activation of the pseudokinase mlkl unleashes the four ... · activation of the pseudokinase mlkl...

6
Activation of the pseudokinase MLKL unleashes the four-helix bundle domain to induce membrane localization and necroptotic cell death Joanne M. Hildebrand a,b,1 , Maria C. Tanzer a,b,1 , Isabelle S. Lucet a,b,c , Samuel N. Young a , Sukhdeep K. Spall a , Pooja Sharma a,b , Catia Pierotti a , Jean-Marc Garnier a,b , Renwick C. J. Dobson d,e , Andrew I. Webb a,b , Anne Tripaydonis a,b , Jeffrey J. Babon a,b , Mark D. Mulcair f , Martin J. Scanlon f , Warren S. Alexander a,b , Andrew F. Wilks g , Peter E. Czabotar a,b , Guillaume Lessene a,b,h , James M. Murphy a,b,2,3 , and John Silke a,b,2,3 a The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Departments of b Medical Biology and h Pharmacology and Therapeutics, University of Melbourne, Parkville, VIC 3050, Australia; c Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Monash University, Clayton, VIC 3800, Australia; d Biomolecular Interactions Centre, School of Biological Sciences, University of Canterbury, Christchurch, New Zealand; e Department of Biochemistry, Bio21 Institute, University of Melbourne, Parkville, VIC 3052, Australia; f Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia; and g SynThesis MedChem, Parkville, VIC 3052, Australia Edited by James A. Wells, University of California, San Francisco, CA, and approved September 15, 2014 (received for review May 15, 2014) Necroptosis is considered to be complementary to the classical caspase-dependent programmed cell death pathway, apoptosis. The pseudokinase Mixed Lineage Kinase Domain-Like (MLKL) is an essential effector protein in the necroptotic cell death pathway downstream of the protein kinase Receptor Interacting Protein Kinase-3 (RIPK3). How MLKL causes cell death is unclear, however RIPK3mediated phosphorylation of the activation loop in MLKL trips a molecular switch to induce necroptotic cell death. Here, we show that the MLKL pseudokinase domain acts as a latch to re- strain the N-terminal four-helix bundle (4HB) domain and that unleashing this domain results in formation of a high-molecular- weight, membrane-localized complex and cell death. Using alanine- scanning mutagenesis, we identified two clusters of residues on opposing faces of the 4HB domain that were required for the 4HB domain to kill cells. The integrity of one cluster was essential for membrane localization, whereas MLKL mutations in the other cluster did not prevent membrane translocation but prevented killing; this demonstrates that membrane localization is necessary, but insufficient, to induce cell death. Finally, we identified a small molecule that binds the nucleotide binding site within the MLKL pseudokinase domain and retards MLKL translocation to mem- branes, thereby preventing necroptosis. This inhibitor provides a novel tool to investigate necroptosis and demonstrates the feasibility of using small molecules to target the nucleotide binding site of pseudokinases to modulate signal transduction. pseudoenzyme | RIP kinase | ATP mimetic | programmed necrosis P rogrammed necrosis or necroptosishas emerged in the past 5 years as a cell death mechanism that complements the conventional cell death pathway, apoptosis, in multicellular organisms. In contrast to apoptosis, necroptosis does not appear to serve an important role in multicellular organism development (13) but participates in the defense against pathogens and is a likely culprit in destructive inflammatory conditions (47). Receptor Interacting Protein Kinase-3 (RIPK3) was identified as a key effector of necroptosis in 2009 (4, 5) and its substrate, the pseudokinase Mixed Line- age Kinase Domain-Like (MLKL), in 2012 (8, 9), but the mo- lecular events following RIPK3-mediated phosphorylation of MLKL required to induce cell death are unclear. The RIPK1/ RIPK3/MLKL necrosome has been proposed to activate PGAM5 (phosphoglycerate mutase 5) and Drp1 (Dynamin-related pro- tein 1) to cause mitochondrial fragmentation and cell death (10), but the requirement for PGAM5, Drp1, and mitochondria for necroptosis has been questioned (1, 1113). We described the structure of mouse MLKL revealing that MLKL contains a C-terminal pseudokinase domain and an N-terminal four-helix bundle (4HB) domain connected by a two-helix linker (the bracehelices) (1). Based on our muta- tional and biochemical analyses, we proposed that the catalyti- cally inactive pseudokinase domain functions as a molecular switch and that RIPK3-mediated phosphorylation triggers this switch by inducing a conformational change in MLKL (1, 14). Recently it has been proposed that the 4HB domain is the death effector domain within MLKL and that the killing function of MLKL relies on its oligomerization and plasma membrane association (1518). The stoichiometry of the oligomer is, how- ever, contentious and has been reported to contain three (15), four (16), and possibly six (17) MLKL protomers. Furthermore, several mechanisms for how this oligomer causes cell death have been proposed: Cai et al. proposed it activates the calcium channel protein Tprm7 and promotes calcium influx (15), Chen et al. showed it increased sodium influx (16), and Wang et al. proposed that the oligomerized form of MLKL has the ability to bind negatively charged lipids, including phosphoinositides and Significance The four-helix bundle (4HB) domain of Mixed Lineage Kinase Domain-Like (MLKL) bears two clusters of residues that are required for cell death by necroptosis. Mutations within a cluster centered on the α4 helix of the 4HB domain of MLKL prevented its membrane translocation, oligomerization, and ability to induce necroptosis. This cluster is composed princi- pally of acidic residues and therefore challenges the idea that the 4HB domain engages negatively charged phospholipid membranes via a conventional positively charged interaction surface. The importance of membrane translocation to MLKL- mediated death is supported by our identification of a small molecule that binds the MLKL pseudokinase domain and retards membrane translocation to inhibit necroptotic signaling. Author contributions: J.M.H., M.C.T., I.S.L., C.P., J.-M.G., M.J.S., P.E.C., G.L., J.M.M., and J.S. designed research; J.M.H., M.C.T., I.S.L., S.N.Y., S.K.S., P.S., C.P., J.-M.G., R.C.J.D., A.I.W., A.T., M.D.M., and J.M.M. performed research; J.M.H., M.C.T., I.S.L., P.S., R.C.J.D., A.I.W., J.J.B., M.D.M., M.J.S., W.S.A., A.F.W., P.E.C., G.L., J.M.M., and J.S. analyzed data; and J.M.H., J.M.M., and J.S. wrote the paper. Conflict of interest statement: J.S. is a member of the Scientific Advisory Board of Tetralogic Pharmaceuticals. A.F.W. is Chair of SynThesis MedChem and a Director of Catalyst Therapeutics. This article is a PNAS Direct Submission. 1 J.M.H. and M.C.T. contributed equally to this work. 2 J.M.M. and J.S. contributed equally to this work. 3 To whom correspondence may be addressed. Email: [email protected] or silke@wehi. edu.au. This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10. 1073/pnas.1408987111/-/DCSupplemental. 1507215077 | PNAS | October 21, 2014 | vol. 111 | no. 42 www.pnas.org/cgi/doi/10.1073/pnas.1408987111 Downloaded by guest on May 26, 2020

