throbber
Dimethyl fumarate blocks pro-inflammatory cytokine production via inhi...
`
`https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4976367/
`
`Sci Rep. 2016; 6: 31159.
`
`Published online 2016 Aug 8. doi: 10.1038/srep31159
`
`PMCID: PMC4976367
`
`Dimethyl fumarate blocks pro-inflammatory cytokine production via
`inhibition of TLR induced M1 and K63 ubiquitin chain formation
`
`1
`1,2
`3
`3
`3
`Victoria A. McGuire, Tamara Ruiz-Zorrilla Diez, Christoph H. Emmerich, Sam Strickson, Maria Stella Ritorto, Ruhcha V.
`1
`1
`1
`3
`4
`1
`4
`Sutavani, Anne Weiβ, Kirsty F. Houslay, Axel Knebel, Paul J. Meakin,
`Iain R. Phair, Michael L. J. Ashford, Matthias
`3
`a,1
`Trost, and J. Simon C. Arthur
`
`Division of Cell Signaling and Immunology, School of Life Sciences, Wellcome Trust Building, University of Dundee, Dow St, Dundee, DD1 5EH, UK
`
`Department of Chemistry and Biochemistry, Faculty of Pharmacy, CEU San Pablo University, Urbanización Montepríncipe, 28668 Madrid, Spain
`
`MRC Protein Phosphorylation and ubiquitylation Unit, School of Life Sciences, Sir James Black Centre, University of Dundee, Dow St, Dundee, DD1
`
`1 2 3
`
`5EH, UK
`4
`
`Cardiovascular and Diabetes Medicine, Medical Research Institute, School of Medicine, University of Dundee, Ninewells Hospital, Dundee, DD1 9SY,
`
`UK
`a
`
`Email: j.s.c.arthur@dundee.ac.uk
`
`Received 2015 Oct 19; Accepted 2016 Jul 15.
`
`Copyright © 2016, The Author(s)
`
`This work is licensed under a Creative Commons Attribution 4.0 International License. The images or other third party material in this article are
`
`included in the article’s Creative Commons license, unless indicated otherwise in the credit line; if the material is not included under the Creative
`
`Commons license, users will need to obtain permission from the license holder to reproduce the material. To view a copy of this license, visit
`
`http://creativecommons.org/licenses/by/4.0/
`
`Abstract
`
`Go to:
`
`Dimethyl fumarate (DMF) possesses anti-inflammatory properties and is approved for the treatment of psoriasis
`
`and multiple sclerosis. While clinically effective, its molecular target has remained elusive - although it is known
`
`to activate anti-oxidant pathways. We find that DMF inhibits pro-inflammatory cytokine production in response
`
`to TLR agonists independently of the Nrf2-Keap1 anti-oxidant pathway. Instead we show that DMF can inhibit
`
`the E2 conjugating enzymes involved in K63 and M1 polyubiquitin chain formation both in vitro and in cells. The
`
`formation of K63 and M1 chains is required to link TLR activation to downstream signaling, and consistent with
`
`the block in K63 and/or M1 chain formation, DMF inhibits NFκB and ERK1/2 activation, resulting in a loss of
`
`pro-inflammatory cytokine production. Together these results reveal a new molecular target for DMF and show
`
`that a clinically approved drug inhibits M1 and K63 chain formation in TLR induced signaling complexes.
`
`Selective targeting of E2s may therefore be a viable strategy for autoimmunity.
`
`Autoimmune disorders represent a diverse range of conditions that remain challenging to treat. The advent of
`
`1
`biological drugs, such as anti-TNF agents, provided a significant advance in the treatment of these conditions ,
`
`however they have the disadvantages of not being orally available and that a proportion of patients do not
`
`respond. The development of new orally available small molecule drugs for autoimmunity is therefore desirable.
`
`Several breakthroughs in this area have recently been made, such as the development of Jak inhibitors and S1P
`2,3,4
`receptor modulating agents, which illustrate the potential of this approach
`.
`
`Dimethyl fumarate (DMF) is a methyl ester known to have immuno-modulatory properties. In combination with
`
`1 of 15
`
`10/18/2016 5:39 PM
`
`Biogen Exhibit 2387
`Coalition v. Biogen
`IPR2015-01993
`
`Page 1 of 15
`
`

`
`Dimethyl fumarate blocks pro-inflammatory cytokine production via inhi...
