首页 | 本学科首页   官方微博 | 高级检索  
   检索      


HSP90 activity is required for MLKL oligomerisation and membrane translocation and the induction of necroptotic cell death
Authors:A V Jacobsen  K N Lowes  M C Tanzer  I S Lucet  J M Hildebrand  E J Petrie  M F van Delft  Z Liu  S A Conos  J-G Zhang  D C S Huang  J Silke  G Lessene  J M Murphy
Institution:1The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia;2Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia;3Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, VIC, Australia
Abstract:Necroptosis is a caspase-independent form of regulated cell death that has been implicated in the development of a range of inflammatory, autoimmune and neurodegenerative diseases. The pseudokinase, Mixed Lineage Kinase Domain-Like (MLKL), is the most terminal known obligatory effector in the necroptosis pathway, and is activated following phosphorylation by Receptor Interacting Protein Kinase-3 (RIPK3). Activated MLKL translocates to membranes, leading to membrane destabilisation and subsequent cell death. However, the molecular interactions governing the processes downstream of RIPK3 activation remain poorly defined. Using a phenotypic screen, we identified seven heat-shock protein 90 (HSP90) inhibitors that inhibited necroptosis in both wild-type fibroblasts and fibroblasts expressing an activated mutant of MLKL. We observed a modest reduction in MLKL protein levels in human and murine cells following HSP90 inhibition, which was only apparent after 15 h of treatment. The delayed reduction in MLKL protein abundance was unlikely to completely account for defective necroptosis, and, consistent with this, we also found inhibition of HSP90 blocked membrane translocation of activated MLKL. Together, these findings implicate HSP90 as a modulator of necroptosis at the level of MLKL, a function that complements HSP90''s previously demonstrated modulation of the upstream necroptosis effector kinases, RIPK1 and RIPK3.Necroptosis is an inflammatory, caspase-independent form of regulated cell death characterised by loss of cellular membrane integrity and release of cytoplasmic contents.1 It is believed to have evolved as a defence mechanism against viruses;2, 3 however, there is increasing evidence that deregulated necroptosis has a role in the pathogenesis of a range of inflammatory, autoimmune and neurodegenerative diseases.4, 5, 6, 7, 8 Reduced capacity to undergo necroptosis has been correlated to increased aggressiveness of cancers;9, 10 and therapeutic initiation of necroptosis is currently being investigated as a cancer therapy.11, 12 Additionally, there is emerging evidence that the necroptotic signalling pathway has a general role in the modulation of inflammation.13, 14, 15, 16, 17 As such, unravelling the molecular events governing necroptosis, and potential avenues for therapeutic intervention, is of enormous interest.Necroptosis is initiated through activation of death receptors, such as Tumour Necrosis Factor Receptor 1 (TNFR1), or through microbial activation of pattern recognition receptors, such as Toll-like receptors or intracellular viral DNA sensors.3, 18, 19, 20 Receptor ligation initiates a signalling cascade, whereby Receptor Interacting Protein Kinase (RIPK)-3 oligomerises and is phosphorylated, a process known to be regulated by association with other effectors, such as the protein kinase RIPK1, TIR-domain-containing adapter-inducing IFN-β (TRIF), or DNA-dependent activator of IFN regulatory factors (DAI), via their RIP Homotypic Interaction Motifs (RHIMs).2, 21, 22 Once activated, RIPK3 phosphorylates the pseudokinase domain of Mixed Lineage Kinase domain-Like (MLKL), the most downstream known obligate effector of the necroptotic signalling pathway, to induce its activation.23, 24 MLKL phosphorylation is thought to trigger a molecular switch,25, 26, 27 leading to the unleashing of the N-terminal executioner four-helix bundle (4HB) domain,28 MLKL oligomerisation and translocation to cellular membranes where cell death occurs via an incompletely-understood mechanism.28, 29, 30Molecular chaperones have an integral role in modulating both the structure and function of proteins. One such chaperone is heat-shock protein 90 (HSP90), which interacts with a diverse group of protein ‘clients'', the largest group comprising the kinases and pseudokinases, with 50% of the human kinome estimated to interact with HSP90.31 These interactions are dependent on the recognition of the kinase or pseudokinase domain by the HSP90 co-chaperone Cdc37, which enables HSP90 to confer protein stabilisation, assist in late-stage folding and conformational modifications, and mediate intracellular transport.32, 33, 34, 35It has already been demonstrated that the necroptotic pathway is subject to modulation by HSP90. RIPK1 is well established as an HSP90 client protein, with a number of studies finding HSP90 inhibition affects both the stability and function of RIPK1 and promotes an apoptotic phenotype.36, 37, 38, 39, 40, 41 More recently, RIPK3 was also identified as an HSP90 client.2, 42, 43 Surprisingly, HSP90 inhibition did not markedly impact RIPK3 abundance or stability, but rather was essential for RIPK3''s necroptotic functions, such as phosphorylation of MLKL.42 However, whether MLKL itself is a client of HSP90 has not been investigated.In this study, using a phenotypic screen for small-molecule inhibitors of MLKL-driven cell death, we identified HSP90 as a modulator of necroptosis that functions on, or downstream of, the terminal effector, MLKL. HSP90 inhibition did not markedly reduce levels of MLKL in human U937 or mouse dermal fibroblasts, suggesting instead that HSP90 has an active role in governing MLKL-mediated cell death. This idea is supported by our finding that cell death driven by the S345D activated mutant of MLKL in Ripk3-deficient fibroblasts in the absence of necroptotic stimuli was suppressed by three distinct chemical classes of HSP90 inhibitor, but MLKL abundance was not impacted by HSP90 inhibition. Although our data indicate that MLKL binds HSP90 weakly or transiently, HSP90 activity was essential for the assembly of MLKL into high molecular weight complexes and the membrane translocation known to precede cell death. These findings suggest an expanded role for HSP90 in regulating necroptosis, and further our understanding of the mechanisms controlling MLKL-mediated cell death.
Keywords:
设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号