Upload: others

Post on 25-May-2020

8 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Activation of the pseudokinase MLKL unleashes the four ... · Activation of the pseudokinase MLKL unleashes the four-helix bundle domain to induce membrane localization and necroptotic

Activation of the pseudokinase MLKL unleashes thefour-helix bundle domain to induce membranelocalization and necroptotic cell deathJoanne M. Hildebranda,b,1, Maria C. Tanzera,b,1, Isabelle S. Luceta,b,c, Samuel N. Younga, Sukhdeep K. Spalla,Pooja Sharmaa,b, Catia Pierottia, Jean-Marc Garniera,b, Renwick C. J. Dobsond,e, Andrew I. Webba,b, Anne Tripaydonisa,b,Jeffrey J. Babona,b, Mark D. Mulcairf, Martin J. Scanlonf, Warren S. Alexandera,b, Andrew F. Wilksg, Peter E. Czabotara,b,Guillaume Lessenea,b,h, James M. Murphya,b,2,3, and John Silkea,b,2,3

aThe Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Departments of bMedical Biology and hPharmacology andTherapeutics, University of Melbourne, Parkville, VIC 3050, Australia; cDepartment of Biochemistry and Molecular Biology, School of Biomedical Sciences,Monash University, Clayton, VIC 3800, Australia; dBiomolecular Interactions Centre, School of Biological Sciences, University of Canterbury, Christchurch, NewZealand; eDepartment of Biochemistry, Bio21 Institute, University of Melbourne, Parkville, VIC 3052, Australia; fMedicinal Chemistry, Monash Institute ofPharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia; and gSynThesis MedChem, Parkville, VIC 3052, Australia

Edited by James A. Wells, University of California, San Francisco, CA, and approved September 15, 2014 (received for review May 15, 2014)

Necroptosis is considered to be complementary to the classicalcaspase-dependent programmed cell death pathway, apoptosis.The pseudokinase Mixed Lineage Kinase Domain-Like (MLKL) is anessential effector protein in the necroptotic cell death pathwaydownstream of the protein kinase Receptor Interacting ProteinKinase-3 (RIPK3). How MLKL causes cell death is unclear, howeverRIPK3–mediated phosphorylation of the activation loop in MLKLtrips a molecular switch to induce necroptotic cell death. Here, weshow that the MLKL pseudokinase domain acts as a latch to re-strain the N-terminal four-helix bundle (4HB) domain and thatunleashing this domain results in formation of a high-molecular-weight, membrane-localized complex and cell death. Using alanine-scanning mutagenesis, we identified two clusters of residues onopposing faces of the 4HB domain that were required for the4HB domain to kill cells. The integrity of one cluster was essentialfor membrane localization, whereas MLKL mutations in the othercluster did not prevent membrane translocation but preventedkilling; this demonstrates that membrane localization is necessary,but insufficient, to induce cell death. Finally, we identified a smallmolecule that binds the nucleotide binding site within the MLKLpseudokinase domain and retards MLKL translocation to mem-branes, thereby preventing necroptosis. This inhibitor providesa novel tool to investigate necroptosis and demonstrates thefeasibility of using small molecules to target the nucleotidebinding site of pseudokinases to modulate signal transduction.

pseudoenzyme | RIP kinase | ATP mimetic | programmed necrosis

Programmed necrosis or “necroptosis” has emerged in the past5 years as a cell death mechanism that complements the

conventional cell death pathway, apoptosis, in multicellularorganisms. In contrast to apoptosis, necroptosis does notappear to serve an important role in multicellular organismdevelopment (1–3) but participates in the defense againstpathogens and is a likely culprit in destructive inflammatoryconditions (4–7). Receptor Interacting Protein Kinase-3(RIPK3) was identified as a key effector of necroptosis in2009 (4, 5) and its substrate, the pseudokinase Mixed Line-age Kinase Domain-Like (MLKL), in 2012 (8, 9), but the mo-lecular events following RIPK3-mediated phosphorylation ofMLKL required to induce cell death are unclear. The RIPK1/RIPK3/MLKL necrosome has been proposed to activate PGAM5(phosphoglycerate mutase 5) and Drp1 (Dynamin-related pro-tein 1) to cause mitochondrial fragmentation and cell death (10),but the requirement for PGAM5, Drp1, and mitochondria fornecroptosis has been questioned (1, 11–13).We described the structure of mouse MLKL revealing that

MLKL contains a C-terminal pseudokinase domain and an

N-terminal four-helix bundle (4HB) domain connected by atwo-helix linker (the “brace” helices) (1). Based on our muta-tional and biochemical analyses, we proposed that the catalyti-cally inactive pseudokinase domain functions as a molecularswitch and that RIPK3-mediated phosphorylation triggers thisswitch by inducing a conformational change in MLKL (1, 14).Recently it has been proposed that the 4HB domain is the

death effector domain within MLKL and that the killing functionof MLKL relies on its oligomerization and plasma membraneassociation (15–18). The stoichiometry of the oligomer is, how-ever, contentious and has been reported to contain three (15),four (16), and possibly six (17) MLKL protomers. Furthermore,several mechanisms for how this oligomer causes cell death havebeen proposed: Cai et al. proposed it activates the calciumchannel protein Tprm7 and promotes calcium influx (15), Chenet al. showed it increased sodium influx (16), and Wang et al.proposed that the oligomerized form of MLKL has the ability tobind negatively charged lipids, including phosphoinositides and

Significance

The four-helix bundle (4HB) domain of Mixed Lineage KinaseDomain-Like (MLKL) bears two clusters of residues that arerequired for cell death by necroptosis. Mutations within acluster centered on the α4 helix of the 4HB domain of MLKLprevented its membrane translocation, oligomerization, andability to induce necroptosis. This cluster is composed princi-pally of acidic residues and therefore challenges the idea thatthe 4HB domain engages negatively charged phospholipidmembranes via a conventional positively charged interactionsurface. The importance of membrane translocation to MLKL-mediated death is supported by our identification of a smallmolecule that binds the MLKL pseudokinase domain and retardsmembrane translocation to inhibit necroptotic signaling.

Author contributions: J.M.H., M.C.T., I.S.L., C.P., J.-M.G., M.J.S., P.E.C., G.L., J.M.M., and J.S.designed research; J.M.H., M.C.T., I.S.L., S.N.Y., S.K.S., P.S., C.P., J.-M.G., R.C.J.D., A.I.W.,A.T., M.D.M., and J.M.M. performed research; J.M.H., M.C.T., I.S.L., P.S., R.C.J.D., A.I.W.,J.J.B., M.D.M., M.J.S., W.S.A., A.F.W., P.E.C., G.L., J.M.M., and J.S. analyzed data; and J.M.H.,J.M.M., and J.S. wrote the paper.