`
`https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4976367/
`
`5
`other fumaric acid esters, DMF has been in use for many years as a treatment for moderate and severe psoriasis .
`6
`The first report of its use was in 1959 , although it did not gain widespread acceptance until some time later
`7
`following the publication of the first clinical trials demonstrating its efficacy in 1990 . Subsequently, DMF in
`8,9
`.
`
`combination with three salts of ethylhydrogenfumarate was licensed for use in psoriasis in Germany in 1994
`10
`.
`
`More recently, a slow release formulation of DMF has been approved for the treatment of multiple sclerosis
`
`The molecular target of DMF that accounts for its ability to modulate the immune system has been elusive.
`
`Amongst the possible explanations for its action, DMF has been shown to reduce T cell numbers, inhibit NFκB
`11,12
`). In addition, DMF has been found to
`
`mediated transcription and activate the Nrf2 pathway (reviewed in
`
`modulate cytokine production in a number of immune cell types: cytokine production is regulated by several
`
`intracellular signaling systems including NFκB and the ERK1/2 and p38 MAPK pathways, and DMF has been
`
`suggested to modulate these pathways. For example, DMF has been shown to prevent the induction of NFκB
`
`dependent transcription in LPS stimulated dendritic cells as well as TNF stimulated Human Umbilical Vein
`13,14,15
`.
`
`Endothelial Cells (HUVEC) or airway smooth muscle cells (ASMC)
`
`decrease ERK1/2 activation in cells, others have found it to have no effect
`
`The reported effects of DMF on MAPK signaling are less clear. While some studies have shown that DMF could
`14,16,17
`. For p38, DMF has been
`14,18
`. MAPKs can,
`
`reported to either have no effect on activation or to result in an increase in p38 phosphorylation
`
`in part, mediate their cellular effects via the activation of downstream kinases. For example, p38α activates the
`19
`downstream kinases MK2 and MK3 to promote the production of TNF . In addition, p38α can also activate the
`20
`20
`kinases MSK1 and MSK2 . These two kinases, which can also be activated by ERK1/2 , have been found to
`
`have anti-inflammatory functions in macrophages and are required for the maximal induction of IL-10 by
`21,22
`23
`. The ERK1/2 pathway can also activate RSK , however the role that this
`
`macrophages and dendritic cells
`
`activation of both MSKs and RSKs
`
`kinase plays in the regulation of cytokine production is less well established. DMF has been shown to affect the
`14,16,17
`. For instance, in keratinocytes DMF selectively blocked MSK1
`16
`phosphorylation but not ERK1/2 or p38α activation in response to IL-1 stimulation . Similarly DMF also
`
`blocked MSK1 and RSK activation in MIF (Macrophage Inhibitory Factor) stimulated keratinocytes and
`17
`24
`prevented the induction of Cox2 , a known MSK target gene . DMF has also been reported to inhibit MSK1
`
`phosphorylation in LPS stimulated dendritic cells, however in contrast to the data in keratinocytes, in dendritic
`14
`cells DMF was able to reduce LPS induced ERK1/2, although not p38 or JNK, phosphorylation .
`
`In this study we examine the mechanism by which DMF blocks cytokine induction in primary macrophages and
`
`demonstrate that it affects signaling by inhibiting the formation of M1/K63 hybrid polyubiquitin chains.
`
`Results
`
`Go to:
`
`DMF inhibits the transcription of cytokines independently of Nrf2
`
`To test the ability of DMF to block cytokine production in response to TLR agonists, BMDMs were incubated
`
`with various concentrations of DMF for 4 h (Fig. 1A). The cells were then stimulated with the TLR4 agonist LPS
`
`for a further 8 h and cytokine release determined. LPS promoted the secretion of TNF, IL-6, IL-10, IL-13 and
`
`GM-CSF; this was blocked by 50 µM DMF (Fig. 1A). To ensure this was not due to a loss of cell viability, cells
`
`were incubated with 50 µM DMF and viability determined by FACS. DMF did cause some cell death that
`
`increased over time, however the majority of cells were still alive following 12 h of DMF treatment (Fig. 1B).
`
`Cells were then treated with or without DMF in the presence of brefeldin A and monensin to block cytokine
`
`secretion. TNF levels were then measured at a single cell level by flow cytrometry following gating on the live
`
`cell population. This showed that DMF blocked LPS stimulated TNF production in the live cells (Fig. 1C). In line
`
`with the loss of cytokine secretion (Fig. 1A), DMF also repressed the induction of various cytokine mRNAs,
`
`including TNF, IL-6, IL-10, GM-CSF, IL-12p40, IL-23p19 and IFNβ, in response to 1 h of LPS stimulation (
`
`Fig. 2). DMF also suppressed the induction of IκBα mRNA, an established NFκB target gene.
