throbber
Cellular Signalling 21 (2009) 79–86
`
`Contents lists available at ScienceDirect
`
`Cellular Signalling
`
`Cellular
`Signalling
`- •-=:.;;:::_.
`
`.....LS~VIER
`
`j o u r n a l h om e p a g e : w w w. e l s ev i e r. c o m / l o c a t e / c e l l s i g
`
`Bruton's tyrosine kinase is dispensable for the Toll-like receptor-mediated activation
`of mast cells
`Carolin N. Zorn a, Simone Keck a, Rudi W. Hendriks b, Michael Leitges c,
`Marina A. Freudenberg a, Michael Huber a,d,⁎
`a Department of Molecular Immunology, Biology III, Albert-Ludwigs-University Freiburg and Max-Planck-Institute for Immunobiology, 79108 Freiburg, Germany
`b Department of Pulmonary Medicine, Erasmus Medical Center Rotterdam, NL-3000 CA Rotterdam, The Netherlands
`c Biotechnology Centre of Oslo, University of Oslo, Oslo, Norway
`d Centre for Biological Signaling Studies (bioss), Albert-Ludwigs-University Freiburg, Germany
`
`a r t i c l e
`
`i n f o
`
`a b s t r a c t
`
`Article history:
`Received 21 May 2008
`Received in revised form 11 September 2008
`Accepted 22 September 2008
`Available online 26 September 2008
`
`Keywords:
`Bruton's tyrosine kinase
`Mast cells
`Macrophages
`Lipopolysaccharide
`Lipopeptide
`Protein kinase C-β
`Signal transduction
`
`1. Introduction
`
`Bruton's tyrosine kinase (Btk) represents an important signaling element downstream of ITAM-containing
`receptors, e.g. FcεR1 and BCR. Btk is part of the calcium signalosome and thus, critically involved in
`intracellular calcium mobilization. Loss of Btk or expression of mutant forms results in severe disease
`phenotypes, X-linked agammaglobulinemia (XLA) and Xid in humans and mice, respectively. Previously,
`roles for Btk in TLR-mediated signal transduction have been found in monocytes/macrophages. In the present
`study we show that Btk deficiency moderately enhances or has no influence on the LPS- or lipopeptide-
`induced secretion of IL-6 and TNF-α from murine bone marrow-derived mast cells (BMMCs).
`Furthermore, activation of p38 kinase, which is required for cytokine production, is comparable in WT and
`Btk−/− BMMCs. Moreover, stability of the adaptor protein Mal as well as LPS-induced H2O2 production does
`not vary between WT and Btk−/− cells. Interestingly, PKC-β deficiency, which results in a Xid-like phenotype
`as well, has also no negative effect on LPS-induced cytokine secretion, suggesting that proteins of the calcium
`signalosome are not involved in TLR-mediated BMMC activation. In conclusion, the study reveals that Btk is
`dispensable for TLR signaling and function in murine BMMCs.
`
`© 2008 Elsevier Inc. All rights reserved.
`
`Btk is a cytoplasmic tyrosine kinase expressed in certain
`hemopoietic cells including B-lymphocytes, macrophages, mast
`cells, and platelets [1]. The importance of Btk is particularly obvious
`during B cell development and activation [2,3]. Btk deficiency
`manifests as X-linked agammaglobulinemia (XLA) and Xid in humans
`and mice, respectively [1]. Not only complete absence of Btk, but also
`defects in Btk, like point mutations in the PH and SH3 domains, can
`cause this severe disease [4]. Due to its role in B cell development, Btk-
`deficient patients/mice lack mature B cells and express dramatically
`reduced amounts of immunoglobulins [2]. Btk is part of the so-called
`calcium signalosome downstream of the BCR. It participates in the
`
`Abbreviations: BMMC, bone marrow-derived mast cell; BMMΦ, bone marrow-
`derived macrophage; Btk, Bruton's tyrosine kinase; LP,
`lipopeptide; Mal, MyD88
`adapter-like protein; MC, mast cell; SOCS1, suppressor of cytokine signaling 1; XLA, X-
`linked agammaglobulinemia.
`⁎ Corresponding author. Molecular Immunology, Institute for Biology III, University of
`Freiburg, Germany and Max-Planck-Institute for Immunobiology Stübeweg 51, 79108
`Freiburg, Germany. Tel.: +49 761 5108 438; fax: +49 761 5108 423.
`E-mail address: huberm@immunbio.mpg.de (M. Huber).
`
`0898-6568/$ – see front matter © 2008 Elsevier Inc. All rights reserved.
`doi:10.1016/j.cellsig.2008.09.010
`
`activation of PLC-γ2 and thus regulates the release of intracellular
`calcium from the endoplasmic reticulum [3]. The same function for
`Btk has been reported in mast cells (MCs) activated via the FcεR1 [5].
`Both FcεR1 and BCR are ITAM-containing receptors [6] and share
`elements and mechanisms of signal transduction [5].
