15 ,GLUT, glucose transporter ,Cell biology ,Cardiovascular Diseases ,TGF-β1, transforming growth factor-β1 ,Phosphorylation ,Death effector domain ,Associate editor: S. Kennedy ,HNF-4α, hepatocyte nuclear factor 4alpha ,Endocrine System Diseases ,NSCLC, non-small cell lung cancer ,Ca2+/calmodulin-dependent protein kinase ,PKC, protein kinase C ,Animals ,Humans ,PLD1, phospholipase D1 ,Protein kinase C ,Death domain ,Pharmacology ,DISC, death initiation signalling complex ,business.industry ,Phosphoproteins ,FADD, Fas-associated death domain protein ,IL, interleukin ,Phosphoprotein ,VSM, vascular smooth muscle ,biology.protein ,Apoptosis Regulatory Proteins ,PCOS, polycystic ovary syndrome ,business - Abstract
Phosphoprotein enriched in astrocytes-15 (PEA-15) is a cytoplasmic protein that sits at an important junction in intracellular signalling and can regulate diverse cellular processes, such as proliferation and apoptosis, dependent upon stimulation. Regulation of these processes occurs by virtue of the unique interaction of PEA-15 with other signalling proteins. PEA-15 acts as a cytoplasmic tether for the mitogen-activated protein kinases, extracellular signal-regulated kinase 1/2 (ERK1/2) preventing nuclear localisation. In order to release ERK1/2, PEA-15 requires to be phosphorylated via several potential pathways. PEA-15 (and its phosphorylation state) therefore regulates many ERK1/2-dependent processes, including proliferation, via regulating ERK1/2 nuclear translocation. In addition, PEA-15 contains a death effector domain (DED) which allows interaction with other DED-containing proteins. PEA-15 can bind the DED-containing apoptotic adaptor molecule, Fas-associated death domain protein (FADD) which is also dependent on the phosphorylation status of PEA-15. PEA-15 binding of FADD can inhibit apoptosis as bound FADD cannot participate in the assembly of apoptotic signalling complexes. Through these protein–protein interactions, PEA-15-regulated cellular effects have now been investigated in a number of disease-related studies. Changes in PEA-15 expression and regulation have been observed in diabetes mellitus, cancer, neurological disorders and the cardiovascular system. These changes have been suggested to contribute to the pathology related to each of these disease states. As such, new therapeutic targets based around PEA-15 and its associated interactions are now being uncovered and could provide novel avenues for treatment strategies in multiple diseases.
- Published
- 2014
22. Structural determinants of DISC function: New insights into death receptor-mediated apoptosis signalling
- Author
-
Afshin Samali, Tamás Sessler, Eva Szegezdi, and Sandra Healy
- Subjects
Pharmacology ,Death Domain Receptor Signaling Adaptor Proteins ,Apoptosis ,Receptors, Death Domain ,Biology ,Fas receptor ,TRADD ,Cell biology ,TNF receptor associated factor ,biology.protein ,Animals ,Humans ,Pharmacology (medical) ,Death effector domain ,FADD ,Signal transduction ,Receptor ,Protein Processing, Post-Translational ,Signal Transduction ,Death domain - Abstract
Death receptors are members of the tumour necrosis factor (TNF) receptor superfamily characterised by an ~80 amino acid long alpha-helical fold, termed the death domain (DD). Death receptors diversified during early vertebrate evolution indicating that the DD fold has plasticity and specificity that can be easily adjusted to attain additional functions. Eight members of the death receptor family have been identified in humans, which can be divided into four structurally homologous groups or clades, namely: the p75(NTR) clade (consisting of ectodysplasin A receptor, death receptor 6 (DR6) and p75 neurotrophin (NTR) receptor); the tumour necrosis factor receptor 1 clade (TNFR1 and DR3), the CD95 clade (CD95/FAS) and the TNF-related apoptosis-inducing ligand receptor (TRAILR) clade (TRAILR1 and TRAILR2). Receptors in the same clade participate in similar processes indicating that structural diversification enabled functional specialisation. On the surface of nearly all human cells multiple death receptors are expressed, enabling the cell to respond to a plethora of external signals. Activation of different death receptors converges on the activation of three main signal transduction pathways: nuclear factor-κB-mediated differentiation or inflammation, mitogen-associated protein kinase-mediated stress response and caspase-mediated apoptosis. While the ability to induce cell death is true for nearly all DRs, the FAS and TRAILR clades have specialised in inducing cell death. Here we summarise recent discoveries about the molecular regulation and structural requirements of apoptosis induction by death receptors and discuss how this information can be used to better explain the biological functions, similarities and distinguishing features of death receptors.
- Published
- 2013
23. 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
-
Jay M. McDonald, Tong Zhou, Qi Yan, and Yuhua Song
- Subjects
Fas-Associated Death Domain Protein ,Static Electricity ,Molecular Dynamics Simulation ,urologic and male genital diseases ,Caspase 8 ,Biochemistry ,Protein Structure, Secondary ,Article ,Fas ligand ,Structural Biology ,Protein Interaction Mapping ,fas Receptor ,FADD ,Molecular Biology ,Death domain ,Principal Component Analysis ,Binding Sites ,biology ,Protein Stability ,Chemistry ,Computational Biology ,Fas receptor ,Cell biology ,Multiprotein Complexes ,Death-inducing signaling complex ,biology.protein ,Caspase 10 ,Death effector domain ,Protein Multimerization ,biological phenomena, cell phenomena, and immunity ,Protein Binding ,Signal Transduction - 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.
- Published
- 2012
24. Regulating TRAIL Receptor-Induced Cell Death at the Membrane: A Deadly Discussion
- Author
-
Sarah Shirley, Olivier Micheau, Alexandre Morizot, Lipides - Nutrition - Cancer (U866) (LNC), Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Bourgogne (UB)-Ecole Nationale Supérieure de Biologie Appliquée à la Nutrition et à l'Alimentation de Dijon (ENSBANA)-AgroSup Dijon - Institut National Supérieur des Sciences Agronomiques, de l'Alimentation et de l'Environnement, Centre Régional de Lutte contre le cancer Georges-François Leclerc [Dijon] (UNICANCER/CRLCC-CGFL), UNICANCER, This work is supported by grants of the Conseil Regional de Bourgogne, the INCa (Institut National du Cancer), Cancéropôle Grand-Est, ANR (Agence Nationale de la Recherche, ANR-06-JCJC-0103 and 07-PCV-0031), and the European Community (ApopTrain Marie Curie RTN) (O.M.). A.M., is supported by fellowships from the Ministry of Research and Education and the ARC (Association pour la Recherche sur le Cancer). S.S. is supported by the INCa., ANR-06-JCJC-0103,TRAILCHIM,TRAIL et chimiothérapies anticancéreuses(2006), Lipides - Nutrition - Cancer (U866) ( LNC ), Université de Bourgogne ( UB ) -Institut National de la Santé et de la Recherche Médicale ( INSERM ) -AgroSup Dijon - Institut National Supérieur des Sciences Agronomiques, de l'Alimentation et de l'Environnement-Ecole Nationale Supérieure de Biologie Appliquée à la Nutrition et à l'Alimentation de Dijon ( ENSBANA ), Centre Régional de Lutte contre le cancer - Centre Georges-François Leclerc ( CRLCC - CGFL ), and ANR-06-JCJC-0103,TRAILCHIM,TRAIL et chimiothérapies anticancéreuses ( 2006 )
- Subjects
MESH: Cell Death ,MESH: Signal Transduction ,Cancer Research ,Apoptosis ,TRAIL ,MESH : Models, Biological ,scaffold ,Cell membrane ,0302 clinical medicine ,Drug Discovery ,MESH: Animals ,Pharmacology (medical) ,Receptor ,death effector domain ,0303 health sciences ,Cell Death ,General Medicine ,TRAIL-R4 ,3. Good health ,Cell biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Signal transduction ,MESH : Apoptosis Regulatory Proteins ,Signal Transduction ,Programmed cell death ,c-FLIP ,death domain ,medicine.drug_class ,MESH : Cell Membrane ,Cancer therapy ,Biology ,Monoclonal antibody ,Models, Biological ,Article ,03 medical and health sciences ,medicine ,Animals ,Humans ,Chemotherapy ,[SDV.BBM]Life Sciences [q-bio]/Biochemistry, Molecular Biology ,MESH: Receptors, TNF-Related Apoptosis-Inducing Ligand ,MESH : Receptors, TNF-Related Apoptosis-Inducing Ligand ,[ SDV.BBM ] Life Sciences [q-bio]/Biochemistry, Molecular Biology ,030304 developmental biology ,MESH : Signal Transduction ,MESH: Humans ,MESH: Apoptosis Regulatory Proteins ,MESH: Apoptosis ,MESH : Humans ,Cell Membrane ,MESH: Models, Biological ,DISC ,Receptors, TNF-Related Apoptosis-Inducing Ligand ,MESH : Cell Death ,FADD ,Cancer cell ,MESH : Animals ,Apoptosis Regulatory Proteins ,MESH : Apoptosis ,MESH: Cell Membrane - Abstract
Article Open access plus; International audience; The use of TRAIL/APO2L and monoclonal antibodies targeting TRAIL receptors for cancer therapy holds great promise, due to their ability to restore cancer cell sensitivity to apoptosis in association with conventional chemotherapeutic drugs in a large variety of tumors. TRAIL-induced cell death is tightly regulated right from the membrane and at the DISC (Death-Inducing Signaling Complex) level. The following patent and literature review aims to present and highlight recent findings of the deadly discussion that determines tumor cell fate upon TRAIL engagement.