Conflict of interest statement: J.S. is a member of the Scientific Advisory Board ofTetralogic Pharmaceuticals. A.F.W. is Chair of SynThesis MedChem and a Director ofCatalyst Therapeutics.

This article is a PNAS Direct Submission.1J.M.H. and M.C.T. contributed equally to this work.2J.M.M. and J.S. contributed equally to this work.3To whom correspondence may be addressed. Email: [email protected] or [email protected].

This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10.1073/pnas.1408987111/-/DCSupplemental.

15072–15077 | PNAS | October 21, 2014 | vol. 111 | no. 42 www.pnas.org/cgi/doi/10.1073/pnas.1408987111

Dow

nloa

ded

by g

uest

on

May

26,

202

0

Page 2: Activation of the pseudokinase MLKL unleashes the four ... · Activation of the pseudokinase MLKL unleashes the four-helix bundle domain to induce membrane localization and necroptotic

cardiolipin, which facilitates its disruption of membrane integrity(17), a model supported by a subsequent paper (18).Here, we show that the MLKL 4HB domain is sufficient to

induce necroptosis and identify several charged residues clus-tered on two faces that are required for this function. Surpris-ingly the polarity of several of these charged residues is notconserved between mouse and human MLKL, and alanine sub-stitution of negatively charged residues on the α4 helix of the4HB domain disrupted function. This finding challenges the im-portance of phospholipid binding to the killing activity of the4HB domain and illustrates that membrane association cannotsolely be attributed to the interaction of poorly conserved basicresidues within the MLKL 4HB domain. Intriguingly, mutationof a second cluster of residues on the 4HB domain did not pre-clude membrane localization or oligomerization but did preventcell death, illustrating that additional function(s) beyond mem-brane translocation are required for the 4HB domain to inducecell death. MLKL oligomerization and membrane translocationwere also inhibited by a small molecule, compound 1, which weidentified on the basis of its affinity for the nucleotide bindingsite of the MLKL pseudokinase domain. These data support amodel for MLKL function whereby the pseudokinase domain ofMLKL holds the 4HB domain in check until phosphorylated byRIPK3, which causes a conformational change in the pseudo-kinase domain to unleash the 4HB domain to oligomerize andassociate with membranes. Activation of MLKL can be thwartedby a small MLKL binding molecule, indicating the feasibility oftargeting the nucleotide binding or “pseudoactive” sites of pseudo-kinases, a hitherto unexplored class of therapeutic targets.

ResultsThe N-Terminal 4HB Domain of MLKL Is the Necroptotic Effector Domain.We sought to define the contributions of MLKL’s componentdomains to necroptotic signaling in light of our recent X-raycrystal structure and functional analysis of full-length mouseMLKL (1). We inducibly expressed a suite of mouse MLKLtruncation constructs (Fig. 1A) in Mlkl−/− and wild-type mousedermal fibroblasts (MDFs). Their capacity to induce cell deathwas determined by propidium iodide (PI) uptake using flowcytometry, in the absence or presence of the necroptotic stimu-lus, TNF (T), Smac mimetic (S), and the pan-caspase inhibitorQ-VD-OPh (Q). TNF initiates signaling upon ligation of TNFReceptor 1 on the cell surface, Smac mimetic inhibits the E3ubiquitin ligase activity of the cellular Inhibitor of APoptosisproteins known to ubiquitylate and prevent the participation ofRIPK1 in apoptotic and necroptotic signaling, and Q-VD-OPhinhibits the activity of caspase-8, thereby preventing the cleavageand inactivation of RIPK1 (19, 20). As we previously showed (1),Mlkl−/− MDFs are sensitive to TS-induced apoptosis but areresistant to TSQ-induced necroptosis, and constructs encodinguntagged full-length mouse MLKL reconstitute TSQ-inducednecroptosis in Mlkl−/− MDFs (Fig. 1B). Surprisingly, taggingthe N terminus of MLKL with the eight-amino-acid FLAG-tagprevented full-length MLKL from reconstituting necroptosis inMlkl−/− MDFs (Fig. 1C), despite being expressed (Fig. S1A).Expression of different C-terminal pseudokinase domain con-structs (Fig. 1 D and E) also failed to reconstitute the necroptoticpathway when overexpressed in Mlkl−/− MDFs. On the contrary,inducible expression of MLKL(124–464), encompassing the braceand pseudokinase domain, inhibited TSQ-stimulated cell death by∼50% in wild-type MDFs compared with the uninduced controls(Fig. 1F).These data pointed to an essential role for the N-terminal

domain of MLKL in necroptotic signaling, while implicatingthe C-terminal pseudokinase domain as a suppressor of MLKL-mediated cell death. Indeed, inducible expression of untaggedMLKL constructs that lacked the pseudokinase domain in eitherMlkl−/− or wild-type MDFs led to constitutive cell death in theabsence of TSQ stimulation (Fig. 1 G and H). The capacity ofMLKL(1–180) to induce cell death was independent of caspaseand RIPK1 kinase activities (Fig. 1G) or the presence of RIPK3

(Fig. 1I). We principally characterized MLKL fragments encom-passing residues 1–180 because our monoclonal MLKL antibody(3H1) recognizes the brace region (Fig. 1A), and like the full-length MLKL, addition of an N-terminal FLAG- or HA-tag pre-vented its killing activity in wild-type and Mlkl−/− cells (Fig. 1 C andJ and Fig. S1 B and C). However, the 4HB domain alone wassufficient to induce cell death in the absence of TSQ stimulationin both Mlkl−/− and wild-type MDFs (Fig. 1 K and L). Theseresults demonstrate that the 4HB domain alone mediates thekilling function of MLKL, and this function is suppressed in thecontext of full-length MLKL by its pseudokinase domain untilactivation by RIPK3-mediated phosphorylation.

Two Charged Clusters on the 4HB Surface Are Essential for MLKL-Induced Cell Death. Having established that the 4HB domain ofMLKL mediates necroptotic cell death, we performed alanine-scanning mutational analysis to define key residues(s) requiredfor this function. We selected residues on the surface of the 4HBdomain, based on our recent mouse MLKL structure (Fig. 1A)(1), and mutated clusters of typically two to three residues to ala-nine (Fig. 2A). We did not mutate residues that participate in thehelical core of the 4HB domain because such mutations easilydisrupt domain folding and/or stability. Mutations were introduced

A B Mlkl-/- MDFs + FLAG-full length MLKL

C

EDwt MDFs + 124-464

FMlkl-/- MDFs + 124-464Mlkl-/- + 179-464-FLAG

Mlkl-/- MDFs + FLAG-1-180J K L

Mlkl-/- MDFs + 1-125 wild-type MDFs + 1-125

HG IRipk3-/- MDFs + 1-180Mlkl-/- MDFs + 1-180 wild-type MDFs + 1-180

Mlkl-/- MDFs + full length MLKL

Fig. 1. The 4HB domain of MLKL is sufficient to induce necroptosis. (A)Schematic representing the different constructs used and the correspondingMLKL structure. (B–L) MDF cell lines derived from wild-type or Mlkl−/− micewere stably infected with the indicated MLKL constructs. MLKL variants wereinduced for 4 h (white bars) or not (black bars), then either left untreated(UT) or treated with the apoptotic stimulus (TS) or necroptotic stimulus (TSQ)for 24 h. Q, Q-VD-OPh; S, Smac-mimetic; T, TNF. PI-permeable cells werequantified using flow cytometry. (G) As above, but in addition, cells werealso treated with N, Necrostatin-1. Data are plotted as the mean ± SEM ofat least two biological replicates each assayed in a minimum of three in-dependent experiments.