`Btk has also been reported to be involved in the signal transduction of
`ITAM-independent receptor systems, e.g. TLR. However, the current
`literature provides in part conflicting data. Btk has been found to interact
`with components of the TLR4 and TLR2 signaling machinery, i.e. the
`adapter proteins, MyD88 and MyD88 adapter-like protein (Mal), as well
`as the interleukin-1 receptor-associated kinase-1 in HEK293 transfec-
`tants [7], and to be involved in transactivation of NFκB [8]. Furthermore,
`it was reported that Mal is subject to Btk-mediated tyrosine phosphor-
`ylation in response to TLR4 or TLR2 engagement [9] and this
`phosphotyrosine serves as an acceptor site for the suppressor of
`cytokine signaling 1 (SOCS1) protein. Binding of SOCS1 to Mal conveys
`this adaptor protein for degradation by the 26S proteasome [10]. Thus,
`Btk seems to be acting as part of a negative regulatory mechanism
`limiting primary innate immune responses. In accordance, degradation
`of Mal was absent in LPS-stimulated mouse Xid splenocytes [10]. From
`the involvement of Btk in degradation of Mal one would expect that cells
`lacking Btk function exhibit enhanced reactivity to LPS. However, the
`
`

`

`80
`
`C.N. Zorn et al. / Cellular Signalling 21 (2009) 79–86
`
`opposite effect of Btk deficiency, an impaired TNF-α response to LPS, was
`observed in mononuclear cells from XLA patients [11]. XLA mononuclear
`cells showed attenuated LPS-induced phosphorylation of p38 kinase
`compared to cells from control donors and this seemed to cause
`decreased stability of TNF-α mRNA in XLA cells [11]. In another study
`[12] TLR2-mediated stimulation of human XLA mononuclear cells
`resulted in impaired production of TNF-α and IL-1β, while production
`of IL-6, IL-8, and IL-10 remained unimpaired [12]. Finally, in a very
`thorough study involving seven XLA patients expressing no Btk protein,
`no effect of Btk deficiency on LPS-induced TNF-α and IL-6 production as
`well as activation of p38 kinase in monocytes was found [13]. Contrary,
`in a murine study Btk-deficient macrophages stimulated with LPS or
`other TLR ligands secreted enhanced levels of IL-6, as a result of
`suppressed IL-10 production [14].
`So far reported, investigations on the role of Btk in TLR signaling
`were carried out in monocytes/macrophages, but not in MCs. Unlike
`macrophages MCs do not express membrane (m)CD14 and differ from
`mCD14-positive macrophages in their LPS recognition properties [15].
`MCs, in the absence of soluble (s)CD14, recognize and react to rough
`(R)-chemotypes of LPS, while the reactivity to smooth (S)-chemotypes
`is practically absent. In this respect, they behave like macrophages
`deficient for CD14 or those expressing a mutant variant of CD14
`(‘heedless’) [16]. In contrast, wild-type macrophages are also able to
`recognize S-chemotypes of LPS in an LPS-binding protein/mCD14-
`dependent fashion [15,16].
`In this work studying the requirement of Btk for LPS- as well as
`lipopeptide (LP) -induced MC activation, we demonstrate that Btk
`moderately inhibits or has no influence on the activation of bone
`marrow-derived MCs (BMMCs) mediated by TLR4 or TLR2. Induction
`of TNF-α and IL-6 by LPS or LP, and activation of p38 kinase, and
`H2O2 production by LPS in Btk-deficient MCs is in comparison to WT
`MCs moderately enhanced or comparable. Interestingly, deficiency of
`PKC-β, which also results in a Xid-like phenotype in mice [17], does
`not influence LPS-induced cytokine production in BMMCs, suggest-
`ing that proteins involved in the organization of the calcium
`signalosome do not, or only marginally participate in TLR4-mediated
`signal transduction in MCs. Finally, Btk plays no obvious role in
`activation of murine bone marrow-derived macrophages (BMMΦs)
`by LPS or LP.
`
`2. Materials and methods
`
`2.1. Reagents
`
`R-form LPS from S. minnesota mutant R595 and S-form LPS from S.
`abortus equi were extracted and purified as described [18–20]. Synthetic
`lipopetides (Pam3CSK4, FSL-1) were obtained from Echaz Microcollec-
`tions (Tübingen, Germany). Polyclonal anti-Mal (T-16) antibody was
`obtained from Santa Cruz (Heidelberg, Germany) and polyclonal anti-
`p85 antibody (#06195) from Upstate/Biomol (Hamburg, Germany).
`Polyclonal anti-P-p38 (T180/Y182) antibody was purchased from Cell
`Signaling Technology (Frankfurt a. M., Germany). DNP-HSA containing
`30–40 mol DNP per mole albumin and monoclonal IgE with specificity
`for DNP (SPE-7) were purchased from SIGMA (Deisenhofen, Germany)
`and DMSO from J.T. Baker (Griesheim, Germany). Btk inhibitor, LFM A13
`[alpha-cyano-beta-hydroxy-beta-methyl-N-(2, 5-dibromophenyl) pro-
`penamide], and cPKC-specific inhibitor, Gö6976, were obtained from
`Calbiochem (Schwalbach, Germany). p38 inhibitor, BIRB0796, was
`purchased from the Division of Signal Transduction Therapy, College
`Of Life Sciences, University of Dundee, Dundee, Scotland, U.K.