- Published
- 2011
25. DJ-1 inhibits TRAIL-induced apoptosis by blocking pro-caspase-8 recruitment to FADD
- Author
-
Guanghui Wang, Yubao Wang, Haigang Ren, Kai Fu, Hongfeng Wang, and Erkang Fei
- Subjects
Cancer Research ,Fas-Associated Death Domain Protein ,Protein Deglycase DJ-1 ,Apoptosis ,Caspase 8 ,TNF-Related Apoptosis-Inducing Ligand ,Genetics ,Humans ,RNA, Messenger ,FADD ,Molecular Biology ,Cells, Cultured ,Death domain ,Oncogene Proteins ,Oncogene ,biology ,Intracellular Signaling Peptides and Proteins ,Caspase Inhibitors ,Cell biology ,Protein Transport ,Receptors, TNF-Related Apoptosis-Inducing Ligand ,Death-inducing signaling complex ,biology.protein ,Death effector domain ,Tumor necrosis factor alpha ,Tumor Suppressor Protein p53 - Abstract
DJ-1 was initially identified as an oncogene product involved in human tumorigenesis in cooperation with Ras. Increased DJ-1 expression is associated with tumorigenesis in many cancers, whereas the loss of DJ-1 function is linked to an autosomal recessive form of Parkinson's disease (PD). It has been reported that DJ-1 protects cells from TRAIL (tumor necrosis factor-related apoptosis-inducing ligand)-induced apoptosis. However, the mechanism by which DJ-1 is involved is still largely unknown. Here we show that DJ-1 inhibits TRAIL-induced apoptosis by blocking Fas-associated protein death domain (FADD)-mediated pro-caspase-8 activation. Wild-type DJ-1, but not the PD-associated mutant L166P, binds to FADD to inhibit the formation of the death-inducing signaling complex (DISC). DJ-1 competes with pro-caspase-8 to bind to FADD at the death effector domain, thereby repressing the recruitment and activation of pro-caspase-8 to the active form of caspase-8. Thus, our study suggests that DJ-1 protects against TRAIL-induced apoptosis through the regulation of DISC formation.
- Published
- 2011
26. Detection of apoptosis by RT–PCR array in mefloquine–induced cochlear damage
- Author
-
Qi Wei–dong, Da Lian Ding, Yu Dong–zhen, Richard Salvi, Jiang Hai–yan, Someya Shinichi, and Masaru Tanokura
- Subjects
hair cells ,business.industry ,mefloquine ,cochlea ,apoptosis ,spiral ganglion neurons ,Anatomy ,Cell biology ,CFLAR ,ototoxicity ,medicine.anatomical_structure ,Otorhinolaryngology ,Apoptosis ,Gene expression ,gene expression ,otorhinolaryngologic diseases ,medicine ,Death effector domain ,sense organs ,Signal transduction ,BCL2-related protein A1 ,business ,Spiral ganglion ,Death domain - Abstract
Objective To investigate the occurrence and possible mechanisms of apoptosis in cochlear epithelium and spiral ganglion neurons after mefloquine treatment. Methods We used quantitative RT–PCR apoptosis–focused gene arrays (96–well, 84 apoptosis related genes) to assess changes of gene expression in the cochlear basilar membrane (hair cells–supporting cells) and spiral ganglion neurons of rat cochlear organotypic cultures treated with 100 μM mefloquine for 3 h. Results Significant up–or down–regulation in gene expression was detected in 23 genes in the cochlear basilar membrane, and in 32 genes in the spiral ganglion neurons compared with time–matched controls. The responding genes could be classified as pro–or anti–apoptotic, and were mainly implicated in the Bcl–2, Caspase, Card, IAP, TNF ligand/TNF receptor, Death domain/Death effector domain, DNA damage/p53, and NF–kappa B families. Synthetic analysis suggested that these families could be revised to two major pathways mainly involved in the death receptor–mediated signaling pathway and apoptotic mitochondrial pathway. In addition, it was found that numerous anti–apoptotic genes such as Bcl2a1, Birc1b, Birc3, Birc4, Bnip1, Cflar, Il10, Lhx4, Mcl1, Nfkb1, Prlr, Prok2, and TNF were greatly up–regulated in the cochlear tissue, which might imply the co–existence of protective response in the cells at the early stage of mefloquine–induced damage.
- Published
- 2011
27. The Fas–FADD death domain complex structure reveals the basis of DISC assembly and disease mutations
- Author
-
Se Bok Jang, Liwei Wang, Venkataraman Kabaleeswaran, Stefan Raunser, Jin Kuk Yang, Anthony C. Cruz, Ah Young Park, Hao Wu, Qian Yin, Thomas Walz, Amanda J. Rice, Ermelinda Damko, Carol V. Robinson, and Richard M. Siegel
- Subjects
Models, Molecular ,Spectrometry, Mass, Electrospray Ionization ,Fas-Associated Death Domain Protein ,Molecular Sequence Data ,Apoptosis ,urologic and male genital diseases ,Caspase 8 ,medicine.disease_cause ,Article ,Fas ligand ,Cell Line ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Tandem Mass Spectrometry ,Structural Biology ,medicine ,Animals ,Humans ,Amino Acid Sequence ,fas Receptor ,FADD ,Molecular Biology ,030304 developmental biology ,Death domain ,0303 health sciences ,Mutation ,biology ,Autoimmune Lymphoproliferative Syndrome ,Fas receptor ,Molecular biology ,Protein Structure, Tertiary ,030220 oncology & carcinogenesis ,Death-inducing signaling complex ,biology.protein ,Death effector domain ,biological phenomena, cell phenomena, and immunity ,Sequence Alignment - Abstract
The death-inducing signaling complex (DISC) formed by the death receptor Fas, the adaptor protein FADD and caspase-8 mediates the extrinsic apoptotic program. Mutations in Fas that disrupt the DISC cause autoimmune lymphoproliferative syndrome (ALPS). Here we show that the Fas-FADD death domain (DD) complex forms an asymmetric oligomeric structure composed of 5-7 Fas DD and 5 FADD DD, whose interfaces harbor ALPS-associated mutations. Structure-based mutations disrupt the Fas-FADD interaction in vitro and in living cells; the severity of a mutation correlates with the number of occurrences of a particular interaction in the structure. The highly oligomeric structure explains the requirement for hexameric or membrane-bound FasL in Fas signaling. It also predicts strong dominant negative effects from Fas mutations, which are confirmed by signaling assays. The structure optimally positions the FADD death effector domain (DED) to interact with the caspase-8 DED for caspase recruitment and higher-order aggregation.
- Published
- 2010
28. NMR backbone dynamics studies of human PED/PEA-15 outline protein functional sites
- Author
-
Biancamaria Farina, Luciano Pirone, Carlo Pedone, Francesca Viparelli, Luigi Russo, Roberto Fattorusso, Emilia Pedone, and Nunzianna Doti
- Subjects
Regulator ,food and beverages ,Cell Biology ,Nuclear magnetic resonance spectroscopy ,Plasma protein binding ,Biology ,Biochemistry ,Protein–protein interaction ,Molecular dynamics ,Phosphoprotein ,Biophysics ,Death effector domain ,Molecular Biology ,Death domain - Abstract
PED/PEA-15 (phosphoprotein enriched in diabetes/phosphoprotein enriched in astrocytes) is a ubiquitously expressed protein and a key regulator of cell growth and glucose metabolism. PED/PEA-15 mediates both homotypic and heterotypic interactions and is constituted by an N-terminal canonical death effector domain and a C-terminal tail. In the present study, the backbone dynamics of PED/PEA-15 via (15)N R(1) and R(2) and steady-state [(1)H]-(15)N NOE measurements is reported. The dynamic parameters were analyzed using both Lipari-Szabo model-free formalism and a reduced spectral density mapping approach. The results obtained define a polar and charged surface of the death effector domain characterized by internal motions in the micro- to millisecond timescale, which is crucial for the multiple heterotypic functional protein-protein interactions in which PED/PEA-15 is involved. The present study contributes to a better understanding of the molecular basis of the PED/PEA-15 functional interactions and provides a more detailed surface for the design and development of PED/PEA-15 binders.