Hildebrand et al. PNAS | October 21, 2014 | vol. 111 | no. 42 | 15073

BIOCH

EMISTR

Y

Dow

nloa

ded

by g

uest

on

May

26,

202

0

Page 3: Activation of the pseudokinase MLKL unleashes the four ... · Activation of the pseudokinase MLKL unleashes the four-helix bundle domain to induce membrane localization and necroptotic

into the MLKL(1–180) construct, as expression of these un-tagged constructs could be monitored using the 3H1 antibody.Wild-type and mutant MLKL(1–180) constructs were induciblyexpressed in Mlkl−/− (Fig. 2B and Fig. S2B) and wild-type (Fig.S2 A and B) MDFs, and were deemed to have retained nec-roptotic killing function when the percentage of PI-positive cellswas ≥threefold higher than in uninduced controls. As before,induction of wild-type MLKL(1–180) killed both Mlkl−/− andwild-type MDFs (Figs. 1 G and H and 2B and Fig. S2A), buta subset of the mutant MLKL(1–180) constructs were unable tokill, despite expressing similar levels as wild-type MLKL(1–180)(Fig. S2B). One group of MLKL(1–180) mutants were unable toinduce cell death in either Mlkl−/− or wild-type MDFs: C18S/C24S/C28S, K22A/R30A, R63A/D65A, K80A/K81A, H98A/E99A,E102A/K103A, R105A/D106A, E109A/E110A, and LLLL112–115AAAA (highlighted in red in Fig. 2 A and C). On the otherhand, the Y15A/E16A, E70A/N72A, and E76A/K77A mutationsprevented MLKL(1–180)-induced cell death when expressedin Mlkl−/− MDFs, but not wild-type MDFs. These residues(highlighted in orange in Fig. 2 A and C) are therefore essential

for 4HB domain killing but can induce cell death via a mecha-nism dependent on endogenous MLKL.The mutations that compromise 4HB domain function in cell

death were centered on two clusters positioned on opposingsides of the domain (Fig. 2C): Cluster 1 centered around the α4helix (E102/K103, R105/D106, E109/E110, and L112–L115) andincluded residues from the adjacent α3 helix (R63/D65), andcluster 2 centered around the C-terminal part of the α1 helix, theN-terminal part of the α2 helix, and the connecting α1–α2 loop(Y15/E16, K22/R30, and C18/C24/C28) and includes residuesfrom the α3–α4 loop that was disordered in the MLKL struc-ture (K80A/K81A).

Membrane Localization of MLKL Is Not Sufficient to Induce Cell Death.We sought to establish the localization of MLKL by performingsubcellular fractionation and Blue-Native PAGE of wild-typeMDFs ± TSQ-induced necroptosis. These studies revealed thatMLKL resides in the cytoplasm (C) of a healthy cell in a complexthat migrates similarly to the 146-kDa molecular weight marker(complex I; Fig. 3A) until necroptosis is induced, when it trans-located to the 0.025% digitonin-insoluble, 1% digitonin-solublecell fraction (containing most biological membranes; M) andformed part of a complex that migrated above the 480-kDamolecular weight marker (complex II; Fig. 3A) with concomitantdepletion from the cytoplasm. These complexes, formed by en-dogenous (rather than tagged or overexpressed) MLKL, wereextracted from cells and resolved on Blue-Native PAGE undernondenaturing conditions in the presence of the N-ethyl maleimide,a compound that reacts with free thiols. Previous demonstrationsof MLKL oligomerization in situ have used nonreducing SDS/PAGE (15–18). Although the physiological relevance of theobserved disulfide bonding and the reported oligomer stoichi-ometry to MLKL function remains to be established, all reportsto date (15–18) support the idea that MLKL association with theplasma membrane is an essential step for the induction of nec-roptotic cell death. We tested this hypothesis by monitoring mem-brane translocation of wild-type MLKL(1–180) when expressedin Mlkl−/− MDFs (Fig. 3B). MLKL(1–180) translocated from thecytoplasm to the membrane following induction of proteinexpression, and like full-length MLKL, MLKL(1–180) was in-corporated into a higher molecular weight complex (complex II)by Blue-Native PAGE. Based on migration relative to molecularweight markers in Blue-Native PAGE, we speculated that cyto-plasmic full-length MLKL (Fig. 3A) and MLKL(1–180) (Fig. 3B)formed homotrimers. Analytical ultracentrifugation studies ofrecombinant MLKL(1–169) confirmed that the MLKL 4HBand brace exists as a stable homotrimer in solution with a KD

3-1

of 7.2 ± 0.7 × 10−5 μM2 (Fig. S3 A–C), and like the cytosolicfraction of cells expressing MLKL(1–180) (Fig. 3B), recombinantMLKL(1–169) comigrated with the 66-kDa marker in Blue-Native PAGE (Fig. 3C).Accumulation of MLKL(1–180) at the membrane correlated

with the time course of cell death postinduction inMlkl−/− MDFsin the absence of TSQ stimulation (Fig. S3D). This suggestedthat lack of necroptotic potency among the loss-of-function mutantsmight arise from defective translocation to the membrane fraction.We therefore selected eight MLKL(1–180) constructs for closerexamination. The cluster 1 mutants—R63A/D65A, E102A/K103A,R105A/D106A, E109A/E110A, and LLLL112–115AAAA—showed negligible or no capacity to incorporate into high-molecular-weight complexes in the membrane fraction, despitehigh levels of expression (Fig. 3D). Small amounts of R105A/D106A and E109A/E110A mutants were detected in membranefractions, but notably did not assemble into high molecularcomplexes (complex II; Fig. 3D). Interestingly, in contrast to thecluster 1 mutants, the cluster 2 mutant, Y15A/E16A, and to alesser extent C18S/C24S/C28S and N-terminally FLAG-taggedwild-type MLKL(1–180) were capable of membrane trans-location and formed the high-molecular-weight complexestypical of cell death-inducing complexes, but did not induce celldeath in Mlkl−/− MDFs. Despite observing less translocation of