`
`2 of 15
`
`10/18/2016 5:39 PM
`
`Page 2 of 15
`
`

`
`Dimethyl fumarate blocks pro-inflammatory cytokine production via inhi...
`
`https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4976367/
`
`Figure 1
`
`Inhibition of LPS stimulated cytokine induction by DMF.
`
`Figure 2
`
`DMF inhibits LPS induced gene transcription.
`
`DMF has previously been suggested to act via targeting cysteine residues in Keap1 and activating the Nrf2
`25,26,27
`anti-oxidant pathway
`. To test the potential involvement of this pathway in the regulation of cytokine
`
`transcription, Nrf2−/− BMDMs were stimulated with LPS in the presence or absence of DMF and cytokine
`28,29
`. While LPS
`
`mRNA levels measured at 1 h. Nrf2 regulates the transcription of several genes including HO-1
`
`alone did not induce the mRNA for HO-1, this mRNA was induced in DMF treated wild type but not Nrf2
`
`knockout BMDMs, indicating that DMF was able to activate Nrf2 in macrophages (Fig. 3). Nrf2 knockout did not
`
`affect the induction of TNF, IL-6, IL-12p40, IL-23p19 or IκBα mRNAs in response to LPS relative to wild type
`
`cells. There was a small but significant increase (p < 0.05, Students unpaired two tailed t-test) in LPS stimulated
`
`IL-10 and GM-CSF mRNA induction in Nrf2 knockout cells compared to wild type BMDMs (Fig. 3). This
`
`increase in IL-10 mRNA induction is consistent with a recent report showing increased IL-10 production in Nrf2
`30
`knockout dendritic cells
`
`. DMF was able to inhibit cytokine and IκBα mRNA induction in response to LPS in
`
`both wild type and Nrf2−/− cells (Fig. 3). These results suggest that the effects of DMF on LPS induced cytokine
`
`induction are largely independent of any effects on the Nrf2 pathway.
`
`Figure 3
`
`Effect of DMF on LPS stimulated mRNA induction in Nrf2 knockout
`
`BMDMs.
`
`DMF has been proposed to inhibit the action of MSK1 in dendritic cells
`
`14
`. However, the reduced cytokine
`
`production caused by DMF in Fig. 1 is inconsistent with the decreased IL-10 production but increased
`22
`pro-inflammatory cytokine production previously reported in MSK1/2 double knockout macrophages
`
`,
`
`suggesting that DMF must have MSK independent effects. To confirm this, wild type and MSK1/2 knockout
`
`BMDCs were tested. As expected MSK1/2 knockout BMDCs exhibited a lower induction of the MSK1/2 target
`
`gene IL-10 relative to wild type cells (Fig. 4). Induction of IL-12p35 mRNA was increased in the MSK1/2
`
`knockout BMDCs while IL-6, IL-12p40 and IκBα mRNA induction was not greatly changed. As in macrophages,
`
`DMF blocked the LPS induced expression of both IκBα and the cytokine mRNAs tested. This inhibition was
`
`comparable in wild type and MSK1/2 knockout cells (Fig. 4).
`
`Figure 4
`
`Effect of DMF on LPS stimulated mRNA induction in MSK1/2 knockout BMDCs.
`
`DMF inhibits NFκB and ERK1/2 activation in response to TLR agonists
`
`3 of 15
`
`10/18/2016 5:39 PM
`
`Page 3 of 15
`
`

`
`Dimethyl fumarate blocks pro-inflammatory cytokine production via inhi...
`
`https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4976367/
`
`TLR dependent transcription and secretion of cytokines is regulated by the NFκB as well as the ERK1/2 and p38
`31
`
`. The ability of DMF to block the induction of multiple LPS induced genes (Figs 1
`
`MAPK signaling pathways
`
`and 2) suggested that it might have a suppressive effect on both MAPK and NFκB activation in response to the
`
`TLR4 agonist LPS. TLR4 signals via both MyD88 and Trif dependent pathways, which are thought to converge
`31,32,33,34
`. Tak1 in turn activates a complex of IKKβ, IKKα and NEMO. IKKβ then
`
`on the activation of Tak1
`
`activates the classical NFκB pathway via the phosphorylation of IκBα. In addition IKKβ also phosphorylates
`
`p105, and this is required for the activation of Tpl2, which is then able to activate the ERK1/2 pathway. Tak1 also
`31,32,33,34
`.