`
`2.2. Animals
`
`Breeding of mice and all experiments were done in accordance
`with national guidelines and were approved by a local ethics
`committee.
`
`2.3. Bone marrow-derived mast cells and macrophages
`
`To obtain BMMCs, bone marrow cells (1×106/ml) from 6 to 8 week
`old male mice (129/Sv) were cultured (37 °C, 5% CO2) as a single cell
`suspension in RPMI 1640 medium containing 15% FCS, 1% X63Ag8-
`653-conditioned medium as a source of IL-3 [21], 2 mM L-glutamine,
`1×10− 5 M 2-mercaptoethanol, 50 units/ml penicillin, and 50 mg/ml
`streptomycin. At weekly intervals, the non-adherent cells were
`reseeded at 5 ×105 cells/ml in fresh medium. By 4–6 weeks in culture,
`greater than 99% of the cells were c-kit and FcεR1 positive as assessed
`by phycoerythrin-labeled anti-c-kit antibodies (Pharmingen, Missis-
`sauga, Canada) and FITC-labeled rat anti-mouse IgE antibodies
`(Southern Biotechnology, Birmingham, AL, USA), respectively. Btk+/+
`and −/− MCs were in vitro differentiated using the same protocol but
`starting from BM cells of 6 to 8 week old Btk+/+ and −/− littermates
`(C57BL/6). Wildtype and Xid MCs were in vitro differentiated from BM
`cells of 6 to 8 week old CBA/CaHN-Btkxid/J and CBA/J mice (The Jackson
`Laboratory, Bar Harbor, Maine, USA). PKC-α+/+, PKC-α−/−, PKC-β +/+,
`and PKC-β −/− BMMC were in vitro differentiated from BM cells of 6 to
`8 week old littermates (129/Sv).
`To obtain BMMΦs, BM cells from Btk+/+ and −/− mice were grown
`in the presence of L-cell-conditioned medium in teflon bags as
`described previously [22].
`
`2.4. MC stimulation and Western blotting
`
`IgE-preloaded cells were washed twice in RPMI/0.1% BSA and
`resuspended in RPMI/0.1% BSA. Cells were adapted to 37 °C for 30 min
`and stimulated with the indicated concentrations of DNP-HSA, LPS, or
`LP. After stimulation for different length of time, cells were pelleted
`and solubilized with 0.5% NP-40 and 0.5% deoxycholate in 4 °C
`phosphorylation solubilization buffer [23]. After normalizing for
`protein content, the postnuclear supernatants were subjected
`directly to SDS-PAGE and Western blot analysis as described pre-
`viously [24]. For measurement of extracellular cytokines, cell-free
`supernatants for cytokine measurements were stored in aliquots at
`−80 °C until use.
`
`2.5. Macrophage stimulation
`
`For induction of cytokines, macrophages were resuspended in
`serum-free DMEM (105 cells/0.2 ml/well), placed in 96-well plates
`(Nunc, Roskilde, Denmark) and cultured for 24 h at 37 °C in a
`humidified atmosphere containing 8% CO2. Thereafter macrophages
`were washed and stimulated in triplicates with different amounts of
`LPS or LP in 0.2 ml of DMEM. The culture supernatants for TNF-α and
`IL-6 measurements were collected 4 h and 24 h later, respectively, and
`stored in aliquots at −80 °C until used for cytokine measurements.
`
`2.6. Cytokine measurement
`
`Mouse IL-6 and TNF-α ELISA's (BD Pharmingen, Heidelberg,
`Germany) were performed according to the manufacturer's instruc-
`tions. The levels of cytokines in culture supernatants varied between
`experiments due to genetic background or age of the cells. Qualitative
`differences or similarities between WT and mutant cells, however,
`were consistent throughout the study.
`
`2.7. Flow cytometric analysis of H2O2 production
`
`IgE-sensitized BMMCs were washed twice with PBS, resuspended
`in 500 μl PBS, and stained with H2DCFDA (final concentration: 10 μM)
`for 30 min at 37 °C in the dark. Incubation in the dark was continued
`after the addition of the stimuli (Ag or LPS) for 5 min and flow
`cytometric analysis of cell samples was carried out subsequently
`using a FACScan cell analyzer (Beckton Dickinson, San Jose, USA).
`
`

`

`A
`1800 ,~----,
`D Btk+/+
`■ Btk-/-
`
`1400
`
`l 1000
`
`g
`<D
`...'.J
`
`600
`
`200
`
`B 400
`
`300
`
`l
`g 200
`d
`LL. z
`t-
`
`100
`
`** ,-,
`
`D Btk+/+
`■ Btk-/-
`
`C.N. Zorn et al. / Cellular Signalling 21 (2009) 79–86
`
`81
`
`**
`r-1
`
`Fluorescence intensity data (channel FL1) were collected from 25,000
`cells per measurement.