CN

Cluster 2

A

full le

ngth

MLKL

wild-ty

pe N

TD

K3A/Q

6A/K

9A

Y15A/E

16A

Q17A/K

20A

C18S/C

24S/C

28S

K22A/R

30A

Q29A/N

33A

D55A/D

56A

R63A/D

65A

E66A/K

69A

E70A/N

72A

E76A/K

77A

K80A/K

81A

H83A/K

86A

H98A/E

99A

E102A

/K10

3A

R105A

/D10

6A

E109A

/E11

0A

LLLL

112-1

15AAAA

0

20

40

60

80

100

PI-p

ositi

ve c

ells

(%) Uninduced

InducedB

C

Mlkl-/- MDFs +MLKL(1-180) mutants

1 a a a b b c d c e d d f e f 42M. musculus M D K L G Q I I K L G Q L I Y E Q C E K M K Y C R K Q C Q R L G N R V H G L L Q P LH. sapiens M E N L K H I I T L G Q V I H K R C E E M K Y C K K Q C R R L G H R V L G L I K P LG. gallus M D I I E K V F S I A H A I H S Q F E H V K C C K H Q C Q R L V E R I H I L L E P VX. tropicalis M E I L G N V L E I A Q T I Y N L C D Q A S S N K R Q C S R L K K R I Q I L L M A A

43 g g h h i i j j k k l l mM. musculus Q R L Q A Q G K K N L P D – D I T A A L G R F D E V L K E A N Q Q I E K F S K K S HH. sapiens E M L Q D Q G K R S V P S E K L T T A M N R F K A A L E E A N G E I E K F S N R S NG. gallus R V L Q A Q P S W R I S H – H E E Q M L T K L L Q A L G E A Q K L V T K Y S Q T S WX. tropicalis E K L K K Q P E K S G – – – E L K I V L R E M Q L T L R N A K S W V L K Y S N Q G W

84 m n n o o p p q q r r r r 125M. musculus I W K F V S V G N D K I L F H E V N E K L R D V W E E L L L L L Q V Y H W N T V S DH. sapiens I C R F L T A S Q D K I L F K D V N R K L S D V W K E L S L L L Q V E Q R M P V S PG. gallus I Q K F L S A R S S G E E F V W V N R S L E D I A Q G L S L L L Q A E Q K Q A L L EX. tropicalis W M K I I K A N G I K E E F D L I N D R L K D A A D D I S V M L A M E H R E L L L K

Cluster 1

Fig. 2. Residues required for 4HB killing cluster into two motifs. (A) Align-ment of MLKL orthologs. Groups of mutated residues are indicated by low-ercase letters. Residues in red when mutated to Alanine prevented 4HB killingin both Mlkl−/− and wild-type cells; mutation of residues highlighted in or-ange prevented 4HB killing in Mlkl−/− but not wild-type cells. (B) Three bi-ologically independent MDF cell lines derived from Mlkl−/− mice were stablyinfected with the indicated doxycycline-inducible 4HB MLKL wild-type andmutant constructs and each assayed in two independent experiments. Celllines were induced for 20 h (white bars) or not (black bars) before viabilitywas quantitated. All data are plotted as mean ± SEM. (C) Depiction of the4HB domain (Protein Data Bank ID code 4BTF) (1) drawn using PyMOLsoftware with residues colored according to the scheme in panel A.

15074 | www.pnas.org/cgi/doi/10.1073/pnas.1408987111 Hildebrand et al.

Dow

nloa

ded

by g

uest

on

May

26,

202

0

Page 4: Activation of the pseudokinase MLKL unleashes the four ... · Activation of the pseudokinase MLKL unleashes the four-helix bundle domain to induce membrane localization and necroptotic

the C18S/C24S/C28S than Y15A/E16A mutant to the mem-brane fraction, both mutations are spatially adjacent (forming“cluster 2”) and exhibit deficits in cell death signaling. Thesedata demonstrate that the formation of high-molecular-weight,MLKL-containing complexes in biological membranes alone isinsufficient to initiate cell death. We then examined the cluster1 mutations, R105A/D106A and E109A/E110A, and the cluster2 mutation, Y15A/E16A, in the context of full-length MLKL.Cluster 1 mutations completely compromised the ability of full-length MLKL to reconstitute necroptotic signaling in Mlkl−/−

MDFs and partially antagonized endogenous MLKL to reduceTSQ-induced death in wild-type MDFs (Fig. S4 B–D). How-ever, in the context of full-length MLKL, the cluster 2 mutant,Y15A/E16A, restored TSQ sensitivity toMlkl−/− MDFs (Fig. S4A and D). These data support the idea that cluster 2 is dis-pensable for MLKL to assemble into a high-molecular-weight,membrane-associated complex, but is crucial for orienting proto-mers within the complex or binding to a downstream effector toinduce death, a function that can be complemented by the pseu-dokinase domain in the context of full-length MLKL.

A Small-Molecule Compound That Binds the MLKL Nucleotide BindingSite Retards Membrane Association to Inhibit Necroptosis. Based onthese observations and our recent studies (1, 14), we hypothe-sized that the pseudokinase domain functions as a switch that,until activated, restrains the necroptotic activity of the 4HB

domain. Our data suggest a model whereby release of the 4HBdomain occurs following a conformational change in the pseudo-kinase domain induced by RIPK3 phosphorylation. Previously,using thermal shift assays, we established that the pseudo-kinase domain of MLKL could engage nucleotides in a cation-independent, noncatalytic manner (1, 14, 21), although thebiological significance of nucleotide binding is unclear. To probethe role of nucleotide binding in MLKL function, we screeneda library of 367 small molecules (22) against the recombinantmouse MLKL pseudokinase domain using a thermal stabilityshift assay (21, 23) and identified compound 1 as an MLKLinteractor (Fig. 4 A and B). Subsequent analyses by SurfacePlasmon Resonance (SPR) provided further validation thatcompound 1 bound the MLKL pseudokinase domain (Fig. 4Cand Fig. S5A), yielding a Kd value of 9.3 μM. Saturation transferdifference NMR (STD–NMR) studies inferred that compound 1bound the nucleotide binding site in the MLKL pseudokinasedomain, because compound 1 competed with either ATP orADP for binding to MLKL (Fig. S5B). This idea was furthersupported by thermal shift assays where compound 1 exhibiteddiminished binding to K219M MLKL, a mutant known to pos-sess defective ATP binding compared with wild-type MLKL (Fig.S5C) (1). We next examined the capacity of compound 1 to in-hibit TSQ-induced necroptosis in cells. Compound 1 rescued50% of wild-type MDFs from TSQ-induced necroptosis withan IC50 < 50 nM, with >50-fold greater potency than Nec-1, when1 ng/mL TNF was used (Fig. 4D). However, supraphysiologicalTNF concentrations (100 ng/mL) overwhelmed the ability ofcompound 1 to inhibit necroptosis, leading to an IC50 value of100–500 nM with maximally ∼50% of cells protected fromTSQ-induced death (Fig. S5D). Although compound 1 boundthe MLKL pseudokinase domain with a relatively high Kd invitro, the efficiency of compound 1 as an inhibitor of necroptosisin cells may be attributed to the slow off-rates observed in SPRstudies. This indicates that despite a significant energy penaltyduring the association phase, the resulting complex betweencompound and protein is relatively stable (Fig. 4C). We observedthat compound 1 affected cell viability at high concentrations(above 5 μM; Fig. 4D), presumably due to off-target effects.Consequently, we used 1 μM compound 1 in subsequent experi-ments. Compound 1 has previously been described as a nanomolarinhibitor of the protein kinase, VEGFR2 (24), raising the possibilitythat inhibition of VEGFR2 might block necroptosis. However,sorafenib, a potent VEGFR2, Ret, and c-Kit inhibitor, was unableto inhibit necroptosis in wild-type MDFs (Fig. S5E). Although wecannot exclude the possibility that compound 1 inhibits other targetsin addition to MLKL, the experiments with sorafenib suggest thatinhibition of VEGFR2, Ret, and c-Kit cannot prevent necroptosis.To establish the mechanism by which compound 1 binding to