`
`directly activates the MKKs required for the activation of the p38 and JNK pathways (Fig. 5A)
`
`Figure 5
`
`DMF inhibits multiple signals downstream of TLR4.
`
`DMF was able to inhibit the activation of the classical NFκB pathway, as 50 µM of DMF was sufficient to block
`
`the degradation of IκBα in response to 30 min stimulation with LPS (Fig. 5B). In addition, 50 µM DMF blocked
`35
`the phosphorylation of IKKβ on Ser177 and 181 (Fig. 5B), sites that correlate with its activation . In agreement
`
`with the loss of IKKβ activity, DMF also blocked the phosphorylation of the IKKβ substrate p105 and inhibited
`
`the activation of ERK1/2, as judged by phosphorylation on its TXY activation motif. DMF treatment also resulted
`
`in a partial inhibition of JNK activation, that was maximal at 75 µM DMF. In contrast, DMF did not inhibit the
`
`activation of p38 at any of the concentrations tested. In the MyD88 pathway, IRAKs are involved in propagating
`
`the signal from MyD88 to Traf6 and then Tak1. During this process IRAK1 becomes modified with K63
`
`polyubiquitin chains, resulting in the disappearance of the IRAK1 band at its predicted molecular weight on
`36,37
`. Interestingly,
`
`immunoblots that can be reversed by treatment of the lysates with deubiquitinating enzymes
`
`the loss of the IRAK1 band was partially reversed by 25 µM DMF and completely reversed by 50 µM DMF
`
`suggesting that DMF may interfere with IRAK1 ubiquitination (Fig. 5B).
`
`To examine this process in more detail, cells were incubated in 50 µM DMF and then a time course of LPS
`
`stimulation carried out (Fig. 5C). As in the previous experiment, DMF inhibited the loss of IRAK1 and IκBα,
`
`blocked the phosphorylation of IKKβ and p105 and greatly reduced the phosphorylation of ERK1/2. The effect on
`
`JNK phosphorylation was more complex. DMF reduced the phosphorylation of JNK at 30 min. In the absence of
`
`DMF, JNK phosphorylation was transient and not observed at 60 or 90 min. In contrast, JNK phosphorylation was
`
`still observed at 60 and 90 min of LPS stimulation in the presence of DMF.
`
`LPS has the ability to signal via both the MyD88 and Trif adaptors. To determine if the effects of DMF on
`
`signaling were common to pathways downstream of both adaptors, cells were stimulated with either Pam CSK
`3
`4
`or poly(I:C). Pam CSK stimulates TLR1/2 and acts via MyD88 and not Trif. The effects of DMF on Pam CSK
`4
`3
`3
`induced signaling mirrored the results seen with LPS (Fig. 5C). Poly(I:C) activates TLR3 and this utilizes Trif
`
`4
`
`and not MyD88 for downstream signaling. Consistent with this poly(I:C) did not strongly promote the
`
`ubiquitination of IRAK1. Poly(I:C) was able to induce both IKKβ and p105 phosphorylation and this was blocked
`
`by DMF. In addition DMF blocked poly(I:C) induced ERK1/2 activation, however, as for LPS and Pam CSK
`3
`4
`there was little effect on p38 activation (Fig. 5C). Poly(I:C) was only a weak activator of JNK, and this activation
`
`was slightly increased in the presence of DMF.
`
`Unexpectedly in these experiments, incubation in DMF alone induced the phosphorylation of p38 and, to a lesser
`
`extent, JNK in the absence of any other stimuli (Fig. 5C, lane 5). To examine this further a time course of DMF
`
`treatment alone was carried out. DMF treatment in the absence of any additional TLR stimulation did not affect
`
`4 of 15
`
`10/18/2016 5:39 PM
`
`Page 4 of 15
`
`

`
`Dimethyl fumarate blocks pro-inflammatory cytokine production via inhi...