`
`2.8. RNA extraction and RT-PCR analysis
`
`Total RNA was isolated from 5 × 106 cells by a guanidinium
`isothiocyanate-phenol-chloroform-isoamyl alcohol procedure [25],
`as described previously [26]. Total RNA (1 μg) was reverse-transcribed
`with Moloney Murine Leucaemia Virus reverse transcriptase and
`oligo-p(dT) primers (Expand reverse transcriptase kit; Roche) accord-
`ing to the manufacturer's recommendations. PCR was performed
`using HotStart Taq DNA polymerase (Genaxxon) according to the
`manufacturer's instructions. The primer pairs for amplification
`of murine S0CS1, S0CS3, CISH, and actin were: S0CS1: sense, CCCTGG-
`CGACACTCACTT; antisense, GAGCGCGAAGAAGCAGTT (199-bp product
`size); S0CS3: sense, AGCGTCAAGACCCAGTCG; antisense, ACTTCGGAC-
`GAGGGTTCC (194-bp product size); CISH: sense, TCGGGAATCTGGG-
`TGGTA; antisense, TCCAGCCGGAAGCTAGAA (192-bp product size),
`and actin: sense, TGG AAT CCT GTG GCA TCC ATG AAA; antisense, TAA
`AAC GCA GCT CAG TAA CAG TCC (348-bp product size). An annealing
`temperature of 56 °C was used for actin primer pairs and 61 °C was
`used for S0CS1, S0CS3, and CISH primer pairs. An annealing time of
`25 s was used for S0CS1 as well as actin and of 45 s for S0CS3 and CISH.
`All PCR products were resolved by 2% agarose gel electrophoresis and
`were visualized by ethidium bromide staining.
`
`A
`
`3000
`
`2500
`
`* ,-,
`
`□ Btk+/+
`■ Btk-/-
`
`***
`r-1
`
`0 ..__ ___ ..__
`DNP
`con
`
`0,5
`
`2
`µg/ml LPS
`
`5
`
`C 1500
`
`1000
`
`l g
`
`<D
`...'.J
`
`500
`
`***
`r-1
`
`***
`r-1
`
`0
`
`con
`
`20 ng/ml 0.5µg/ml 2 µg/ml
`DNP
`LPS
`LPS
`
`Fig. 1. Btk is not involved in the positive regulation of LPS-induced cytokine secretion in
`BMMCs. (A) IgE-loaded Btk+/+ and Btk−/− BMMCs were stimulated for 3 h with Ag
`(DNP), increasing concentrations of R-LPS, or left unstimulated (con). Secreted IL-6 was
`measured by ELISA. Each point is the mean of triplicates ±SD. Comparable results were
`obtained with independent BMMC cultures. (B) Supernatants from Btk+/+ and Btk−/−
`BMMCs treated as described in (A) were analyzed by ELISA for the presence of TNF-α.
`Each point is the mean of triplicates± SD. Comparable results were obtained with
`independent BMMC cultures. (C) IgE-loaded WT and Xid BMMCs were treated for 3 h
`with Ag (DNP), increasing concentrations of R-LPS, or left unstimulated (con). IL-6 in the
`supernatants was measured by ELISA. Each point is the mean of triplicates± SD.
`Comparable results were obtained with independent BMMC cultures.
`
`_ 2000
`E
`°' g 1500
`
`<D
`...'.J
`
`1000
`
`500
`
`5
`10 20 0.5
`con DNP 5
`µg/ml FSL-1
`µg/ml Pam3CSK4
`
`***
`;:;:-;
`
`□ Btk+/+
`■ Btk-/-
`
`B 300
`
`=200
`
`t g
`
`d
`LL.
`~ 100
`
`Fig. 2. No attenuation of TLR2-mediated cytokine secretion in Btk-deficient BMMCs.
`(A) IgE-loaded Btk+/+ and Btk−/− BMMCs were stimulated for 3 h with Ag (DNP, 20 ng/
`ml), indicated concentrations of Pam3CSK4 (TLR2/TLR1 ligand) or of FSL-1 (TLR2/TLR6
`ligand), or left unstimulated (con). Secreted IL-6 was measured by ELISA. Each point is
`the mean of triplicates ±SD. Comparable results were obtained with independent
`BMMC cultures. (B) Supernatants from Btk+/+ and Btk−/− BMMCs treated as described
`under (A) were analyzed by ELISA for TNF-α expression. Each point is the mean of
`triplicates± SD. Comparable results were obtained with independent BMMC cultures.