the MLKL pseudokinase domain inhibited necroptosis, we ex-amined whether compound 1 might prevent phosphorylation ofMLKL by its upstream activator, RIPK3, using in vitro kinase as-says, but neither the catalytic activity of recombinant RIPK3 norRIPK3-mediated phosphorylation of MLKL were inhibited bycompound 1 (Fig. S5F). On the contrary, in the presence of10 μMof compound 1, RIPK3-mediated phosphorylation of MLKLwas enhanced. Mass spectrometry analyses confirmed thatphosphorylation of established activation loop substrate residuesS345 and S347 (1) was enhanced (Fig. S6), consistent with com-pound 1 increasing solvent exposure of the MLKL activation loop.We therefore tested whether compound 1 inhibited the membranelocalization of endogenous MLKL in wild-type MDFs followingTSQ-induced necroptosis. TSQ stimulation of MDFs led to theaccumulation of MLKL in the membrane fraction over a 6-h timecourse, and this was reproducibly retarded in three independentexperiments by preincubation of MDFs with 1 μM of compound 1(Fig. 4E). Consistent with a mode of action targeting the MLKLpseudokinase domain, compound 1 (i) did not protect MDFsfrom MLKL(1–180)-mediated death (Fig. S5G) and (ii) con-ferred no significant protection on the predominantly apoptoticdeath arising from TS stimulation (Fig. S5D). Collectively, these

I

II II

I

A B C

D

Fig. 3. MLKL and the 4HB domain of MLKL form high-molecular-weightcomplexes in biological membranes. (A) Blue-Native PAGE showed that en-dogenous MLKL translocated from cytoplasm (C) to membrane (M) fractionin wild-type MDFs following TSQ treatment. (B) MLKL(1–180) similarlytranslocated to the membranes of Mlkl−/− MDFs following induction. (C)Recombinant MLKL(1–169) resolved by Blue-Native PAGE. (D) Membranecomplex (complex II) formation monitored by Blue-Native PAGE after a 6-hinduction of wild-type MLKL(1–180), N-FLAG–tagged MLKL(1–180), cluster 2mutants (Y15A/E16A and C18S/C24S/C28S), and cluster 1 mutants (R63A/D65A, E102A/K103A, R105A/D105A, E109A/E110A, and LLLL112–115AAAA).Data presented were obtained from three independent experiments per-formed on two biological replicate cell lines.

Hildebrand et al. PNAS | October 21, 2014 | vol. 111 | no. 42 | 15075

BIOCH

EMISTR

Y

Dow

nloa

ded

by g

uest

on

May

26,

202

0

Page 5: Activation of the pseudokinase MLKL unleashes the four ... · Activation of the pseudokinase MLKL unleashes the four-helix bundle domain to induce membrane localization and necroptotic

findings support a model in which binding of an ATP mimetic tothe pseudokinase domain of MLKL can jam the switch mecha-nism, thereby preventing RIPK3-mediated phosphorylation ofMLKL from inducing a conformational change within the pseudo-kinase domain to unleash the necroptotic death effector, the4HB domain (Fig. 4F).

DiscussionAlthough the pseudokinase MLKL has been confirmed as anecroptotic effector downstream of the protein kinase RIPK3, itis unclear how it induces cell death. We show that the 4HBdomain of MLKL is sufficient to induce necroptosis and is ableto oligomerize and translocate to membranes without an ectopicoligomerization domain or any other stimulus. Two clusters ofamino acids on opposing faces of the 4HB domain are requiredfor killing by this domain, and mutations in these clusters affectthe ability of the 4HB domain to either form high-molecular-weight complexes and/or localize to membranes or to inducedeath once incorporated into high-molecular-weight, membrane-associated complexes. Notably, solvent exposure of both 4HBdomain clusters in our recent structure of full-length MLKL (1)is highly suggestive that the crystallized conformation representsan activated form of MLKL. Endogenous MLKL also translocatesto the membrane fraction following a necroptotic stimulus, and

this translocation and ensuing cell death can be inhibited bya small molecule that binds the ATP binding site of MLKL.Several groups have shown that MLKL can oligomerize, but

there is no consensus regarding the stoichiometry of the oligo-mer; Cai et al. reported it contains three protomers (15), whereasothers claim four (16) and possibly six (17) units. Chen et al.showed that the N-terminal domain of MLKL(1–130) was suf-ficient to trigger necroptosis, formed tetramers, and translocatedto lipid rafts in the plasma membrane (16). However, to inducedeath in their hands, forced oligomerization of the 4HB domainwith an inducible dimerization domain was required. In contrast,we show that the MLKL 4HB domain is sufficient to inducedeath by necroptosis and does not require fusion to an exog-enous oligomerization domain. Additionally, our Blue-NativePAGE and analytical ultracentrifugation studies support the ideathat both endogenous MLKL and the N-terminal domain ofMLKL exist as stable homotrimers in the cytoplasm, before theirincorporation into membrane-associated, high-molecular-weightcomplexes and subsequent necroptotic cell death.We were intrigued by the poor conservation of charged resi-

dues in the 4HB domain between orthologs from different species,often with oppositely charged amino acids (Fig. 2A). We thereforechose to replace charged residues, as well as a partially conserved,solvent-exposed hydrophobic stretch (L112LLL115), with alanine,and potential metal coordinating cysteines C18, C24, and C28,which were also partially conserved, with serine. None of thesemutations are expected to disrupt the structure of the 4HBdomain, and accordingly, the expression levels of all mutantswere comparable to wild-type MLKL(1–180). When we testedthese mutants in wild-type and Mlkl−/− cells, we identified threeclasses of mutants: The first class did not affect MLKL func-tion, and these mutants were dispersed over the α1, α2, and α3helices and the α3–α4 loop; the second completely prevented4HB domain killing whether expressed in Mlkl−/− or wild-typecells; and the third class of mutants, located on the α1 and α3helices, presumably retain the ability to interact with endogenousMLKL because they killed wild-type but not Mlkl−/− cells. Loss-of-function mutants clustered into two groups on opposite facesof the 4HB domain, suggesting that the 4HB domain performstwo independent activities required to kill cells. Although thecluster 2 mutants retained the ability to form high-molecularcomplexes (complex II) in membranes, mutations on the op-posite face of the 4HB domain (cluster 1; on the α4 helix)prevented formation of complex II in the membrane fraction.Whether membrane localization is required for MLKL com-plex II formation or complex II formation is required formembrane localization currently remains unclear.It is notable that the polarity of the charged residues that we