`
`https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4976367/
`
`IRAK1 ubiquitination or p105 phosphorylation (Fig. 6A). DMF treatment did however induce phosphorylation of
`
`MAPK signaling in cells
`
`p38, and to a lesser extent JNK, over time. Tyrosine phosphatase inhibitors have previously been found to activate
`38
`
`. Tyrosine phosphatases possess an active site cysteine and may therefore be a target
`39
`for electrophilic drugs such as DMF . Treatment with the tyrosine phosphatase inhibitor pervanadate induced the
`
`phosphorylation of p38 and JNK although, in contrast to DMF, pervanadate also strongly induced ERK1/2
`
`phosphorylation (Fig. 6A). As would be expected pervanadate strongly induced global tyrosine phosphorylation
`
`levels. In contrast, DMF had little effect on global tyrosine phosphorylation levels indicating that it does not act as
`
`a general tyrosine phosphatase inhibitor in cells (Fig. 6B). MAPKs can be dephosphorylated by DUSPs (Dual
`
`Specificity Phosphatases) and inhibition of these enzymes by DMF may account for the p38 and JNK activation.
`
`Inhibition of the DUSP would be expected to be specific for the MAPK and not affect the upstream MKK. DMF
`
`was found to increase the phosphorylation of MKK3 and 6 suggesting that its effects on p38 and JNK
`
`phosphorylation were independent of DUSP inhibition (Fig. 6A).
`
`Figure 6
`
`DMF can activate p38 MAPK and can covalently modify its targets in
`
`the cell.
`
`DMF has the ability to covalently modify cysteine residues in its targets, as is proposed for Keap1. We assessed
`
`whether DMF inhibition remained following washout of DMF from the cells. BMDMs were treated for 4 h with
`
`50 µM DMF and then either directly stimulated with LPS or washed extensively and incubated in media without
`
`DMF for various times. This showed that signaling was still inhibited by DMF 4 h after DMF being washed off
`
`the cells, but that inhibition was lost by 16 h (Fig. 6C). This slow loss of inhibition would be consistent with DMF
`
`covalently modifying its target in cells.
`
`In vivo DMF can be converted to MMF. We therefore checked if MMF was able to mimic the effects of DMF on
`
`cell signaling. Unlike DMF, MMF did not inhibit LPS induced ERK1/2 or p105 phosphorylation, or the
`
`ubiquitination of IRAK1 (Supplementary Figure 1).
`
`DMF inhibits E2 conjugating enzymes in vitro
`
`Together these results suggest that DMF may act at an upstream point in the TLR signaling cascade. The
`
`activation of Tak1 and the recruitment of the IKK complex involves the formation of K63/M1 hybrid
`
`polyubiquitin chains which can be added to several proteins in the MyD88 signaling cascade including MyD88
`34,40
`. The formation of polyubiquitin chains requires a cascade of three enzymes; an E1 activating
`
`and IRAKs
`
`enzyme, an E2 conjugating enzyme and an E3 ligase to mediate the transfer of the ubiquitin to the substrate. E2
`
`enzymes use a cysteine in their active site to covalently couple to ubiquitin. DMF is an electrophilic compound
`
`and therefore has the potential to react with the SH group in cysteines (Fig. 7A). We therefore investigated if
`
`M1 chains
`
`DMF could inhibit Ubc13 or UbcH7, the major E2 enzymes proposed to be involved in the formation of K63 and
`41,42,43,44,45
`. In vitro, addition of the E1 UBE1, ubiquitin and Mg-ATP to an E2 is able to promote the
`45
`.
`
`loading of ubiquitin onto the E2, which can be resolved as a mobility shift on SDS polyacrylamide gels
`
`Addition of DMF to this reaction inhibited the loading of ubiquitin onto Ubc13 with an IC50 of between 10 and
`
`20 µM (Fig. 7B). In parallel assays, UbcH7 was more strongly inhibited by DMF with an IC50 of less than 10 µM
`
`and complete inhibition by 50 µM (Fig. 7B). If DMF were to inhibit Ubc13 or UbcH7 via covalent modification
`
`of a cysteine, the molecular mass of the E2 should be increased following DMF treatment. MALDI-TOF mass
`
`spectrometry was therefore used to determine the molecular mass of recombinant Ubc13 and UbcH7 before and
`
`after treatment with DMF. The recombinant Ubc13 has a theoretical mono-isotopic mass of 17295 Da, which
`
`corresponded well with the observed mass of 17301 Da. For Ubc13, an increase in the molecular mass of 144 Da
`
`was seen following incubation in DMF, which would be consistent with the labeling of a cysteine residue in the
`
`5 of 15
`
`10/18/2016 5:39 PM
`
`Page 5 of 15
`
`

`
`Dimethyl fumarate blocks pro-inflammatory cytokine production via inhi...