`
`

`

`82
`
`C.N. Zorn et al. / Cellular Signalling 21 (2009) 79–86
`
`2.9. Statistical analysis
`
`All values in the figures (concerning bar graphs) are expressed as
`mean of SD of n observations (with n indicated in the respective figure
`
`legends). The relevant data sets were compared by unpaired two
`tailed Student's t test using Graph Pad Prism3 evaluation software
`(Graph Pad, San Diego, CA). P values of ⁎ b0.05, ⁎⁎ b0.005, and ⁎⁎⁎
`b0.0005 were considered statistically significant. Densitometric
`
`A
`Btk-/-
`Btk+/+
`- -- - - - - - - - - - - - - - -- - - - - - -
`DNP
`DNP
`LPS
`LPS
`
`5
`
`15
`
`30
`
`15
`
`30
`
`5
`
`15 30
`
`15
`
`30 min
`
`-----·--- - - _..--1
`WB: o.-P-p38 1--------------·------
`
`WB: o.-p85
`B
`
`□ Btk +/+
`160 ■ Btk -/-
`
`ns
`r7
`
`ns
`r7
`
`120
`
`:::,
`<Ii 80
`
`40
`
`0
`
`C
`
`-
`
`--
`
`WB: o.-actin
`D 3000
`
`= 2000
`~
`3
`(0
`....J
`-
`
`1000
`
`15
`DNP
`Btk +/+
`Pam
`FSL
`DNP
`------
`5 15 30 15 30 15 30
`
`5
`
`30
`
`30
`15
`LPS
`
`min
`
`Btk -/-
`Pam
`FSL
`DNP
`- - - - - -
`15 30 15 30 15 30 min
`
`5
`
`2 93 78 35 52 50 40 38 3 51 27 8 52 48 45 51
`WB: o.-P-p38
`
`--
`
`E
`
`fooNPj
`~
`2000
`
`E
`O>
`3
`~ 1000
`
`7
`
`ODMSO
`■ BI RB0796
`
`0
`
`OM
`
`0,01 0,03 0, 10 0,3
`µM BIRB0796
`
`0 ,....___.L-
`eon DNP FSL Pam LPS
`
`Fig. 3. p38 activation is important for LPS-induced IL-6 secretion, but is not dependent on Btk activity. (A) Btk+/+ and Btk−/− BMMCs were pre-loaded with IgE and stimulated with Ag
`(DNP, 20 ng/ml) or R-LPS (5 μg/ml) for the indicated time points, or were left untreated (−). Subsequently, postnuclear supernatants were analyzed by anti-P-p38 (upper panel) and
`anti-p85 (lower panel; loading control) immunoblotting. (B) Anti-P-p38 Western Blot shown in (A) together with two further experiments performed with independently cultured
`BMMCs were statistically evaluated with respect to changes between Btk+/+ and Btk−/− BMMCs. (C) IgE-preloaded Btk+/+ and Btk−/− BMMCs were stimulated with Ag (DNP, 20 ng/
`ml), FSL-1 (1 μg/ml), or Pam3CSK4 (Pam; 10 μg/ml) for the indicated time points, or were left untreated (−). Subsequently, postnuclear supernatants were analyzed by anti-P-p38
`(upper panel) and anti-actin (lower panel; loading control) immunoblotting. Densitometry was performed, and relative expression levels are indicated under each band. (D) IgE-
`loaded BMMCs were stimulated with Ag (DNP, 20 ng/ml) or R-LPS (5 μg/ml) for 3 h in the presence of the indicated concentrations of p38-specific inhibitor, BIRB0796, or vehicle
`(DMS0, DM). Inhibitor treatment was started 30 min before the addition of stimulus. IL-6 in the supernatants was measured by ELISA. Each point is the mean of triplicates±SD.
`Comparable results were obtained with independent BMMC cultures. (E) IgE-loaded BMMCs were left untreated (con) or stimulated with Ag (DNP, 20 ng/ml), FSL-1 (FSL, 1 μg/ml),
`Pam3CSK4 (Pam, 10 μg/ml), or R-LPS (5 μg/ml) for 3 h in the presence of 0.1 μM BIRB0796 or vehicle (DMS0). Inhibitor treatment was started 30 min before the addition of the stimuli.
`IL-6 in the supernatants was measured by ELISA. Each point is the mean of triplicates± SD. Comparable results were obtained with independent BMMC cultures.
`
`

`

`analysis of Western blots was performed using publicly available
`ImageJ software [27].
`
`A
`
`Btk+/+
`0.5
`
`2
`
`3
`
`0.5
`
`2
`
`3 h
`
`Btk-/-
`
`C.N. Zorn et al. / Cellular Signalling 21 (2009) 79–86
`
`83
`
`3. Results
`
`3.1. Btk deficiency enhances or does not affect LPS- or LP-triggered
`cytokine secretion in BMMCs, respectively
`
`The role of Btk in TLR signaling in MCs was not studied yet. Such
`investigation appeared interesting since activation of MCs via TLR4 or
`TLR2, unlike a similar activation of macrophages, proceeds without
`help of mCD14 and may thus differ from the latter [15]. Here, mast
`cells generated from bone marrow precursors, BMMCs, of wild type
`and Btk-deficient mice were stimulated with LPS in vitro and cytokine
`responses were measured. As shown in Fig. 1A, the levels of IL-6
`secreted by LPS-stimulated Btk−/− and WT BMMCs were either
`comparable or higher in the cultures of Btk−/− cells. Likewise, LPS-
`stimulated Btk−/− BMMCs secreted higher TNF-α levels than WT cells
`(Fig. 1B). However, as reported previously [28], marked reduction of
`secretion of IL-6 and TNF-α was observed in Btk-deficient BMMCs
`compared to WT cells after stimulation via the FcεR1 (Fig. 1A and B).
