SPATA2 links CYLD to the TNF‐α receptor signaling complex and modulates the receptor signaling outcomes

TNF‐α is a key regulator of innate immune and proinflammatory responses. However, the composition of the TNF‐α receptor‐associated signaling complexes (TNF‐RSC) and the architecture of the downstream signaling networks are incompletely understood. We employed quantitative mass spectrometry to demonstrate that TNF‐α stimulation induces widespread protein phosphorylation and that the scope of phosphorylation expands in a temporal manner. TNF‐α stimulation also induces rapid ubiquitylation of components of the TNF‐RSC. Temporal analysis of the TNF‐RSC composition identified SPATA2 as a novel component of the TNF‐RSC. The predicted PUB domain in the N‐terminus of SPATA2 interacts with the USP domain of CYLD, whereas the C‐terminus of SPATA2 interacts with HOIP. SPATA2 is required for recruitment of CYLD to the TNF‐RSC. Downregulation of SPATA2 augments transcriptional activation of NF‐κB and inhibits TNF‐α‐induced necroptosis, pointing to an important function of SPATA2 in modulating the outcomes of TNF‐α signaling. Taken together, our study draws a detailed map of TNF‐α signaling, identifies SPATA2 as a novel component of TNF‐α signaling, and provides a rich resource for further functional investigations.

[1]  K. Rittinger,et al.  LUBAC-Recruited CYLD and A20 Regulate Gene Activation and Cell Death by Exerting Opposing Effects on Linear Ubiquitin in Signaling Complexes , 2015, Cell reports.

[2]  Chunaram Choudhary,et al.  Systems-wide analysis of BCR signalosomes and downstream phosphorylation and ubiquitylation , 2015, Molecular systems biology.

[3]  Narmada Thanki,et al.  CDD: NCBI's conserved domain database , 2014, Nucleic Acids Res..

[4]  K. Iwai,et al.  Linear ubiquitin chains: NF-κB signalling, cell death and beyond , 2014, Nature Reviews Molecular Cell Biology.

[5]  Roger J. Davis,et al.  TNF and MAP kinase signalling pathways. , 2014, Seminars in immunology.

[6]  P. R. Elliott,et al.  Molecular Basis and Regulation of OTULIN-LUBAC Interaction , 2014, Molecular cell.

[7]  Michael H. Olma,et al.  Binding of OTULIN to the PUB domain of HOIP controls NF-κB signaling. , 2014, Molecular cell.

[8]  Y. Saeki,et al.  Suppression of LUBAC‐mediated linear ubiquitination by a specific interaction between LUBAC and the deubiquitinases CYLD and OTULIN , 2014, Genes to cells : devoted to molecular & cellular mechanisms.

[9]  J. Silke,et al.  IAP family of cell death and signaling regulators. , 2014, Methods in enzymology.

[10]  M. Mann,et al.  Status of Large-scale Analysis of Post-translational Modifications by Mass Spectrometry* , 2013, Molecular & Cellular Proteomics.

[11]  F. Chan,et al.  CYLD Deubiquitinates RIP1 in the TNFα-Induced Necrosome to Facilitate Kinase Activation and Programmed Necrosis , 2013, PloS one.

[12]  Toru Okamoto,et al.  The pseudokinase MLKL mediates necroptosis via a molecular switch mechanism. , 2013, Immunity.

[13]  Sebastian A. Wagner,et al.  Lysine succinylation is a frequently occurring modification in prokaryotes and eukaryotes and extensively overlaps with acetylation. , 2013, Cell reports.

[14]  Yingying Zhang,et al.  Mlkl knockout mice demonstrate the indispensable role of Mlkl in necroptosis , 2013, Cell Research.

[15]  Sebastian A. Wagner,et al.  OTULIN restricts Met1-linked ubiquitination to control innate immune signaling. , 2013, Molecular cell.

[16]  Kay Hofmann,et al.  OTULIN Antagonizes LUBAC Signaling by Specifically Hydrolyzing Met1-Linked Polyubiquitin , 2013, Cell.

[17]  Tharan Srikumar,et al.  The linear ubiquitin-specific deubiquitinase gumby regulates angiogenesisThe linear ubiquitin-specific deubiquitinase gumby regulates angiogenesis , 2013 .

[18]  Wade H. Dunham,et al.  The linear ubiquitin-specific deubiquitinase Gumby/Fam105b regulates angiogenesis , 2013, Nature.

[19]  S. Mohammed,et al.  Robust phosphoproteome enrichment using monodisperse microsphere–based immobilized titanium (IV) ion affinity chromatography , 2013, Nature Protocols.

[20]  C. Benedict,et al.  Clinical targeting of the TNF and TNFR superfamilies , 2013, Nature Reviews Drug Discovery.

[21]  D. Tang,et al.  PKR-Dependent Inflammatory Signals , 2012, Science Signaling.

[22]  R. Aebersold,et al.  Mass spectrometry-based proteomics and network biology. , 2012, Annual review of biochemistry.

[23]  Richard J. Lavallee,et al.  Optimized fast and sensitive acquisition methods for shotgun proteomics on a quadrupole orbitrap mass spectrometer. , 2012, Journal of proteome research.

[24]  Xiaodong Wang,et al.  Mixed Lineage Kinase Domain-like Protein Mediates Necrosis Signaling Downstream of RIP3 Kinase , 2012, Cell.

[25]  Chuan-Qi Zhong,et al.  Programmed necrosis: backup to and competitor with apoptosis in the immune system , 2011, Nature Immunology.

[26]  H. Walczak TNF and ubiquitin at the crossroads of gene activation, cell death, inflammation, and cancer , 2011, Immunological reviews.

[27]  Edward L. Huttlin,et al.  Systematic and quantitative assessment of the ubiquitin-modified proteome. , 2011, Molecular cell.

[28]  J. Silke The regulation of TNF signalling: what a tangled web we weave. , 2011, Current opinion in immunology.

[29]  R. Xavier,et al.  CASPASE 8 inhibits programmed necrosis by processing CYLD , 2011, Nature Cell Biology.

[30]  A. High,et al.  The E3 Ubiquitin Ligase Mind Bomb-2 (MIB2) Protein Controls B-cell CLL/Lymphoma 10 (BCL10)-dependent NF-κB Activation* , 2011, The Journal of Biological Chemistry.

[31]  Sebastian A. Wagner,et al.  A Proteome-wide, Quantitative Survey of In Vivo Ubiquitylation Sites Reveals Widespread Regulatory Roles* , 2011, Molecular & Cellular Proteomics.

[32]  J. Colgan,et al.  HOIL-1L Interacting Protein (HOIP) Is Essential for CD40 Signaling , 2011, PloS one.

[33]  Sebastian A. Wagner,et al.  Phosphorylation of the Autophagy Receptor Optineurin Restricts Salmonella Growth , 2011, Science.

[34]  M. Mann,et al.  Mass Spectrometry-based Proteomics Using Q Exactive, a High-performance Benchtop Quadrupole Orbitrap Mass Spectrometer* , 2011, Molecular & Cellular Proteomics.

[35]  M. Mann,et al.  Andromeda: a peptide search engine integrated into the MaxQuant environment. , 2011, Journal of proteome research.

[36]  Damian Szklarczyk,et al.  The STRING database in 2011: functional interaction networks of proteins, globally integrated and scored , 2010, Nucleic Acids Res..

[37]  V. Dixit,et al.  Deubiquitinases in the regulation of NF-κB signaling , 2011, Cell Research.

[38]  M. Mann,et al.  Decoding signalling networks by mass spectrometry-based proteomics , 2010, Nature Reviews Molecular Cell Biology.

[39]  K. Gevaert,et al.  Improved visualization of protein consensus sequences by iceLogo , 2009, Nature Methods.

[40]  P. Vandenabeele,et al.  RIP Kinases at the Crossroads of Cell Death and Survival , 2009, Cell.

[41]  Tao Wang,et al.  Receptor Interacting Protein Kinase-3 Determines Cellular Necrotic Response to TNF-α , 2009, Cell.

[42]  S. Akira,et al.  Involvement of linear polyubiquitylation of NEMO in NF-κB activation , 2009, Nature Cell Biology.

[43]  Alexei Degterev,et al.  Identification of a Molecular Signaling Network that Regulates a Cellular Necrotic Cell Death Pathway , 2008, Cell.

[44]  M. Mann,et al.  MaxQuant enables high peptide identification rates, individualized p.p.b.-range mass accuracies and proteome-wide protein quantification , 2008, Nature Biotechnology.

[45]  V. Dötsch,et al.  Ubiquitin binding mediates the NF-κB inhibitory potential of ABIN proteins , 2008, Oncogene.

[46]  J. Bradley,et al.  TNF‐mediated inflammatory disease , 2008, The Journal of pathology.

[47]  Michael L. Creech,et al.  Integration of biological networks and gene expression data using Cytoscape , 2007, Nature Protocols.

[48]  M. Mann,et al.  Higher-energy C-trap dissociation for peptide modification analysis , 2007, Nature Methods.

[49]  P. Vandenabeele,et al.  RIP1, a kinase on the crossroads of a cell's decision to live or die , 2007, Cell Death and Differentiation.

[50]  Steven P Gygi,et al.  Target-decoy search strategy for increased confidence in large-scale protein identifications by mass spectrometry , 2007, Nature Methods.

[51]  M. Mann,et al.  Protocol for micro-purification, enrichment, pre-fractionation and storage of peptides for proteomics using StageTips , 2007, Nature Protocols.

[52]  M. Mann,et al.  Global, In Vivo, and Site-Specific Phosphorylation Dynamics in Signaling Networks , 2006, Cell.

[53]  Zhijian J. Chen Ubiquitin signalling in the NF-κB pathway , 2005, Nature Cell Biology.

[54]  Alexei Degterev,et al.  Chemical inhibitor of nonapoptotic cell death with therapeutic potential for ischemic brain injury , 2005, Nature chemical biology.

[55]  Zhijian J. Chen Ubiquitin signalling in the NF-kappaB pathway. , 2005, Nature cell biology.

[56]  L. Cantley,et al.  The Crohn's Disease Protein, NOD2, Requires RIP2 in Order to Induce Ubiquitinylation of a Novel Site on NEMO , 2004, Current Biology.

[57]  S. Johansson,et al.  β1‐Integrins induce phosphorylation of Akt on serine 473 independently of focal adhesion kinase and Src family kinases , 2004, EMBO reports.

[58]  B. Aggarwal Signalling pathways of the TNF superfamily: a double-edged sword , 2003, Nature Reviews Immunology.

[59]  René Bernards,et al.  Loss of the cylindromatosis tumour suppressor inhibits apoptosis by activating NF-κB , 2003, Nature.

[60]  A. Ashworth,et al.  CYLD is a deubiquitinating enzyme that negatively regulates NF-κB activation by TNFR family members , 2003, Nature.

[61]  G. Courtois,et al.  The tumour suppressor CYLD negatively regulates NF-κB signalling by deubiquitination , 2003, Nature.

[62]  Jinyu Ren,et al.  Omi/HtrA2 catalytic cleavage of inhibitor of apoptosis (IAP) irreversibly inactivates IAPs and facilitates caspase activity in apoptosis. , 2003, Genes & development.

[63]  D. Goeddel,et al.  TNF-R1 Signaling: A Beautiful Pathway , 2002, Science.

[64]  M. Mann,et al.  Stable Isotope Labeling by Amino Acids in Cell Culture, SILAC, as a Simple and Accurate Approach to Expression Proteomics* , 2002, Molecular & Cellular Proteomics.

[65]  S. Srinivasula,et al.  The serine protease Omi/HtrA2 is released from mitochondria during apoptosis. Omi interacts with caspase-inhibitor XIAP and induces enhanced caspase activity , 2002, Cell Death and Differentiation.

[66]  H. Nakayama,et al.  A serine protease, HtrA2, is released from the mitochondria and interacts with XIAP, inducing cell death. , 2001, Molecular cell.

[67]  R. Beyaert,et al.  Functional redundancy of the zinc fingers of A20 for inhibition of NF‐κB activation and protein–protein interactions , 2001, FEBS letters.

[68]  R. Locksley,et al.  The TNF and TNF Receptor Superfamilies Integrating Mammalian Biology , 2001, Cell.

[69]  E. Harhaj,et al.  NF-κB-Inducing Kinase Regulates the Processing of NF-κB2 p100 , 2001 .

[70]  E. Harhaj,et al.  NF-kappaB-inducing kinase regulates the processing of NF-kappaB2 p100. , 2001, Molecular cell.

[71]  M. Karin,et al.  Phosphorylation meets ubiquitination: the control of NF-[kappa]B activity. , 2000, Annual review of immunology.

[72]  A. Israël,et al.  I kappa B epsilon, a novel member of the IκB family, controls RelA and cRel NF‐κB activity , 1997 .

[73]  A. Israël,et al.  I kappa B epsilon, a novel member of the I kappa B family, controls RelA and cRel NF-kappa B activity. , 1997, The EMBO journal.

[74]  S. Gerstberger,et al.  Control of I kappa B-alpha proteolysis by site-specific, signal-induced phosphorylation , 1995, Science.

[75]  J. Darnell,et al.  Stat3: a STAT family member activated by tyrosine phosphorylation in response to epidermal growth factor and interleukin-6. , 1994, Science.