- Published
- 2010
29. Gene expression in cisplatin ototoxicity and protection with p53 inhibitor
- Author
-
Richard Salvi, Da Lian Ding, Jiang Hai-yan, Wang Ping, and Donald E. Coling
- Subjects
p53 ,hair cells ,cochlea ,Biology ,Ototoxicity ,Gene expression ,otorhinolaryngologic diseases ,medicine ,Spiral ganglion ,Death domain ,Cisplatin ,Genetics ,spiral ganglion neurons ,medicine.disease ,ototoxicity ,medicine.anatomical_structure ,Otorhinolaryngology ,Apoptosis ,gene expression ,Cancer research ,Death effector domain ,sense organs ,Signal transduction ,Pifithrin–a ,medicine.drug - Abstract
Abxtract Cisplatin damages cochlear hair cells and spiral ganglion neurons through cell death signaling pathways that are not fully understood. We used focused apoptosis gene microarrays to study early changes in gene expression in cochlear cultures from P3 neonatal rats treated with cisplatin (0.2 mM). After 12 hours of cisplatin treatment, more than 50% of the 96 genes on the array showed a significant decrease in expression, consistent with widespread cell death. However, after 3 hours of cisplatin treatment, 10 genes showed significant increase in expression in total cochlear tissue. In experiments with subsets of cochlear tissues, at 3h, cisplatin induced increased expression of 12 genes in the cochlear sensory epithelium (basilar membrane) and 11 genes in the spiral ganglion (tissue of Rosenthal’s canal, containing the spiral ganglion). These included pro– and anti–apoptotic genes involved in the p53 signaling pathway, TNF receptor family, NF–kappaB pathway, death domain family, death effector domain family, Bcl–2 family, CARD family, TRAF family, and GTP signal transduction. Although the changes in gene expression showed an overlap between basilar membrane and spiral ganglion, other changes, which may reflect the unique response of each tissue, were also observed. Pifithrin–a blocked cisplatin–induced up–regulation of genes in the p53 signaling pathway when assayed by both superarray and real time PCR. The data add to our understanding of the involvement of p53 in cisplatin–induced ototoxicity and otoprotection, conferred by the p53 inhibitor Pifithrin–a.
- Published
- 2009
30. Paclitaxel promotes a caspase 8-mediated apoptosis through death effector domain association with microtubules
- Author
-
Ainhoa Mielgo, Vicente A. Torres, Dwayne G. Stupack, Simone Barbero, and Kiran Clair
- Subjects
Cancer Research ,Paclitaxel ,microtubule organizing center ,caspase ,Apoptosis ,Caspase 8 ,Microtubules ,cell survival ,Article ,Neuroblastoma ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Cell Line, Tumor ,Genetics ,Humans ,death effector domain ,Molecular Biology ,Caspase ,030304 developmental biology ,Death domain ,Centrosome ,0303 health sciences ,biology ,NLRP1 ,Antineoplastic Agents, Phytogenic ,Protein Structure, Tertiary ,3. Good health ,Cell biology ,chemistry ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,Caspase 10 ,Death effector domain - Abstract
Microtubule-perturbing drugs have become front-line chemotherapeutics, inducing cell-cycle crisis as a major mechanism of action. However, these agents show pleiotropic effects on cells and can induce apoptosis through other means. Paclitaxel, a microtubule-stabilizing agent, induces a caspase-dependent apoptosis, although the precise mechanism(s) remain unclear. Here, we used genetic approaches to evaluate the role of caspase 8 in paclitaxel-mediated apoptosis. We observed that caspase 8-expressing cells are more sensitive to paclitaxel than caspase 8-deficient cells. Mechanistically, caspase 8 was found associated with microtubules, and this interaction increased after paclitaxel treatment. The prodomains death effector domains (DEDs) of caspase 8 were sufficient for interaction with microtubules, but the caspase 8 holoprotein was required for apoptosis. DED-only forms of caspase 8 were found in both primary and tumor cell lines, associating with perinuclear microtubules and the centrosome. Microtubule association, and paclitaxel sensitivity, depends on a critical lysine (K156) within a microtubule-binding motif (KLD) in DED-b of caspase 8. The results show an unexpected pathway of apoptosis mediated by caspase 8.
- Published
- 2009
31. A New C-Terminal Cleavage Product of Procaspase-8, p30, Defines an Alternative Pathway of Procaspase-8 Activation
- Author
-
Alexander Pappa, Julia C. Hoffmann, Inna N. Lavrik, and Peter H. Krammer
- Subjects
Receptor complex ,CASP8 and FADD-Like Apoptosis Regulating Protein ,Apoptosis ,Biology ,Cleavage (embryo) ,Jurkat Cells ,Humans ,fas Receptor ,FADD ,Molecular Biology ,Death domain ,Caspase 8 ,Antibodies, Monoclonal ,Receptors, Death Domain ,Articles ,Cell Biology ,Fas receptor ,Heterotetramer ,Molecular biology ,Peptide Fragments ,Protein Structure, Tertiary ,Cell biology ,Enzyme Activation ,Caspases ,Death-inducing signaling complex ,biology.protein ,Death effector domain ,Signal Transduction - Abstract
Apoptosis can be triggered by a number of factors, including UV or γ-irradiation, chemotherapeutic drugs, and signaling from death receptors (11, 12). CD95 (APO-1/Fas) is a member of the death receptor family, a subfamily of the tumor necrosis factor receptor (TNF-R) superfamily (1, 30). Eight members of the death receptor subfamily have been characterized so far: TNF-R1 (DR1, CD120a, p55, p60), CD95 (DR2, APO-1, Fas), DR3 (APO-3, LARD, TRAMP, WSL1), TRAIL-R1 (APO-2, DR4), TRAIL-R2 (DR5, KILLER, TRICK2), DR6, EDA-R, and NGF-R (13). Cross-linking of CD95 by its natural ligand, CD95L (CD178) (29), or by agonistic antibodies induces apoptosis in sensitive cells (31, 36). The death-inducing signaling complex (DISC) is formed within seconds after CD95 stimulation (9). The DISC consists of oligomerized, probably trimerized CD95 receptors, the adaptor molecule FADD, two isoforms of procaspase-8 (procaspase-8a and -8b), procaspase-10, and c-FLIPL/S/R (6, 19, 21, 25, 27). The interactions between molecules at the DISC are based on homotypic contacts. The death domain of the receptor interacts with the death domain of FADD, while the death effector domain (DED) of FADD interacts with the N-terminal tandem DEDs of procaspase-8 and -10 and c-FLIPL/S/R. Two isoforms of procaspase-8 (procaspase-8a and procaspase-8b) were reported to be bound to the DISC (24). Both isoforms possess two tandem DEDs, as well as the catalytic subunits p18 and p10 (see Fig. Fig.1A).1A). Procaspase-8a contains an additional 2-kDa (15-amino-acid [aa]) fragment, which results from the translation of exon 9. This small fragment is located between the second DED and the large catalytic subunit, resulting in different lengths of procaspase-8a and -8b (p55 and p53 kDa), respectively. FIG. 1. A new 30-kDa protein is detected by the anti-caspase-8 MAb C15. (A) Scheme of procaspase-8 and its cleavage products. The binding sites of the anti-caspase-8 MAbs C5 and C15 are indicated. (B) The B-lymphoblastoid cell lines SKW6.4, Raji, and BJAB and ... Activation of procaspase-8 is believed to follow an “induced-proximity” model in which high local concentrations and a favorable mutual orientation of procaspase-8 molecules at the DISC lead to their autoproteolytic processing (2, 3, 20). There is strong evidence from several in vitro studies that autoproteolytic activation of procaspase-8 occurs after oligomerization at the receptor complex (20). Furthermore, it has been shown that homodimers of procaspase-8 have proteolytic activity and that proteolytic processing of procaspase-8 occurs between precursor homodimers (3). Procaspase-8a/b (p55/p53) processing at the DISC has been described to involve two sequential cleavage steps (see Fig. Fig.1A).1A). This process is referred to as the “two-step model” (3, 17). The first cleavage step occurs between the two protease domains, and the second cleavage step takes place between the prodomain and the large protease subunit (see Fig. Fig.1A)1A) (15). During the first cleavage step, the cleavage at Asp374 generates the two subunits p43/p41 and p12. Both cleavage products remain bound to the DISC: p43/p41 by DED interactions and p12 by interactions with the large protease domain of p43/p41. The second cleavage step takes place at Asp216 and Asp384, producing the active enzyme subunits p18, p10, and the prodomain p26/p24. As a result of procaspase-8 processing, the active caspase-8 heterotetramer p182-p102 is formed at the DISC. This heterotetramer is subsequently released into the cytosol, starting the apoptotic signaling cascade (14). Recent studies have shown that processing of procaspase-8 at the DISC is more complicated and can involve additional steps like the generation of a prolonged prodomain of procaspase-8, termed CAP3 (p27), that is quickly converted to p26 (see Fig. Fig.1A)1A) (7). In addition to its central role in death receptor-induced apoptosis, caspase-8 was reported to be required for proliferation of lymphocytes (12, 23). Recently caspase-8 was shown to be an important factor for NF-κB activation following T-cell receptor stimulation (28). The mechanism underlying the dual role of caspase-8 activity and its regulation is largely unknown. In the present study, we show that upon death receptor stimulation, p30 is formed by cleavage at Asp210, a yet-unknown cleavage product of procaspase-8, which comprises the C terminus of procaspase-8. p30 turned out to be a key intermediate product in the course of procaspase-8 processing. Furthermore, we suggest that the p30-mediated activation of procaspase-8 plays an important role in the amplification of the death signal. Taken together, our findings provide a new mechanism of procaspase-8 activation and extend the current two-step cleavage model by an alternative activation pathway.