mutated is not well conserved among MLKL proteins from dif-ferent species, although the presence of charged residues inthese positions is. For example, mouse MLKL has a glutamate oraspartate in positions 16, 56, and 65, whereas human MLKL hasa lysine in these positions. Human MLKL has glutamates inpositions 20 and 70, whereas mouse MLKL has lysines in thesepositions. This is suggestive of the possibility of charged pairsinteracting between MLKL protomers, although the charge in-version is not conserved in all species. Recent studies suggestedthat MLKL can bind negatively charged phospholipids (17, 18),and this function was attributed to nine basic residues in the 4HBdomain because simultaneous charge reversal of all nine residuescompromised phospholipid binding (18). In contrast, our ob-servation that the requirement for a charged residue, rather thanstrictly a basic residue, argues against charged residues beingimportant for interaction with the phospholipids in the plasmamembrane and does not support the idea that phospholipid, andthus membrane, binding is mediated by these poorly conservedbasic residues via a simple positive–negative charge pairing.In contrast to previous observations (15–17), we observed

by Blue-Native PAGE that endogenous MLKL exists in a cy-toplasmic complex of ∼150 kDa (complex I) in the absence ofexogenous necroptotic stimuli, but is incorporated into a much

A

B E

C

F

D

Fig. 4. Compound 1, a small molecule targeting the nucleotide binding siteof the MLKL pseudokinase domain, retards MLKL membrane translocationand inhibits necroptosis. (A and B) Compound 1 was identified as an MLKLinteractor using a thermal stability shift assay. (C) Compound 1 binding tothe MLKL pseudokinase domain was validated by SPR. Sensorgrams showthe kinetics of compound 1 binding (at a given concentration from 6.25to 200 μM) to MLKL (colored curves) with fit to model overlaid (black curves).x axis, time (s); y axis, response unit (RU) levels. (D) Compound 1 and Nec-1inhibited necroptotic death of wild-type MDFs stimulated with TSQ (1 ng/mLTNF, 500 nM compound A, 10 μM Q-VD-OPh) in a dose-dependent manner.Data shown are the mean ± SEM for three independent experiments. (E)Compound 1 (1 μM) retarded MLKL translocation to the membrane. Cyto-plasmic and membrane fraction purity and protein abundance are illustratedby control blots for GAPDH and VDAC1. Data are representative of threeindependent repeats. (F) A model for MLKL activation and the mechanism ofaction of compound 1.

15076 | www.pnas.org/cgi/doi/10.1073/pnas.1408987111 Hildebrand et al.

Dow

nloa

ded

by g

uest

on

May

26,

202

0

Page 6: Activation of the pseudokinase MLKL unleashes the four ... · Activation of the pseudokinase MLKL unleashes the four-helix bundle domain to induce membrane localization and necroptotic

larger complex (complex II) in membranes following treatmentwith necroptotic stimuli. A similar phenomenon was observedupon induction of MLKL(1–180) expression in Mlkl−/− dermalfibroblasts, indicating a correspondence between assembly of anMLKL-containing, high-molecular-weight complex in biologicalmembranes and necroptotic signaling. The composition of thehigh-molecular-weight, membrane-associated complex (complexII), however, is currently unclear and remains a subject ofongoing investigation.Having established the importance of pseudoactive site in-

tegrity for the MLKL pseudokinase domain to function as anegative regulator of necroptosis (Fig. 1F and ref. 1), we hypothe-sized that small molecules that bind the nucleotide binding site ofMLKL might antagonize necroptosis. Initially, we identifiedan ATP mimetic (termed compound 1) that bound recombi-nant mouse MLKL pseudokinase domain, which we subsequentlyshowed inhibited TSQ-induced death of MDFs by delaying MLKLtranslocation to the membrane. Compound 1 therefore not onlyrepresents a valuable reagent to inhibit necroptosis to aid discoveryin this field but, more broadly, provides an important proof-of-principle that targeting catalytically dead pseudoenzymesrepresents a feasible, emerging therapeutic avenue.

Materials and MethodsExpression Constructs.MouseMLKL cDNA (encoding residues 1–464), PCR-derivedmutants, or a library of MLKL(1–180) mutants (DNA2.0, CA) were cloned into thedoxycycline-inducible, puromycin selectable vector, pF TRE3G PGK puro, as pre-viously (1, 11, 25). Lentiviruses were generated in HEK293T cells (26) before in-fection of target cells and selection/maintenance in 5 μg/mL puromycin.

Reagents and Antibodies. Recombinant hTNF-Fc (27), rat anti-mouseMLKL monoclonal antibody (clone 3H1; available from Millipore, cat. no.MABC604) (1) and compound 1 (available from Synkinase, Australia) wereproduced in-house. Smac mimetic, compound A, was described pre-viously (26). Q-VD-OPh was from R&D Systems. Anti–β-actin and anti-FLAG (M2) antibodies were purchased from Sigma Aldrich; anti-VDAC1(AB10527) was purchased from Millipore; anti-GAPDH was purchasedfrom Cell Signaling Technologies; and HRP-conjugated secondary anti-bodies were from GE Healthcare or Jackson Immunoresearch, with theECL reagent from Millipore.

Cell Lines and Cell Death Assays. Three biologically independent MDF cell lineswere generated from wild-type, Mlkl−/−, and Ripk3−/− mice and cell deathassays performed as described previously (1). Cells were attached over 4 h inthe presence of 10 ng/mL doxycycline before addition of death stimuli(Fig. 1), or incubated with 10 ng/mL doxycycline for 20 h (Fig. 2 and Fig. S2),to induce protein expression from stably transfected constructs and PI-positivecells quantified by flow cytometry.

Fractionation and Blue-Native PAGE. MDFs were stimulated with TSQ (WTMDFs) or doxycycline [MLKL(1–180)] and permeabilized in buffer containing0.025% digitonin. Cytosolic and crude membrane fractions were furthersolubilized in 1% digitonin, resolved by Bis·Tris Native PAGE, and immuno-probed for MLKL.

Recombinant Protein Expression and Purification. Recombinant mouse MLKLpseudokinase domain (residues 179–464) and mRIPK3 kinase domain wereexpressed and purified from Sf21 cells as described previously (1, 28). MLKL(1–169) was expressed and purified from Escherichia coli using an estab-lished strategy (29).