`Comparable results to those obtained with Btk-deficient BMMCs were
`obtained in studies of IL-6 responses to LPS or Ag of cells derived from
`BtkXid mice (Fig. 1C).
`In this study we also investigated the role of Btk in the IL-6 and TNF-α
`responses of BMMCs to LPs. As shown in Fig. 2A and B, WT and Btk-
`deficient BMMCs stimulated with the synthetic analoga of bacterial
`lipopeptides, FSL-1 (TLR2/TLR6 ligand) and Pam3CSK4 (TLR2/TLR1 ligand),
`secreted moderately enhanced or comparable levels of IL-6 and TNF-α,
`indicating that neither TLR2/TLR6- nor TLR2/TLR1-mediated signaling
`depends on Btk activation in BMMCs.
`
`3.2. TLR-mediated activation of p38 is independent of Btk in BMMCs
`
`In addition to the impaired TNF-α response, impaired activation/
`phosphorylation of p38 kinase was observed earlier in Btk-deficient/-
`compromised monocytes stimulated with LPS [11]. Fitting with our
`data on LPS-induced cytokine secretion in BMMCs, no defect in p38
`phosphorylation was observable in LPS-treated Btk−/− compared to
`WT BMMCs (Fig. 3A and B). Furthermore, also lipopeptide-induced
`p38 phosphorylation was independent of Btk (Fig. 3C). These data
`indicate that in MCs TLR-driven activation of p38 is independent of
`Btk. Ag-triggered p38 activation, however, was severely compromised
`in Btk-deficient BMMCs (Fig. 3A–C). To proof that LPS-induced IL-6
`secretion in BMMCs indeed is dependent on p38 activation, we per-
`formed dose–response analyses with the highly specific p38 inhibitor,
`BIRB0769 [29]. This inhibitor, at the dose of 0.1 μM, blocked both Ag-
`and LPS-triggered IL-6 and TNF-α secretion in BMMCs (Fig. 3D and
`data not shown). Likewise, LP-induced IL-6 and TNF-α responses were
`abrogated by using this inhibitor (Fig. 3E and data not shown). These
`data indicate that TLR-mediated cytokine secretion in BMMCs is, like
`in monocytes, dependent on p38 activation. However, this activation
`is, unlike in monocytes [11], independent of Btk.
`
`3.3. Btk deficiency in BMMCs does neither influence stability of Mal nor
`TLR4-mediated H2O2 production
`
`In macrophages, Btk has been demonstrated to be responsible for
`Mal tyrosine phosphorylation and the subsequent S0CS1/proteasome-
`mediated degradation of this adapter protein [10]. Next, we aimed at
`studying this feedback mechanism in BMMCs. However, we did not
`observe degradation of Mal, neither in WT nor Btk−/− BMMCs, during
`the first 3 h of stimulation (Fig. 4A). This suggests that the respective
`pathway (e.g. S0CS1 expression) is not functional in BMMCs. Indeed,
`performing RT-PCR analysis we observed increased expression of the
`S0CS family member CISH in BMMCs, whereas S0CS1 expression was
`
`WB: a-Mal 1--- -
`
`WB: o.-p85
`
`BMMCs
`.5
`1
`
`2
`
`B
`
`SOCS1
`
`SOCS3
`
`GISH
`
`actin
`
`C
`
`.!!J.
`al
`u
`0
`<ii
`.0
`E
`:::,
`C
`
`BMMCbs
`.5
`1
`I
`
`0
`
`I~ y
`
`11
`
`2
`
`CJJ
`,;;.

`
`CJJ
`,;;.
`-+-
`
`10° 101
`
`102
`
`103
`
`10° 101
`H20 2
`
`102
`
`103
`
`104
`
`Fig. 4. Btk deficiency does interfere neither with Mal stability nor LPS-induced H2O2
`production. (A) Btk+/+ and Btk−/− BMMCs were stimulated with R-LPS (5 μg/ml) for the
`indicated times or left unstimulated (−). Postnuclear supernatants were subjected to
`Western blot analysis with anti-Mal (upper panel) and anti-p85 (lower panel; loading
`control) antibodies. (B) BMMCs (left panels) and BMMΦs (right panels) were stimulated
`with 10 μg/ml and 1 μg/ml R-LPS, respectively, for the indicated time points. RT-PCR
`analyses were performed using primers for S0CS1, S0CS3, CISH, and actin (loading
`control). Comparable results were obtained with independent BMMC and BMMΦs
`cultures. (C) Btk+/+ (upper panels) and Btk−/− BMMCs (lower panels) were loaded with
`IgE overnight and subsequently incubated for 30 min with the dye H2DCFDA. Cells were
`then either left unstimulated (thin lines) or treated with Ag (DNP, 20 ng/ml; left panels)
`or R-LPS (5 μg/ml; right panels) (thick lines) and H2O2 production analyzed by FACS.
`Comparable results were obtained with independent BMMC cultures.
`
`reduced in response to LPS (Fig. 4B). In contrast, in BMMΦs S0CS1,
`S0CS3, and CISH expression were induced in response to LPS
`stimulation (Fig. 4B).
`Previously, Mangla et al. reported on the positive role of Btk in the
`generation of bursts of reactive oxygen intermediates in macrophages
`[30]. We and others described PI3K-dependent H2O2 production in
`Ag-triggered BMMCs [31,32]. Here we investigated whether LPS is able
`
`

`

`84
`
`C.N. Zorn et al. / Cellular Signalling 21 (2009) 79–86
`
`to stimulate H2O2 production in BMMCs as well and whether this
`process in Ag- or LPS-simulated cells is dependent on Btk. As shown in
`Fig. 4C, both Ag and LPS induced intracellular H2O2 in BMMCs with LPS
`being the weaker stimulus. Interestingly, whereas the Ag-induced
`response was strongly dependent on Btk, LPS-triggered H2O2 produc-
`tion appeared not to be affected by Btk deficiency (Fig. 4C).
`
`3.4. PKC-β deficiency mimics Btk deficiency with respect to LPS- and
`Ag-triggered IL-6 secretion
`
`In addition to Btk deficiency, several deficiencies have been demon-
`strated to result in a Xid-like phenotype, one of them being PKC-β
`deficiency [17]. In agreement with an earlier publication [33], in this study
`we found that Ag-triggered IL-6 production was markedly compromised
`in PKC-β−/− BMMCs, while the production in PKC-α−/−cells was
`unimpaired (Fig. 5A). However, neither PKC-α nor PKC-β single deficiency
`(Fig. 5A), nor PKC-α/PKC-β double deficiency (data not shown), had a
`negative effect on the LPS-triggered IL-6 response of BMMCs. Using an
`inhibitor specific for cPKCs, Gö6976, Ag-triggered IL-6 production
`was almost completely inhibited at a concentration of 0.3 μM, whereas
`
`*** i
`
`D wr
`□ PKC-a-/(cid:173)
`■ PKC-P-/-
`
`A
`7000
`
`5000
`E
`O> s
`~3000
`
`a
`
`LPS
`
`1000 -
`
`■
`O con DNP
`
`B 250
`
`200
`
`E 150
`O>
`8
`<O
`...'.J 100
`
`50
`
`O DMSO
`
`0.1
`0.3
`- - - - -
`µM Go6976
`
`Fig. 5. PKC-β-deficiency results in compromised IL-6 production after Ag treatment, but
`normal response to LPS. (A) WT, PKC-α−/−, and PKC-β−/− BMMCs were left untreated
`(con) or stimulated for 3 h with Ag (DNP, 20 ng/ml), or R-LPS (2 μg/ml). Subsequently, IL-
`6 in the supernatants was measured by ELISA. Each point is the mean of triplicates± SD.
`Comparable results were obtained with independent BMMC cultures. (B) IgE-loaded
`BMMCs were stimulated with Ag (DNP, 20 ng/ml) or R-LPS (2 μg/ml) for 3 h in the
`presence of vehicle (DMS0) or the indicated concentrations of the cPKC inhibitor,
`Gö6976. Inhibitor treatment was started 30 min before the addition of the stimuli. IL-6
`in the supernatants was measured by ELISA. Each point is the mean of triplicates± SD.
`
`**
`
`D DMSO
`CLFM A13 10 µM
`■ LFM A13 100 µM
`***
`r.;-,
`
`4000
`
`= 3000
`~
`.3
`~ 2000
`
`1000
`
`OJ...J.. ...... -..1--....1....1..---..;;;;;,-....._ ..................... . - - -
`con DNP LPS
`con DNP LPS
`Btk +/+
`Btk -/ -
`
`Fig. 6. LFM A13 exerts off-target effects in Btk-deficient BMMCs. IgE-loaded Btk+/+ and
`Btk−/− BMMCs were left untreated (con) or stimulated with Ag (DNP, 20 ng/ml) or R-LPS
`(2 μg/ml) for 3 h in the presence of vehicle (DMS0), 10 μM LFM A13, or 100 μM LFM A13.
`Inhibitor treatment was started 30 min before the addition of stimulus. IL-6 in the
`supernatants was measured by ELISA. Each point is the mean of triplicates± SD.
`
`LPS-induced IL-6 secretion was only marginally affected (Fig. 5B). This
`indicates that in BMMCs neither PKC-β- nor Btk deficiency exerts a
`negative impact on LPS-induced cytokine production. Moreover, this
`suggests that different deficiencies resulting in a Xid-like phenotype do
`not abrogate TLR-mediated BMMC activation.