`
`https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4976367/
`
`protein with DMF via a Michael addition reaction (Fig. 7A). For unmodified UbcH7 a mass of 18157 Da was
`
`observed relative to a theoretical mono-isotopic mass of 18149 Da. Following DMF treatment of UbcH7 3 peaks
`
`were obtained. The major peaks indicated an addition of 144 and 289 Da, which would correspond to addition of
`
`1 or 2 DMF molecules respectively. A minor peak was also obtained with a mass increase of 432 Da, which
`
`would suggest addition of 3 molecules of DMF (Fig. 7C). Of note, Ubc13 only contains the active site cysteine
`
`whereas UbcH7 also contains 2 further cysteines in addition to its reactive site residue. To further test the ability
`
`of 50 or 100 µM DMF to block E2 activity, it was tested against a panel of 24 different E2 enzymes (Fig. 7D).
`
`This showed that while DMF could inhibit all the E2’s tested to some extent, it did exhibit some selectivity with
`
`UbcH7, Ube2W, Ube2T, Ube2S and Ube2R1 being most strongly inhibited.
`
`Figure 7
`
`DMF can inhibit E2 conjugating enzymes in vitro.
`
`DMF blocks the formation of M1 and M1/K63 polyubuititin chains downstream of TLR signaling
`
`Previous studies have indicated that TLR agonists can stimulate the formation of K63/M1 hybrid polyubiquitin
`40,46
`. As, in vitro, DMF inhibited Ubc13 and UbcH7, the E2 conjugating enzymes proposed to be involved
`
`chains
`
`in the formation K63 and M1 chains respectively, its ability to block formation of M1 and K63 chains in cells was
`
`determined. M1-Ub chains were pulled down from RAW264.7 macrophage cell lysates using Halo-tagged NEMO
`40
`. This approach would be expected to pull
`
`beads, which shows >50 fold selectivity for M1 relative to K63 chains
`
`down M1-Ub chains, complexes containing both M1 and K63 chains as well as M1/K63 hybrid chains that are
`40,46
`. As seen in primary macrophages following TLR
`
`proposed to be formed downstream of TLR signaling
`
`stimulation, DMF inhibited the ubiquitination of IRAK1 and the phosphorylation of ERK1/2, p105 and IKK in
`
`RAW264.7 cells (Fig. 8A). Halo-NEMO pulldowns were blotted for M1 or K63 polyubiquitin chains on IRAK1
`
`and IRAK2. LPS increased the amount of M1 and M1/K63 hybrid chains in the pulldowns, and this increase was
`
`inhibited by pretreatment of the cells with DMF (Fig. 8B). Polyubiquitinated IRAK1 and IRAK2 could also be
`
`seen in the pulldowns from LPS stimulated but not untreated cells, and this was blocked by pretreatment with
`
`DMF (Fig. 8B). DMF did not however block all ubiquitination within the cell. Halo-TUBE (Tandem Ubiquitin
`
`Binding Entities) beads were used to pull down all ubiquitin chains within the cells. Blotting of these pulldowns
`
`showed that LPS did not increase the total amount of K11, K48 or K63 linked polyubiquitin chains in these pull
`
`downs and that these levels were also not reduced by DMF (Supplementary Figure 2). Similarly using
`
`Halo-TAB2-NFZ beads to pull down K63 chains showed that neither LPS or DMF affected the amount of K63
`
`chains in these pull downs (Supplementary Figure 2). This probably reflects a high background level of K63 and
`
`K48 chains within the cell so that the changes induced by TLR signaling are too small to be seen. In line with
`
`this, blotting the Halo-TUBE or Halo-TAB2-NFZ pull downs for IRAK2 showed that LPS did induce the
`
`ubiquitination of this protein and that this was blocked by DMF (supplementary Figure 2). In line with the results
`
`in RAW264.7 cells, DMF also greatly reduced the amount of M1 chains that could be pulled down from BMDM
`
`lysates using either Halo-NEMO or Halo-TUBE beads (Fig. 9). Similarly the amount of ubiquitinated IRAK2 that
`
`came down in the Halo-NEMO pulldowns was also reduced by DMF. In contrast, DMF did not reduce the amount
`
`of K48 linked chains in Halo-TUBE pulldowns from BMDMs (Fig. 9).