Thermal Shift Assays to Screen for Small-Molecule Interactors. Thermal shiftassays were performed on 2.6 μM MLKL(179–464) as described previously (1, 14,21). ATP was added at 0.2 mM; small molecules from the Published Kinase In-hibitor Set (22) (kindly provided by GSK) were added at 40 μM of final con-centration. Shown data are representative of three independent experiments.

SPR Binding Experiments. The kinetics of compound 1 (6.25–200 μM) bindingto MLKL(179–464) was determined by SPR (Biacore T200, GE Healthcare)following protein capture via Ni2+/NTA chelation. The Kd was determinedfrom a global fit of data to a two-state kinetic interaction model.

ACKNOWLEDGMENTS. We thank staff in the Walter and Eliza Hall InstituteBioservices Facility; Drs. Robert Ninnis, Grant Dewson, and James Vince foradvice; Vishva Dixit for Ripk3−/− mice; and Toru Okamoto for developinginducible lentiviral vectors. This work was supported by National Healthand Medical Research Council (NHMRC) Grants 1057905, 1067289,1046984, 1025594, and 461221 and fellowships (to J.M.H., W.S.A., and J.S.);a Victorian International Research Scholarship (to M.C.T.); a C. R. Roper Fel-lowship (to R.C.J.D.); and Australian Research Council fellowships (to J.J.B.,P.E.C., and J.M.M.). Additional support was obtained from the AustralianCancer Research Fund, Victorian State Government Operational Infrastruc-ture Support, and NHMRC Independent Research Institute InfrastructureSupport Grant 361646.

1. Murphy JM, et al. (2013) The pseudokinase MLKL mediates necroptosis via a molec-ular switch mechanism. Immunity 39(3):443–453.

2. Newton K, et al. (2014) Activity of protein kinase RIPK3 determines whether cells dieby necroptosis or apoptosis. Science 343(6177):1357–1360.

3. Wu J, et al. (2013) Mlkl knockout mice demonstrate the indispensable role of Mlkl innecroptosis. Cell Res 23(8):994–1006.

4. Cho YS, et al. (2009) Phosphorylation-driven assembly of the RIP1-RIP3 complex reg-ulates programmed necrosis and virus-induced inflammation. Cell 137(6):1112–1123.

5. He S, et al. (2009) Receptor interacting protein kinase-3 determines cellular necroticresponse to TNF-alpha. Cell 137(6):1100–1111.

6. Rickard JA, et al. (2014) RIPK1 regulates RIPK3-MLKL-driven systemic inflammationand emergency hematopoiesis. Cell 157(5):1175–1188.

7. Dillon CP, et al. (2014) RIPK1 blocks early postnatal lethality mediated by caspase-8and RIPK3. Cell 157(5):1189–1202.

8. Sun L, et al. (2012) Mixed lineage kinase domain-like protein mediates necrosis sig-naling downstream of RIP3 kinase. Cell 148(1-2):213–227.

9. Zhao J, et al. (2012) Mixed lineage kinase domain-like is a key receptor interactingprotein 3 downstream component of TNF-induced necrosis. Proc Natl Acad Sci USA109(14):5322–5327.

10. Wang Z, Jiang H, Chen S, Du F, Wang X (2012) The mitochondrial phosphatasePGAM5 functions at the convergence point of multiple necrotic death pathways. Cell148(1-2):228–243.

11. Moujalled DM, Cook WD, Murphy JM, Vaux DL (2014) Necroptosis induced by RIPK3requires MLKL but not Drp1. Cell Death Dis 5:e1086.

12. Remijsen Q, et al. (2014) Depletion of RIPK3 or MLKL blocks TNF-driven necroptosisand switches towards a delayed RIPK1 kinase-dependent apoptosis. Cell Death Dis5:e1004.

13. Tait SW, et al. (2013) Widespread mitochondrial depletion via mitophagy does notcompromise necroptosis. Cell Reports 5(4):878–885.

14. Murphy JM, et al. (2014) Insights into the evolution of divergent nucleotide-bindingmechanisms among pseudokinases revealed by crystal structures of human andmouse MLKL. Biochem J 457(3):369–377.

15. Cai Z, et al. (2014) Plasma membrane translocation of trimerized MLKL protein isrequired for TNF-induced necroptosis. Nat Cell Biol 16(1):55–65.

16. Chen X, et al. (2014) Translocation of mixed lineage kinase domain-like protein to

plasma membrane leads to necrotic cell death. Cell Res 24(1):105–121.17. Wang H, et al. (2014) Mixed lineage kinase domain-like protein MLKL causes necrotic

membrane disruption upon phosphorylation by RIP3. Mol Cell 54(1):133–146.18. Dondelinger Y, et al. (2014) MLKL compromises plasma membrane integrity by

binding to phosphatidylinositol phosphates. Cell Reports 7(4):971–981.19. Khan N, Lawlor KE, Murphy JM, Vince JE (2014) More to life than death: Molecular

determinants of necroptotic and non-necroptotic RIP3 kinase signaling. Curr Opin

Immunol 26:76–89.20. Murphy JM, Silke J (2014) Ars Moriendi; the art of dying well—New insights into the

molecular pathways of necroptotic cell death. EMBO Rep 15(2):155–164.21. Murphy JM, et al. (2014) A robust methodology to subclassify pseudokinases based on

their nucleotide-binding properties. Biochem J 457(2):323–334.22. Drewry DH, Willson TM, Zuercher WJ (2014) Seeding collaborations to advance kinase

science with the GSK Published Kinase Inhibitor Set (PKIS). Curr Top Med Chem 14(3):

340–342.23. Lucet IS, Babon JJ, Murphy JM (2013) Techniques to examine nucleotide binding by

pseudokinases. Biochem Soc Trans 41(4):975–980.24. Sammond DM, et al. (2005) Discovery of a novel and potent series of dianilinopyr-

imidineurea and urea isostere inhibitors of VEGFR2 tyrosine kinase. Bioorg Med Chem

Lett 15(15):3519–3523.25. Moujalled DM, et al. (2013) TNF can activate RIPK3 and cause programmed necrosis in

the absence of RIPK1. Cell Death Dis 4:e465.26. Vince JE, et al. (2007) IAP antagonists target cIAP1 to induce TNFalpha-dependent

apoptosis. Cell 131(4):682–693.27. Bossen C, et al. (2006) Interactions of tumor necrosis factor (TNF) and TNF receptor

family members in the mouse and human. J Biol Chem 281(20):13964–13971.28. Cook WD, et al. (2014) RIPK1- and RIPK3-induced cell death mode is determined by

target availability. Cell Death Differ 21(10):1600–1612.29. Hercus TR, et al. (2013) High yield production of a soluble human interleukin-3 variant

from E. coli with wild-type bioactivity and improved radiolabeling properties.

PLoS ONE 8(8):e74376.

Hildebrand et al. PNAS | October 21, 2014 | vol. 111 | no. 42 | 15077

BIOCH

EMISTR

Y

Dow

nloa

ded

by g

uest

on

May

26,

202

0