`
`3.5. LFM A13 blocks LPS-induced IL-6 production even in Btk-deficient
`BMMCs
`
`In different studies, the Btk-specific inhibitor, LFM A13, has been
`used to show Btk dependence of TLR-mediated signal transduction
`[8 –10]. However, the potential off-target effect of this drug was not
`investigated in Btk-deficient or -mutant cells. Therefore, we measured
`IL-6 production in WT as well as Btk-deficient BMMCs treated with
`different concentrations of LFM A13 (10 and 100 μM) and subse-
`quently stimulated with Ag or LPS. As shown in Fig. 6, LFM A13 at
`100 μM, a concentration range used by others [8–10,34], completely
`inhibited both Ag- and LPS-triggered IL-6 secretion in both WT and
`Btk-deficient BMMCs. Whereas 10 μM LFM A13 did not show any effect
`on the Ag-induced IL-6 production, it slightly attenuated LPS-induced
`production in WT and even stronger in Btk−/− BMMCs (Fig. 6). This
`clearly indicates that the inhibitory effect of LFM A13 on IL-6 produc-
`tion of MCs is not a result of Btk blockade. Therefore, LFM A13 cannot
`be considered a Btk-specific inhibitor, and that data on the role of
`Btk obtained solely by the usage of this drug might very likely be
`erroneous.
`
`3.6. Btk deficiency does not impair LPS or LP-triggered cytokine secretion
`in bone marrow-derived macrophages
`
`Conflicting results were obtained when the role of Btk for TLR
`signaling was studied in human monocytes or mouse macrophages
`[11–13]. Since we found Btk to be dispensable for TLR-mediated acti-
`vation of MCs derived from mouse bone marrow, we finally analyzed
`the role of Btk in the TLR-triggered induction of cytokines in macro-
`phages grown in vitro from murine bone marrow precursors. For
`this purpose, Btk+/+ and Btk−/− BMMΦs were stimulated with R- and
`S-chemotypes of LPS, as well as with the LPs, FSL-1 and Pam3CSK4, and
`production of IL-6 (Fig. 7A) and TNF-α (Fig. 7B) was measured. In
`agreement with the data by Perez de Diego [13], but contrary to reports
`demonstrating abrogated TLR responses of Btk-deficient macrophages
`[11,12], the LPS- and LP-induced cytokine production was indistin-
`guishable between WT and Btk-deficient BMMΦs. These data in
`
`

`

`C.N. Zorn et al. / Cellular Signalling 21 (2009) 79–86
`
`85
`
`A
`
`□ Btk+/+
`7 ■ Btk-/-
`
`* r-=
`
`**
`r-=
`
`**
`r-=
`
`0
`
`con
`
`B
`
`3
`
`0 Btk+/+
`■ Btk-/-
`
`I
`
`£2
`t:l
`LL
`z
`I-
`
`1
`
`Pam3CSK4
`
`* r-=
`
`* r-=
`
`0
`
`con
`
`1 10 100ng/ml
`Pam3CSK4
`
`Fig. 7. Btk-deficient BMMΦs are not compromised in cytokine production in response to
`different TLR ligands. (A) Btk+/+ and −/− BMMΦs were left untreated (con) or stimulated
`for 24 h with the indicated concentrations of R-LPS, S-LPS, FSL-1, or Pam3CSK4. IL-6 in
`the supernatants was measured by ELISA. Each point is the mean of triplicates± SD.
`(B) Btk+/+ and −/− BMMΦs were left untreated (con) or stimulated for 4 h as in A. TNF-α
`in the supernatants was measured by ELISA. Each point is the mean of triplicates± SD.
`Each point is the mean of triplicates± SD. Comparable results were obtained with
`different BMMΦ cultures.
`
`combination with the published reports suggest that the source and
`differentiation conditions of macrophages are decisive for their
`dependence on Btk for TLR signaling.
`
`4. Discussion
`
`Several research groups reported on the role of Btk in TLR signaling
`in monocytes/macrophages of murine as well as human origin
`[8,10–14].
`In this study,
`loss of Btk function either moderately
`enhanced (LPS triggering) or did not influence (LP triggering) TLR4 and
`TLR2 mediated responses of mouse BMMCs. Thus, in MCs Btk exerts a
`weak inhibitory function on TLR4 signaling and has no obvious effect
`on TLR2 signaling. Earlier, Btk was ascribed a negative regulatory role
`in activation of mouse dendritic cells, mainly through the autocrine
`secretion of IL-10 [35]. Accordingly, Btk−/− DCs produce less IL-10 in
`response to LPS [35]. This mechanism, however, seems to be absent in
`BMMCs, since in preliminary experiments we were unable to detect
`LPS-induced IL-10 production in BMMCs (data not shown). Moreover,
`in another study, production of IL-10 and IL-12p70 was comparable
`between DCs from healthy donors and XLA patients [34].
`We also show in this study (Fig. 7) that macrophages differentiated
`from WT or Btk−/− murine bone marrow cells do not exert differences
`in TLR-mediated production of pro-inflammatory cytokines although
`such differences were reported by others [11,12,14]. One important
`
`question in light of all these apparent discrepancies is how can such
`significant differences between

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