`
`Figure 8
`
`Effect of DMF on LPS stimulated polyubiquitin chain formation in RAW264.7
`
`macrophages. RAW264.7 cells were incubated in the presence or absence of 50 µM DMF
`
`for 4 h.
`
`6 of 15
`
`10/18/2016 5:39 PM
`
`Page 6 of 15
`
`

`
`Dimethyl fumarate blocks pro-inflammatory cytokine production via inhi...
`
`https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4976367/
`
`Figure 9
`
`Effect of DMF on LPS stimulated polyubiquitin chain formation in
`
`BMDMs.
`
`M1 chains are formed by the LUBAC complex which contains HOIP, HOIL and Sharpin. In this complex HOIP
`47
`acts as the E3 and directly conjugates to ubiquitin via a cysteine during ubiquitin transfer
`. As it is possible that
`
`DMF might inhibit HOIP via reacting with this cysteine in cells, HOIP was immunoprecipitated from cells and its
`
`activity examined in an in vitro ubiquitination reaction. Due to the covalent modification of the cysteine by DMF,
`
`inhibition in the cell should be retained during the immunoprecipitation and in vitro assay. Consistent with
`40
`, HOIP was constitutively active in this assay, and this activity was not affected by pretreatment
`
`previous reports
`
`of the cells with DMF (Supplementary Figure 3).
`
`DMF inhibits K63/M1 chain formation in response to IL-1 and TNF
`
`The formation of K63 and M1 chains is not restricted to TLR signaling and also occurs downstream of other
`34,40
`. To examine chain formation downstream of IL-1, HEK293 cells stably
`
`stimuli including IL-1 and TNF
`
`expressing the IL-1 receptor were stimulated with IL-1 in the presence or absence of DMF. In these cells IL-1
`
`stimulated the phosphorylation of p105 and IKK as well as the degradation of IκBα which was blocked by DMF,
`
`as was the activation of ERK1/2 (Fig. 10A). In Halo-NEMO pulldowns, IL-1 stimulation increased the amount of
`
`both M1 and K63 chains observed, and this was blocked by pretreatment of the cells with DMF (Fig. 10B).
`
`Treatment of HeLa cells with TNF induced ERK1/2, IKK and p105 phosphorylation as well as the degradation of
`
`IκBα. ERK1/2 phosphorylation and IκBα degradation were inhibited by DMF, however in these cells DMF did
`
`not block p105 or IKK phosphorylation in response to TNF (Fig. 10C). NEMO pulldowns showed that TNF
`
`stimulated the formation of M1 polyubiquitin chains, and this was reduced by DMF. TNF only resulted in a minor
`
`increase in the signal for K63 chains, but again this was reduced by DMF (Fig. 10D).
`
`Figure 10
`
`Effect of DMF on IL-1 and TNF stimulated M1 chain formation.
`
`Discussion
`
`Go to:
`
`While the ability of DMF to modulate immune function is well established, its mechanism of action has been
`
`more elusive, and several potential in vivo targets have been suggested. While DMF can activate the Nrf2
`48
`
`anti-oxidant pathway, our results, along with those recently published by Gillard et al.
`
`, indicate that this is not
`
`required for the ability of DMF to block cytokine induction. In our hands DMF has a suppressive effect on several
`
`pathways downstream of TLR signaling, including NFκB and ERK1/2 signaling. We demostrate here that DMF
`
`can inhibit cytokine production in macrophages and that this correlates to the inhibition of NFκB and MAPK
`
`signaling pathways. This would suggest that DMF acts at an upstream point in TLR signaling, and in line with
`
`this we show that DMF can block the formation of signaling complexes containing M1 and K63 polyubiquitin
`
`chains downstream of TLR and IL-1 signaling. Furthermore, in vitro DMF can inhibit UbcH7 and Ubc13, the
`
`major E2 enzymes thought to be required for K63 and M1 chain formation in response to IL-1 or TLR agonists.
`
`DMF did not result in a global loss of polyubiquitin chains. Notably the levels of K63 and K48 chains in pull
`
`downs with Halo-TUBE beads was unaffected by DMF. One explanation is that the majority of these chains were
`
`preformed in the cell prior to the addition of DMF. If the turnover of the chains was slow, no effect of DMF
`
`7 of 15
`
`10/18/2016 5:39 PM
`
`Page 7 of 15
`
`

`
`Dimethyl fumarate blocks pro-inflammatory cytokine production via inhi...