- Published
- 2009
32. NMR studies reveal a novel mode for hFADD to bind with the unstructured hRTN3 which initiates the ER-stress activated apoptosis
- Author
-
Wanlong Zhu, Jingxian Liu, Jianxing Song, and Haina Qin
- Subjects
Fas-Associated Death Domain Protein ,Molecular Sequence Data ,Biophysics ,Apoptosis ,Nerve Tissue Proteins ,Plasma protein binding ,Endoplasmic Reticulum ,Biochemistry ,Alzheimer Disease ,Humans ,Amino Acid Sequence ,FADD ,Nuclear Magnetic Resonance, Biomolecular ,Molecular Biology ,Peptide sequence ,Death domain ,biology ,Chemistry ,Membrane Proteins ,Cell Biology ,Nuclear magnetic resonance spectroscopy ,Membrane protein ,Mutation ,biology.protein ,Unfolded protein response ,Death effector domain ,Carrier Proteins ,Protein Binding - Abstract
RTN3 can recruit Fas-associated death domain (FADD), thus initiating the ER-stress activated apoptosis. It also interacts with the beta-secretase and its aggregation is critically associated with Alzheimer's disease. Here, we first investigated the solution conformation of hRTN3, subsequently characterized its binding with hFADD. The results reveal: (1) both hRTN3 N- and C-termini are intrinsically unstructured. Nevertheless, the C-terminus contains two short helix-populated regions. (2) The unstructured hRTN3 C-terminus can bind to hFADD as shown by ITC. Further NMR investigation successfully identified the binding involved hRTN3 residues. (3) Although upon hRTN3-binding, the perturbed hFADD residues were distributed over the whole sequence, the majority of the significantly perturbed are over its death effector domain, very different from the previously observed binding mode for FADD. This study also implies a possible linkage between Alzheimer's disease and ER-stress activated apoptosis.
- Published
- 2009
33. Death Effector Domain-Containing Proteins
- Author
-
Joe W. Ramos and M. Gudur Valmiki
- Subjects
Models, Molecular ,Death Domain Receptor Signaling Adaptor Proteins ,Programmed cell death ,GTPase-activating protein ,Protein Conformation ,DEDD ,Apoptosis ,Biology ,Cellular and Molecular Neuroscience ,Morphogenesis ,Animals ,Humans ,Protein Isoforms ,FADD ,Molecular Biology ,Cell Proliferation ,Death domain ,Pharmacology ,Effector ,Cell Biology ,Cell biology ,biology.protein ,Molecular Medicine ,Death effector domain ,Signal transduction ,Signal Transduction - Abstract
Death effector domains (DEDs) are protein-protein interaction structures that are found in proteins that regulate a variety of signal transduction pathways. DEDs are a part of the larger family of Death Domain structures that have been primarily described in the control of programmed cell death. The seven standard DED-containing proteins are fas associated death domain protein (FADD), Caspase-8 and 10, cellular FLICE-like inhibitory protein (c-FLIP), death effector domain containing DNA binding (DEDD), DEDD2 and phosphoprotein enriched in astrocytes 15-Kda (PEA-15). These proteins are particularly associated with the regulation of apoptosis and proliferation mediated by the tumor necrosis factor alpha (TNFalpha) receptor family. Consequently DED-containing proteins are reported to regulate transcription, migration, and proliferation, in addition to both pro and anti-apoptotic functions. Moreover, DED proteins are essential in embryonic development and homeostasis of the immune system. Here we focus on the role of DED-containing proteins in development and the pathologies arising from abnormal expression of these proteins.
- Published
- 2008
34. MUC1 Oncoprotein Blocks Death Receptor–Mediated Apoptosis by Inhibiting Recruitment of Caspase-8
- Author
-
Donald Kufe, Takeshi Kawano, Naoki Agata, Deepak Raina, Surender Kharbanda, and Rehan Ahmad
- Subjects
Cancer Research ,Apoptosis ,Caspase 8 ,digestive system ,Article ,Fas ligand ,Cell Line, Tumor ,Humans ,FADD ,RNA, Small Interfering ,skin and connective tissue diseases ,neoplasms ,Death domain ,Base Sequence ,biology ,Mucin-1 ,Receptors, Death Domain ,Fas receptor ,biological factors ,digestive system diseases ,Cell biology ,Enzyme Activation ,Oncology ,Death-inducing signaling complex ,biology.protein ,Cancer research ,Death effector domain ,Signal transduction ,Signal Transduction - Abstract
Stimulation of the death receptor superfamily induces the activation of caspase-8 and thereby the apoptotic response. The MUC1 oncoprotein is aberrantly overexpressed by diverse human malignancies and inhibits stress-induced apoptosis. The present results show that MUC1 blocks activation of caspase-8 and apoptosis in the response of malignant cells to tumor necrosis factor α, tumor necrosis factor–related apoptosis-inducing ligand, and Fas ligand. The results show that MUC1 associates constitutively with caspase-8. The MUC1 cytoplasmic domain (MUC1-CD) binds directly to the caspase-8 p18 fragment upstream to the catalytic Cys360 site. The results also show that MUC1-CD binds to Fas-associated death domain (FADD) at the death effector domain. In nonmalignant epithelial cells, MUC1 interacts with caspase-8 and FADD as an induced response to death receptor stimulation. The functional significance of these interactions is supported by the demonstration that MUC1 competes with caspase-8 for binding to FADD and blocks recruitment of caspase-8 to the death-inducing signaling complex. These findings indicate that MUC1 is of importance to the physiologic regulation of caspase-8 activity and that overexpression of MUC1, as found in human malignancies, could contribute to constitutive inhibition of death receptor signaling pathways. [Cancer Res 2008;68(15):6136–44]
- Published
- 2008
35. Calmodulin binding to cellular FLICE-like inhibitory protein modulates Fas-induced signalling
- Author
-
William J. Cook, Jay M. McDonald, Pritish Pawar, John C. Kappes, Keith J. Micoli, Yabing Chen, and Haitao Ding
- Subjects
MAPK/ERK pathway ,Caspase 8 ,Binding Sites ,Calmodulin ,biology ,Kinase ,CASP8 and FADD-Like Apoptosis Regulating Protein ,Apoptosis ,Cell Biology ,Biochemistry ,Cell biology ,Cell Line, Tumor ,biology.protein ,Humans ,Calcium ,Death effector domain ,fas Receptor ,FADD ,Molecular Biology ,Signal Transduction ,Death domain - Abstract
We and others have demonstrated that Fas-mediated apoptosis is a potential therapeutic target for cholangiocarcinoma. Previously, we reported that CaM (calmodulin) antagonists induced apoptosis in cholangiocarcinoma cells through Fas-related mechanisms. Further, we identified a direct interaction between CaM and Fas with recruitment of CaM into the Fas-mediated DISC (death-inducing signalling complex), suggesting a novel role for CaM in Fas signalling. Therefore we characterized the interaction of CaM with proteins recruited into the Fas-mediated DISC, including FADD (Fas-associated death domain)-containing protein, caspase 8 and c-FLIP {cellular FLICE [FADD (Fas-associated death domain)-like interleukin 1β-converting enzyme]-like inhibitory protein}. A Ca2+-dependent direct interaction between CaM and FLIPL, but not FADD or caspase 8, was demonstrated. Furthermore, a 37.3±5.7% increase ( n =6, P =0.001) in CaM–FLIP binding was observed at 30 min after Fas stimulation, which returned to the baseline after 60 min and correlated with a Fas-induced increase in intracellular Ca2+ that reached a peak at 30 min and decreased gradually over 60 min in cholangiocarcinoma cells. A CaM antagonist, TFP (trifluoperazine), inhibited the Fas-induced increase in CaM–FLIP binding concurrent with inhibition of ERK (extracellular-signal-regulated kinase) phosphorylation, a downstream signal of FLIP. Direct binding between CaM and FLIPL was demonstrated using recombinant proteins, and a CaM-binding region was identified in amino acids 197–213 of FLIPL. Compared with overexpression of wild-type FLIPL that resulted in decreased spontaneous as well as Fas-induced apoptosis, mutant FLIPL with deletion of the CaM-binding region resulted in increased spontaneous and Fas-induced apoptosis in cholangiocarcinoma cells. Understanding the biology of CaM–FLIP binding may provide new therapeutic targets for cholangiocarcinoma and possibly other cancers. Abbreviations: AM, acetoxymethyl ester; CaM, calmodulin; CaMS, CaM–Sepharose; c-FLIP, cellular FLICE-like inhibitory protein; Co-IP, co-immunoprecipitation; CS, control Sepharose; DD, death domain; DED, death effector domain; DISC, death-inducing signalling complex; ERK, extracellular-signal-regulated kinase; FADD, Fas-associated DD; FLICE, FADD-like interleukin 1β-converting enzyme; GST, glutathione transferase; HBSS, Hanks balanced salt solution; HRP, horseradish peroxidase; IPTG, isopropyl β-D-thiogalactoside; TFP, trifluoperazine; TNF, tumour necrosis factor; TNF-R1, TNF receptor 1; TRAIL, TNF-related apoptosis-inducing ligand; TRAIL-R1, TRAIL receptor 1; WT, wild-type
- Published
- 2008
36. The PYRIN Domain in Signal Transduction
- Author
-
Christian Stehlik
- Subjects
Models, Molecular ,Protein Conformation ,Molecular Sequence Data ,Apoptosis ,Biology ,Biochemistry ,Pyrin domain ,Article ,Evolution, Molecular ,HAMP domain ,Protein structure ,Animals ,Humans ,Amino Acid Sequence ,Molecular Biology ,Conserved Sequence ,Death domain ,Inflammation ,Genetics ,Binding Sites ,Sequence Homology, Amino Acid ,Immunity ,Chromosome Mapping ,DHR1 domain ,Receptors, Death Domain ,Cell Biology ,General Medicine ,Cell biology ,NACHT domain ,CARD domain ,Death effector domain ,Sequence Alignment ,Signal Transduction - Abstract
The Death Domain Fold superfamily of evolutionarily conserved protein-protein interaction domains consists of 4 subfamilies: the death domain, the death effector domain, the caspase recruitment domain, and the PYRIN domain. Interaction of Death Domain Fold containing proteins modulates the activity of several downstream effectors, such as caspases and transcription factors. Recent studies provide evidence for not only homotypic-, but also heterotypic interactions among different sub-families, and even unconventional non-death domain fold interactions. As the number of potential protein associations among Death Domain Fold containing proteins expands and their influence on cellular responses increases, a challenging field for new investigations opens up. This review will focus on PYRIN domain-containing proteins and discuss the recent advances that provide strong evidence that PYRIN domain-mediated signal transduction has broad implications on cellular functions, including innate immunity, inflammation, differentiation, apoptosis, and cancer.
- Published
- 2007
37. The Inflammasome Adaptor ASC Induces Procaspase-8 Death Effector Domain Filaments*
- Author
-
Darren L. Brown, David P. Sester, Simon O. Cridland, Parimala R. Vajjhala, Siew Wai Pang, Kate Schroder, Jennifer L. Stow, Alvin Lu, Hao Wu, Katryn J. Stacey, Justine M. Hill, and Vitaliya Sagulenko
- Subjects
Programmed cell death ,Death fold ,Inflammasomes ,Apoptosis ,Biochemistry ,Pyrin domain ,Catalytic Domain ,medicine ,Humans ,Molecular Biology ,Caspase ,Death domain ,Inflammation ,Caspase 8 ,biology ,Cell Death ,Caspase 1 ,Signal transducing adaptor protein ,Inflammasome ,hemic and immune systems ,Cell Biology ,eye diseases ,Cell biology ,CARD Signaling Adaptor Proteins ,Cytoskeletal Proteins ,HEK293 Cells ,Microscopy, Fluorescence ,Immunology ,Mutation ,biology.protein ,Death effector domain ,medicine.drug ,Signal Transduction ,Protein Binding - Abstract
Inflammasomes mediate inflammatory and cell death responses to pathogens and cellular stress signals via activation of procaspases-1 and -8. During inflammasome assembly, activated receptors of the NLR or PYHIN family recruit the adaptor protein ASC and initiate polymerization of its pyrin domain (PYD) into filaments. We show that ASC filaments in turn nucleate procaspase-8 death effector domain (DED) filaments in vitro and in vivo. Interaction between ASC PYD and procaspase-8 tandem DEDs optimally required both DEDs and represents an unusual heterotypic interaction between domains of the death fold superfamily. Analysis of ASC PYD mutants showed that interaction surfaces that mediate procaspase-8 interaction overlap with those required for ASC self-association and interaction with the PYDs of inflammasome initiators. Our data indicate that multiple types of death fold domain filaments form at inflammasomes and that PYD/DED and homotypic PYD interaction modes are similar. Interestingly, we observed condensation of procaspase-8 filaments containing the catalytic domain, suggesting that procaspase-8 interactions within and/or between filaments may be involved in caspase-8 activation. Procaspase-8 filaments may also be relevant to apoptosis induced by death receptors.
- Published
- 2015
38. Fuzzy support vector machine model to predict human death domain protein–protein interactions
- Author
-
Kamal Raj Pardasani and Prakash Arun Nemade
- Subjects
Support vector machine ,biology ,Polynomial kernel ,Urology ,biology.protein ,DEDD ,Sequential minimal optimization ,Death effector domain ,FADD ,Computational biology ,Bioinformatics ,Death domain ,Protein–protein interaction - Abstract
Proteins have crucial importance in every living system, and Protein–Protein Interactions (PPIs) play a pivotal role in regulation of virtually all biological processes such as DNA transcription, replication, metabolic cycles and signaling cascades. The PPIs play an important role in the complex process of cell death which mainly occurs via apoptosis and necrosis in eukaryotic cells. Apoptosis is an orderly cellular suicide program critical for the development and homeostasis of multicellular organism. Failure to control apoptosis can have catastrophic consequences. The cascades of amazing reactions carried out by proteins such as Caspase, CARD, NLRP, NOD, FADD, DEDD, POP, Myd88 etc. play important role in the process of cell death. The high throughput experimental methods for determining PPIs are time consuming, expensive and are generating huge amount of PPIs data. Therefore, there is need to develop computational methods to efficiently and accurately predict PPIs. In this work, an attempt has been made to develop a fuzzy support vector machine (F-SVM) model for predicting death domain (DD) PPIs based on sixteen physicochemical, biochemical and structural features of amino acids which are monomers of proteins. First, the protein primary sequences are encoded into sequential features represented by descriptors. Then, the Support Vector Machine and Sequential Minimal Optimization of WEKA software are employed to classify interacting and non-interacting protein pairs. The performance of SVM and F-SVM with various kernel functions has been evaluated and it was observed that libSVM with Polynomial kernel was found to be best with accuracy of 77.94 % via F-SVM which is optimum model in predicting human DD-PPIs. Validation is performed by tenfold cross-validation technique. The F-SVM performance measure is 2.94 % higher than SVM in terms of accuracy with the use of custom designed fuzzy membership function. The results obtained are in agreement with available experimental data. Such models can be useful in providing PPI information of DD proteins which can be useful in understanding the molecular mechanisms involved in death of cells taking place due to aging, programmed cell death and various diseases. It may through some light on the study of cancerous cell and gerontology.
- Published
- 2015
39. Dominant negative FADD dissipates the proapoptotic signalosome of the unfolded protein response in diabetic embryopathy
- Author
-
Fang Wang, Michael J. Quon, Anne-Odile Hueber, Jingwen Yu, Jian-Ying Wang, Hongbo Weng, Peixin Yang, HUEBER, Anne-Odile, University of Maryland School of Medicine, University of Maryland System, Institut de Biologie Valrose (IBV), Université Nice Sophia Antipolis (1965 - 2019) (UNS), COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université Côte d'Azur (UCA), and Université Nice Sophia Antipolis (... - 2019) (UNS)
- Subjects
Physiology ,Fas-Associated Death Domain Protein ,Neurogenesis ,Recombinant Fusion Proteins ,Endocrinology, Diabetes and Metabolism ,[SDV]Life Sciences [q-bio] ,Pregnancy in Diabetics ,Down-Regulation ,Apoptosis ,Mice, Transgenic ,Caspase 3 ,Protein Serine-Threonine Kinases ,Caspase 8 ,urologic and male genital diseases ,Cell Line ,Diabetes Mellitus, Experimental ,Multienzyme Complexes ,Pregnancy ,Physiology (medical) ,Endoribonucleases ,Animals ,Humans ,Neural Tube Defects ,FADD ,[SDV.BDD]Life Sciences [q-bio]/Development Biology ,Caspase ,Death domain ,biology ,Articles ,Embryo, Mammalian ,Endoplasmic Reticulum Stress ,TRADD ,TNF Receptor-Associated Death Domain Protein ,Mice, Inbred C57BL ,[SDV] Life Sciences [q-bio] ,Unfolded Protein Response ,biology.protein ,Cancer research ,Unfolded protein response ,Female ,Mutant Proteins ,Death effector domain ,biological phenomena, cell phenomena, and immunity ,Biomarkers - Abstract
International audience; Endoplasmic reticulum (ER) stress and caspase 8-dependent apoptosis are two interlinked causal events in maternal diabetes-induced neural tube defects (NTDs). The inositol-requiring enzyme 1α (IRE1α) signalosome mediates the proapoptotic effect of ER stress. Diabetes increases tumor necrosis factor receptor type 1R-associated death domain (TRADD) expression. Here, we revealed two new unfolded protein response (UPR) regulators, TRADD and Fas-associated protein with death domain (FADD). TRADD interacted with both the IRE1α-TRAF2-ASK1 complex and FADD. In vivo overexpression of a FADD dominant negative (FADD-DN) mutant lacking the death effector domain disrupted diabetes-induced IRE1α signalosome and suppressed ER stress and caspase 8-dependent apoptosis, leading to NTD prevention. FADD-DN abrogated ER stress markers and blocked the JNK1/2-ASK1 pathway. Diabetes-induced mitochondrial translocation of proapoptotic Bcl-2 members mitochondrial dysfunction and caspase cleavage were also alleviated by FADD-DN. In vitro TRADD overexpression triggered UPR and ER stress before manifestation of caspase 3 and caspase 8 cleavage and apoptosis. FADD-DN overexpression repressed high glucose- or TRADD overexpression-induced IRE1α phosphorylation, its downstream proapoptotic kinase activation and endonuclease activities, and apoptosis. FADD-DN also attenuated tunicamycin-induced UPR and ER stress. These findings suggest that TRADD participates in the IRE1α signalosome and induces UPR and ER stress and that the association between TRADD and FADD is essential for diabetes- or high glucose-induced UPR and ER stress.
- Published
- 2015
40. Structure and Dynamics of ASC2, a Pyrin Domain-only Protein That Regulates Inflammatory Signaling
- Author
-
Justine M. Hill, Ranajeet Ghose, and Aswin Natarajan
- Subjects
Models, Molecular ,Magnetic Resonance Spectroscopy ,Stereochemistry ,Protein Conformation ,Molecular Sequence Data ,Apoptosis ,Biology ,Pyrin domain ,Biochemistry ,Protein structure ,Humans ,Amino Acid Sequence ,Peptide sequence ,Molecular Biology ,Death domain ,Ribonucleoprotein ,Inflammation ,Models, Statistical ,Cell Biology ,Pyrin ,Protein Structure, Tertiary ,Cytoskeletal Proteins ,Ribonucleoproteins ,Helix ,Biophysics ,Death effector domain ,Signal transduction ,Apoptosis Regulatory Proteins ,Signal Transduction - Abstract
Pyrin domain (PYD)-containing proteins are key components of pathways that regulate inflammation, apoptosis, and cytokine processing. Their importance is further evidenced by the consequences of mutations in these proteins that give rise to autoimmune and hyperinflammatory syndromes. PYDs, like other members of the death domain (DD) superfamily, are postulated to mediate homotypic interactions that assemble and regulate the activity of signaling complexes. However, PYDs are presently the least well characterized of all four DD subfamilies. Here we report the three-dimensional structure and dynamic properties of ASC2, a PYD-only protein that functions as a modulator of multidomain PYD-containing proteins involved in NF-kappaB and caspase-1 activation. ASC2 adopts a six-helix bundle structure with a prominent loop, comprising 13 amino acid residues, between helices two and three. This loop represents a divergent feature of PYDs from other domains with the DD fold. Detailed analysis of backbone 15N NMR relaxation data using both the Lipari-Szabo model-free and reduced spectral density function formalisms revealed no evidence of contiguous stretches of polypeptide chain with dramatically increased internal motion, except at the extreme N and C termini. Some mobility in the fast, picosecond to nanosecond timescale, was seen in helix 3 and the preceding alpha2-alpha3 loop, in stark contrast to the complete disorder seen in the corresponding region of the NALP1 PYD. Our results suggest that extensive conformational flexibility in helix 3 and the alpha2-alpha3 loop is not a general feature of pyrin domains. Further, a transition from complete disorder to order of the alpha2-alpha3 loop upon binding, as suggested for NALP1, is unlikely to be a common attribute of pyrin domain interactions.
- Published
- 2006
41. The Structure of FADD and Its Mode of Interaction with Procaspase-8
- Author
-
Gaku Morisawa, Yu Wei, Paul E. Carrington, Yufeng Wei, Evridipis Gavathiotis, Ted Huang, Milton H. Werner, Justine M. Hill, and Cristinel Sandu
- Subjects
Models, Molecular ,Intracellular domain ,Fas-Associated Death Domain Protein ,Amino Acid Motifs ,Molecular Sequence Data ,Biology ,Transfection ,urologic and male genital diseases ,Jurkat Cells ,Humans ,Amino Acid Sequence ,fas Receptor ,FADD ,Molecular Biology ,Adaptor Proteins, Signal Transducing ,Death domain ,Caspase 8 ,Binding Sites ,Mutagenesis ,Cell Biology ,Fas receptor ,Protein Structure, Tertiary ,Cell biology ,Caspases ,Death-inducing signaling complex ,biology.protein ,Death effector domain ,biological phenomena, cell phenomena, and immunity ,Signal Transduction - Abstract
The structure of FADD has been solved in solution, revealing that the death effector domain (DED) and death domain (DD) are aligned with one another in an orthogonal, tail-to-tail fashion. Mutagenesis of FADD and functional reconstitution with its binding partners define the interaction with the intracellular domain of CD95 and the prodomain of procaspase-8 and reveal a self-association surface necessary to form a productive complex with an activated "death receptor." The identification of a procaspase-specific binding surface on the FADD DED suggests a preferential interaction with one, but not both, of the DEDs of procaspase-8 in a perpendicular arrangement. FADD self-association is mediated by a "hydrophobic patch" in the vicinity of F25 in the DED. The structure of FADD and its functional characterization, therefore, illustrate the architecture of key components in the death-inducing signaling complex.
- Published
- 2006
42. Lethal activity of FADD death domain in renal tubular epithelial cells
- Author
-
Jesús Egido, Corina Lorz, Pilar Justo, Ana Belen Sanz, and Alberto Ortiz
- Subjects
kidney ,Programmed cell death ,Serine Proteinase Inhibitors ,Cell Survival ,death domain ,Fas-Associated Death Domain Protein ,Mice, Inbred Strains ,urologic and male genital diseases ,Receptors, Tumor Necrosis Factor ,Mice ,medicine ,Animals ,FADD ,Cells, Cultured ,Caspase ,Adaptor Proteins, Signal Transducing ,Death domain ,Kidney ,biology ,urogenital system ,NF-kappa B ,apoptosis ,Epithelial Cells ,Acute Kidney Injury ,Caspase Inhibitors ,Protein Structure, Tertiary ,Cell biology ,tubular cells ,Kidney Tubules ,medicine.anatomical_structure ,Gene Expression Regulation ,Nephrology ,Apoptosis ,Caspases ,Death-inducing signaling complex ,biology.protein ,Death effector domain ,biological phenomena, cell phenomena, and immunity ,NFκB - Abstract
Fas-associated death domain (FADD) is an adaptor protein that is required for the transmission of the death signal from lethal receptors of the tumor necrosis factor superfamily. FADD contains a death domain (DD) and a death effector domain (DED). As death receptors contribute to renal tubular injury and tubular cell FADD increases in acute renal failure, we have studied the function of FADD in tubular epithelium. FADD expression was studied in kidney samples from mice. In order to study the contribution of FADD to renal tubular cell survival, FADD or FADD-DD were overexpressed in murine tubular epithelium. FADD is expressed in renal tubules of the healthy kidney. Both FADD and FADD-DD induce apoptosis in primary cultures of murine tubular epithelium and in the murine cortical tubular cell line. Death induced by FADD-DD has apoptotic morphology, but differs from death receptor-induced apoptosis in that it is not blocked by inhibitors of caspases. Neither an inhibitor of serine proteases nor overexpression of antiapoptotic BclxL prevented cell death. However, the combination of caspase and serine protease inhibition was protective. FADD and FADD-DD overexpression decreased nuclear factor kappa B activity. These data suggest that FADD has a death regulatory function in renal tubular cells that is independent of death receptors. FADD-DD is sufficient to induce apoptosis in these cells. This information is relevant to understanding the role of FADD in tubular injury.
- Published
- 2006
43. Crystal Structure of MC159 Reveals Molecular Mechanism of DISC Assembly and FLIP Inhibition
- Author
-
Fengyi Wan, Hao Wu, Misonara Ahmed, Jin Kuk Yang, Lixin Zheng, Michael J. Lenardo, and Liwei Wang
- Subjects
Models, Molecular ,Death Domain Receptor Signaling Adaptor Proteins ,Materials science ,Protein Conformation ,Fas-Associated Death Domain Protein ,Molecular Sequence Data ,CASP8 and FADD-Like Apoptosis Regulating Protein ,Apoptosis ,Crystallography, X-Ray ,Caspase 8 ,Article ,Viral Proteins ,Protein structure ,Animals ,Humans ,Amino Acid Sequence ,fas Receptor ,FADD ,Caspase 10 ,Molecular Biology ,Adaptor Proteins, Signal Transducing ,Death domain ,Molluscum contagiosum virus ,biology ,Intracellular Signaling Peptides and Proteins ,Cell Biology ,Tumor Necrosis Factor Receptor-Associated Peptides and Proteins ,Cell biology ,Flip ,Caspases ,Multiprotein Complexes ,Mutation ,Death-inducing signaling complex ,biology.protein ,Death effector domain ,biological phenomena, cell phenomena, and immunity ,Sequence Alignment - Abstract
Summary The death-inducing signaling complex (DISC) comprising Fas, Fas-associated death domain (FADD), and caspase-8/10 is assembled via homotypic associations between death domains (DDs) of Fas and FADD and between death effector domains (DEDs) of FADD and caspase-8/10. Caspase-8/10 and FLICE/caspase-8 inhibitory proteins (FLIPs) that inhibit caspase activation at the DISC level contain tandem DEDs. Here, we report the crystal structure of a viral FLIP, MC159, at 1.2 A resolution. It reveals a noncanonical fold of DED1, a dumbbell-shaped structure with rigidly associated DEDs and a different mode of interaction in the DD superfamily. Whereas the conserved hydrophobic patch of DED1 interacts with DED2, the corresponding region of DED2 mediates caspase-8 recruitment and contributes to DISC assembly. In contrast, MC159 cooperatively assembles with Fas and FADD via an extensive surface that encompasses the conserved charge triad. This interaction apparently competes with FADD self-association and disrupts higher-order oligomerization required for caspase activation in the DISC.
- Published
- 2005
44. Inhibition of Both the Extrinsic and Intrinsic Death Pathways through Nonhomotypic Death-Fold Interactions
- Author
-
Kartik Mani, Young-Jae Nam, Chang Fu Peng, Yukihiro Hayakawa, Peiyee Lee, Stanley J. Korsmeyer, Anthony W. Ashton, Richard N. Kitsis, and Barath Krishnamurthy
- Subjects
Death Domain Receptor Signaling Adaptor Proteins ,Death fold ,Phenylalanine ,Recombinant Fusion Proteins ,Amino Acid Motifs ,Apoptosis ,Mitochondrion ,Models, Biological ,Receptors, Tumor Necrosis Factor ,Adenoviridae ,Cell Line ,Mice ,Proto-Oncogene Proteins ,Two-Hybrid System Techniques ,Animals ,Humans ,Myocytes, Cardiac ,FADD ,fas Receptor ,Molecular Biology ,Caspase ,Cells, Cultured ,Death domain ,bcl-2-Associated X Protein ,Arc (protein) ,biology ,Cell Biology ,Precipitin Tests ,Cell biology ,Protein Structure, Tertiary ,Rats ,Amino Acid Substitution ,Proto-Oncogene Proteins c-bcl-2 ,Caspases ,biology.protein ,Chromatography, Gel ,Death effector domain - Abstract
Death-fold domains constitute an evolutionarily conserved superfamily that mediates apoptotic signaling. These motifs, including CARD (caspase recruitment domain), DD (death domain), and DED (death effector domain), are believed to exert their effects solely through homotypic interactions. Herein we demonstrate that the CARD-containing protein ARC engages in nontraditional death-fold interactions to suppress both extrinsic and intrinsic death pathways. The extrinsic pathway is disrupted by heterotypic interactions between ARC's CARD and the DDs of Fas and FADD, which inhibit Fas-FADD binding and assembly of the death-inducing signaling complex (DISC). The intrinsic pathway is antagonized by ARC-Bax binding, involving ARC's CARD and the Bax C terminus. This inhibits Bax activation and translocation to the mitochondria. Knockdown of endogenous ARC facilitates DISC assembly and triggers spontaneous Bax activation and apoptosis. Conversely, physiological levels of ARC suppress these events. These studies establish a critical role for nonhomotypic death-fold interactions in the regulation of apoptosis.
- Published
- 2004
- Full Text
- View/download PDF
45. Identification of an Expanded Binding Surface on the FADD Death Domain Responsible for Interaction with CD95/Fas
- Author
-
Yu Wei, Justine M. Hill, Tad Kim, Gaku Morisawa, Yufeng Wei, Milton H. Werner, and Ted Huang
- Subjects
Models, Molecular ,Cytoplasm ,Programmed cell death ,DNA, Complementary ,Magnetic Resonance Spectroscopy ,Time Factors ,Cell Survival ,Protein Conformation ,Fas-Associated Death Domain Protein ,Molecular Sequence Data ,Biochemistry ,Protein Structure, Secondary ,Cell Line, Tumor ,Animals ,Humans ,Amino Acid Sequence ,fas Receptor ,FADD ,Molecular Biology ,Adaptor Proteins, Signal Transducing ,Death domain ,Sequence Homology, Amino Acid ,biology ,Cell Membrane ,Cell Biology ,Fas receptor ,Precipitin Tests ,Protein Structure, Tertiary ,Cell biology ,Death-inducing signaling complex ,Mutagenesis, Site-Directed ,biology.protein ,Drosophila ,Death effector domain ,biological phenomena, cell phenomena, and immunity ,Carrier Proteins ,Alpha helix ,Protein Binding ,Signal Transduction - Abstract
The initiation of programmed cell death at CD95 (Fas, Apo-1) is achieved by forming a death-inducing signaling complex (DISC) at the cytoplasmic membrane surface. Assembly of the DISC has been proposed to occur via homotypic interactions between the death domain (DD) of FADD and the cytoplasmic domain of CD95. Previous analysis of the FADD/CD95 interaction led to the identification of a putative CD95 binding surface within FADD DD formed by alpha helices 2 and 3. More detailed analysis of the CD95/FADD DD interaction now demonstrates that a bimodal surface exists in the FADD DD for interaction with CD95. An expansive surface on one side of the domain is composed of elements in alpha helices 1, 2, 3, 5, and 6. This major surface is common to many proteins harboring this motif, whether or not they are associated with programmed cell death. A secondary surface resides on the opposite face of the domain and involves residues in helices 3 and 4. The major surface is topologically similar to the protein interaction surface identified in Drosophila Tube DD and the death effector domain of hamster PEA-15, two physiologically unrelated proteins which interact with structurally unrelated binding partners. These results demonstrate the presence of a structurally conserved surface within the DD which can mediate protein recognition with homo- and heterotypic binding partners, whereas a second surface may be responsible for stabilizing the higher order complex in the DISC.
- Published
- 2004
46. Fas-associated Factor-1 Inhibits Nuclear Factor-κB (NF-κB) Activity by Interfering with Nuclear Translocation of the RelA (p65) Subunit of NF-κB
- Author
-
Min Young Park, Soo Young Lee, Eunhee Kim, Kong-Joo Lee, and Hyunduk Jang
- Subjects
Protein subunit ,Biology ,Biochemistry ,Mice ,chemistry.chemical_compound ,Animals ,Humans ,Molecular Biology ,Adaptor Proteins, Signal Transducing ,Death domain ,Cell Nucleus ,HEK 293 cells ,Intracellular Signaling Peptides and Proteins ,NF-kappa B ,Transcription Factor RelA ,NF-κB ,Cell Biology ,Molecular biology ,Protein Transport ,chemistry ,Apoptosis ,NIH 3T3 Cells ,Tumor necrosis factor alpha ,Death effector domain ,Apoptosis Regulatory Proteins ,Carrier Proteins ,Protein Binding ,Signal Transduction ,Binding domain - Abstract
Fas-associated factor-1 (FAF1) is a Fas-binding pro-apoptotic protein that is a component of the death-inducing signaling complex in Fas-mediated apoptosis. Here, we show that FAF1 is involved in negative regulation of NF-kappaB activation. Overexpression of FAF1 decreased the basal level of NF-kappaB activity in 293 cells. NF-kappaB activation induced by tumor necrosis factor (TNF)-alpha, interleukin-1beta, and lipopolysaccharide was also inhibited by FAF1 overexpression. Moreover, FAF1 suppressed NF-kappaB activation induced by transducers of diverse NF-kappaB-activating signals such as TNF receptor-associated factor-2 and -6, MEKK1, and IkappaB kinase-beta as well as NF-kappaB p65, one of the end point molecules in the NF-kappaB activation pathway, suggesting that NF-kappaB p65 might be a target molecule upon which FAF1 acts. Subsequent study disclosed that FAF1 physically interacts with NF-kappaB p65 and that the binding domain of FAF1 is the death effector domain (DED)-interacting domain (amino acids 181-381), where DEDs of the Fas-associated death domain protein and caspase-8 interact. The NF-kappaB activity-modulating potential of FAF1 was also mapped to the DED-interacting domain. Finally, overexpression of FAF1 prevented translocation of NF-kappaB p65 into the nucleus and decreased its DNA-binding activity upon TNFalpha treatment. This study presents a novel function of FAF1, in addition to the previously known function as a component of the Fas death-inducing signaling complex, i.e. NF-kappaB activity suppressor by cytoplasmic retention of NF-kappaB p65 via physical interaction.
- Published
- 2004
47. Kinetics of cell death in T lymphocytes genetically modified with two novel suicide fusion genes
- Author
-
K Junker, S Zimmerman, Ulrike Koehl, Manuel Grez, T. Klingebiel, Dirk Schwabe, and Stefan Stein
- Subjects
Recombinant Fusion Proteins ,T-Lymphocytes ,Lymphocyte ,Genetic Vectors ,Graft vs Host Disease ,Receptors, Nerve Growth Factor ,Biology ,Antiviral Agents ,Thymidine Kinase ,Viral vector ,Tacrolimus Binding Proteins ,Antigen ,Coenzyme A Ligases ,Genetics ,medicine ,Humans ,Simplexvirus ,FADD ,Ganciclovir ,Molecular Biology ,Death domain ,Cell Death ,Escherichia coli Proteins ,Genetic Therapy ,Virology ,Molecular biology ,Transplantation ,Retroviridae ,medicine.anatomical_structure ,Lymphocyte Transfusion ,biology.protein ,Molecular Medicine ,Death effector domain ,Stem cell ,Genetic Engineering - Abstract
Donor lymphocyte infusions (DLI) following allogeneic stem cell transplantation are known to mediate graft-versus-leukemia effect (GVL). A major side effect of these immunotherapies is the development of graft-versus-host diseases (GVHD). One promising approach to prevent GVHD is to genetically modify donor T cells with a suicide mechanism that can be induced in the case of GVHD. Here we report on a retroviral vector containing the death effector domain (DED) of the human Fas-associated protein with death domain (FADD). The DED was fused to two copies of an FKBP506-binding protein and a truncated version of the human low-affinity receptor for nerve growth factor (LNGFR). Activation of the death signal pathway can be triggered upon the addition of chemical inducers of dimerization. This construct was functionally compared to an optimized HSV-TK vector in which a hypersensitive mutant of the herpes simplex virus thymidine kinase gene (TK39) was fused to a cytoplasmic truncated version of the cell surface antigen CD34. A direct comparison between both vectors in primary T lymphocytes showed that the number of T cells transduced with vectors containing the DED was significantly reduced within 24 h of drug administration whereas ganciclovir treatment of TK39-transduced T cells showed a delay in cell death of approximately 3-4 days. Our results indicate that constructs containing the DED may prove to be the most efficient mechanism to quickly eliminate alloreactive T cells.
- Published
- 2003
48. The death effector domain protein family: regulators of cellular homeostasis
- Author
-
Michael D Tibbetts, Lixin Zheng, and Michael J. Lenardo
- Subjects
Models, Molecular ,Death Domain Receptor Signaling Adaptor Proteins ,Protein family ,Fas-Associated Death Domain Protein ,Molecular Sequence Data ,Immunology ,Cellular homeostasis ,Apoptosis ,Models, Biological ,DNA-binding protein ,Animals ,Homeostasis ,Humans ,Immunology and Allergy ,Amino Acid Sequence ,FADD ,Adaptor Proteins, Signal Transducing ,Death domain ,Caspase 8 ,Sequence Homology, Amino Acid ,biology ,Intracellular Signaling Peptides and Proteins ,Proteins ,Signal transducing adaptor protein ,Caspase 9 ,Protein Structure, Tertiary ,Cell biology ,DNA-Binding Proteins ,Caspases ,biology.protein ,Death effector domain ,Signal transduction ,Carrier Proteins ,Cell Division ,Signal Transduction - Abstract
The death effector domain (DED) occurs in proteins that regulate programmed cell death. Both pro- and anti-apoptotic proteins containing DEDs have been identified. For Fas and possibly other death receptors, homotypic DED interactions connect the Fas-associated death domain (FADD) protein to caspase-8 and caspase-10 to mediate formation of the death-inducing signal complex. This complex can be inhibited by other DED-containing proteins. Accumulating evidence now suggests that DED-containing proteins have additional roles in controlling pathways of cellular activation and proliferation. Thus, the DED defines a family of proteins that may be pivotal to cellular homeostasis by establishing a 'cell renewal set point' that coregulates proliferation and apoptosis in parallel.
- Published
- 2003
49. Fas-associated death domain protein interacts with methyl-CpG binding domain protein 4: A potential link between genome surveillance and apoptosis
- Author
-
Adrian Bird, Owen J. Sansom, Stephan Kiessling, Kathryn Maddison, Robert A. Screaton, Catherine B. Millar, Steven M. Frisch, and Alan Richard Clarke
- Subjects
Fas-Associated Death Domain Protein ,Apoptosis ,Karyopherins ,urologic and male genital diseases ,Fas ligand ,Cell Line ,MBD4 ,Mice ,Animals ,Humans ,FADD ,Adaptor Proteins, Signal Transducing ,Death domain ,Cell Nucleus ,Mice, Knockout ,Endodeoxyribonucleases ,Genome ,Multidisciplinary ,biology ,Biological Sciences ,Fas receptor ,Molecular biology ,ran GTP-Binding Protein ,Death-inducing signaling complex ,biology.protein ,Death effector domain ,biological phenomena, cell phenomena, and immunity ,Carrier Proteins ,Binding domain - Abstract
Fas-associated death domain protein (FADD) is an adaptor protein bridging death receptors with initiator caspases. Thus, its function and localization are assumed to be cytoplasmic, although the localization of endogenous FADD has not been reported. Surprisingly, the data presented here demonstrate that FADD is mainly nuclear in several adherent cell lines. Its accumulation in the nucleus and export to the cytoplasm required the phosphorylation site Ser-194, which was also required for its interaction with the nucleocytoplasmic shuttling protein exportin-5. Within the nucleus, FADD interacted with the methyl-CpG binding domain protein 4 (MBD4), which excises thymine from GT mismatches in methylated regions of chromatin. The MBD4-interacting mismatch repair factor MLH1 was also found in a complex with FADD. The FADD–MBD4 interaction involved the death effector domain of FADD and a region of MBD4 adjacent to the glycosylase domain. The FADD-binding region of MBD4 was downstream of a frameshift mutation that occurs in a significant fraction of human colorectal carcinomas. Consistent with the idea that MBD4 can signal to an apoptotic effector, MBD4 regulated DNA damage-, Fas ligand-, and cell detachment-induced apoptosis. The nuclear localization of FADD and its interaction with a genome surveillance/DNA repair protein that can regulate apoptosis suggests a novel function of FADD distinct from direct participation in death receptor signaling complexes.
- Published
- 2003
50. Caspase- and Serine Protease-dependent Apoptosis by the Death Domain of FADD in Normal Epithelial Cells
- Author
-
Andrew Thorburn, Jacqueline Thorburn, Michael J. Morgan, and Laura M. Bender
- Subjects
Male ,Fas-Associated Death Domain Protein ,Genetic Vectors ,Apoptosis ,Epithelium ,Article ,Adenoviridae ,TNF-Related Apoptosis-Inducing Ligand ,Humans ,FADD ,Molecular Biology ,Caspase ,Adaptor Proteins, Signal Transducing ,Death domain ,Serine protease ,Membrane Glycoproteins ,biology ,Tumor Necrosis Factor-alpha ,Serine Endopeptidases ,Gene Transfer Techniques ,Prostate ,Signal transducing adaptor protein ,Cell Biology ,Molecular biology ,Protein Structure, Tertiary ,Cell biology ,Caspases ,biology.protein ,Death effector domain ,Tumor necrosis factor alpha ,Apoptosis Regulatory Proteins ,Carrier Proteins - Abstract
The adapter protein FADD consists of two protein interaction domains: a death domain and a death effector domain. The death domain binds to activated death receptors such as Fas, whereas the death effector domain binds to procaspase 8. An FADD mutant, which consists of only the death domain (FADD-DD), inhibits death receptor–induced apoptosis. FADD-DD can also activate a mechanistically distinct, cell type–specific apoptotic pathway that kills normal but not cancerous prostate epithelial cells. Here, we show that this apoptosis occurs through activation of caspases 9, 3, 6, and 7 and a serine protease. Simultaneous inhibition of caspases and serine proteases prevents FADD-DD–induced death. Inhibition of either pathway alone does not prevent cell death but does affect the morphology of the dying cells. Normal prostate epithelial cells require both the caspase and serine protease inhibitors to efficiently prevent apoptosis in response to TRAIL. In contrast, the serine protease inhibitor does not affect TRAIL-induced death in prostate tumor cells suggesting that the FADD-DD–dependent pathway can be activated by TRAIL. This apoptosis pathway is activated in a cell type–specific manner that is defective in cancer cells, suggesting that this pathway may be targeted during cancer development.
- Published
- 2003
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.