`
`https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4976367/
`
`would be expected on the levels of these chains. In line with this, LPS did not increase the levels of either K48 or
`
`K63 chains in the Halo-TUBE pulldowns indicating the presence of a significant number of K48 and K63 cells in
`
`resting cells. These observations does not mean TLR signaling cannot promote the K48 or K63 linked
`
`ubiquitination of specific proteins only that any such modifications do not measurably alter the total amount of
`
`K48 and K63 chains in the cell. TLR signaling has been shown to promote K63 and M1 polyubiquitin chain
`
`formation on a number of signaling proteins including IRAK1. Previous reports have indicated that many of these
`40,46
`, and in line with this we see increased amounts of K63
`
`chains actually from as hybrids of K63 and M1 chains
`
`and M1 chains in pull downs with Halo-NEMO beads, which selectively pulls down complexes containing M1
`
`chains. The TLR induced formation of these K63 and M1 chains was blocked by pretreatment with DMF (Figs 8,
`
`9, 10).
`
`DMF is converted to MMF in vivo. The effects we observed on signaling were specific for DMF, as we did not
`
`observe any effect of MMF on LPS induced signaling. This is in agreement with a previous study showing that
`48
`. In macrophages we
`
`DMF, but not MMF, repressed TLR induced cytokine induction in primary human PBMCs
`
`show that DMF blocked the activation of the IKK complex and thus prevented both p105 phosphorylation and
`
`IκBα degradation. Previous reports have also indicated that DMF may inhibit NFκB transcriptional activity. For
`
`example, DMF reduced NFκB activity in LPS stimulated dendritic cells and TNF stimulated HUVEC or
`13,14,15
`
`. A common feature of these earlier studies was that DMF inhibited NFκB DNA binding or
`
`ASMC
`
`relocalization to the nucleus, but did not have a major effect on the degradation of IκBα. The reason for the
`
`difference between these studies and our findings is not clear. One potential explanation is the different cell types
`
`and stimuli used in the various studies may influence the results seen for IκBα degradation. In line with this, the
`
`ability of DMF to inhibit IκBα degradation in TNF stimulated HeLa cells was much less than in TLR stimulated
`
`macrophages. Another issue could relate to how DMF was used. The concentrations of DMF were similar in the
`
`different studies, however the pre-incubation times in these previous papers were not clear and were likely
`
`different to the 4 h preincubation used in our studies. As DMF can act as a covalent inhibitor it is able to display
`
`time dependent kinetics of inhibition (data not shown) and thus longer pre-incubation times might give a more
`
`pronounced inhibition of IκBα degradation.
`
`The effects we observed on NFκB and MAPK activation could be explained by inhibition of TLR signaling at the
`
`level of K63/M1 polyubiquitin chain formation and thus blocking the activation of Tak1. If DMF did act at the
`
`level of Tak1 or ubiquitination, it would also be expected to inhibit p38 activation. This is complicated by the
`
`observation that p38 was activated by DMF alone (Fig. 6), which would obscure any effect of DMF on TLR
`
`induced p38 activation pathways. One potential mechanism would be the inhibition of tyrosine phosphatases, as
`
`this is known to activate MAPK signaling. This is however unlikely as DMF did not result in increased global
`
`levels of phosphotyrosine. Another explanation could be activation of a MAP3K for p38 and JNK. One possible
`
`candidate might be Ask1, which is also known to be expressed in macrophages and can contribute to p38
`49
`50
`activation by LPS . Ask1 is activated by oxidative stress via a complex mechanism involving Trx1 . It is
`
`possible that DMF may activate Ask1 via modification of cysteine residues on Trx1 or via targeting proteins
`
`involved in the response to oxidative stress. Further work would be required to resolve this point.
`
`The effects of DMF on signaling have recently been suggested to have similarities to those produced by BAY
`48
`11-7082 . Interestingly while BAY 11-7082 was originally described as an IKK inhibitor a recent study has
`45
`shown that BAY 11-7082 can act in cells via inhibiting E2 enzymes, including Ubc13 and UbcH7 . As for DMF,
`
`the mechanism found for this involved the covalent modification of the active site cysteine in the E2.
`
`Taken together our results, along with those of others, would indicate that DMF has complex effects in cells and
`
`is unlikely to act via a single target. Instead it is likely that the overall therapeutic effects of DMF in MS or
`
`psoriasis are due to the ability of DMF to directly modify the function of a number of cellular proteins via the
`
`modification of reactive cysteines in th

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket