throbber
FEBS 21942
`
`FEBS Letters 450 (1999) 117^122
`
`A fusion protein of interleukin-11 and soluble interleukin-11 receptor acts
`as a superagonist on cells expressing gp130
`
`Stefan P£anza, Ingrid Tackena, Joachim Gro«tzingera, Yannick Jacquesb, Heike Dahmena,
`Peter C. Heinricha, Gerhard Mu«ller-Newena;*
`aInstitut fu«r Biochemie, Rheinisch-Westfa«lische Technische Hochschule Aachen, PauwelsstraMe 30, D-52057 Aachen, Germany
`bGroupe de Recherche Cytokines et Re¨cepteurs, Unite¨ INSERM 463, 9 Quai Moncousu, 44035 Nantes Cedex 1, France
`
`Received 22 March 1999
`
`IL-11 belongs to a subfamily of the 4-helix bundle cytokines
`including interleukin-6 (IL-6),
`leukemia inhibitory factor
`(LIF), oncostatin M (OSM), ciliary neurotrophic factor
`(CNTF) and cardiotrophin-1 (CT-1) that signal via the com-
`mon signal transducing receptor protein gp130 (for review see
`[8]). IL-11 ¢rst binds to its speci¢c K-receptor subunit (IL-
`11R) [9,10] and subsequently, the complex of IL-11 and IL-
`11R triggers the dimerization of gp130 [11] leading to activa-
`tion of the Janus kinase (Jak)/signal transducer and activator
`of transcription (STAT) signal transduction pathway [12].
`Both receptor components belong to the family of hemato-
`poietic cytokine receptors which are characterized by the pres-
`ence of at least one cytokine binding module (CBM) [13]. A
`CBM consists of two ¢bronectin type III-like domains. The
`N-terminal domain is characterized by four conserved cysteine
`residues, the C-terminal domain contains a WSXWS motif.
`The extracellular part of the IL-11R is proposed to consist
`of an Ig-like domain (D1) followed by a single CBM (D2 and
`D3) [9]. Whereas gp130 is expressed ubiquitously [14], IL-11R
`expression is more restricted [9,10]. Cells expressing gp130 but
`lacking membrane-bound IL-11R can be stimulated by the
`combination of IL-11 and soluble IL-11R (sIL-11R) indicat-
`ing that the cytoplasmic and transmembrane parts of IL-11R
`are not required for signal transduction [15].
`In this study, we show that a fusion protein of human IL-11
`and a fragment of human sIL-11R acts as a superagonist to
`which gp130 expressing cells respond more sensitive than to
`the combination of IL-11 and sIL-11R. Superagonistic activ-
`ity has also been described for a recombinant fusion protein
`consisting of a section of sIL-6R connected to IL-6 by a
`polypeptide linker [16]. The covalent linkage of two soluble
`receptor activating components exists naturally as well: the
`two separately encoded subunits of IL-12 (p35 and p40)
`undergo disul¢de-stabilized heterodimerization to constitute
`its biologically active form [17]. Our results support the idea
`that the covalent linkage of two soluble components required
`for receptor activation potentiates their bioactivity.
`
`2. Materials and methods
`
`Interleukin-11 is a hematopoietic cytokine that
`Abstract
`signals via the signal transducer gp130. Although gp130 is
`ubiquitously expressed,
`interleukine-11 responsiveness is re-
`stricted to cells that express the interleukine-11 receptor KK-
`subunit. The interleukine-11 receptor KK-subunit can be function-
`ally replaced by its soluble form indicating that the transmem-
`brane and cytoplasmic parts are not required for signal
`transduction. Here, we show that a recombinant fusion protein
`of a fragment of the human interleukine-11 receptor KK-subunit
`ectodomain linked to human interleukine-11 acts as a super-
`agonist on cells expressing gp130 but lacking the membrane-
`bound interleukine-11 receptor KK-subunit. It induces acute phase
`protein synthesis in hepatoma cells and efficiently promotes
`proliferation of Ba/F3 cells stably, transfected with gp130. In
`these bioassays, the fusion protein of a fragment of the human
`interleukine-11 receptor KK-subunit ectodomain linked to human
`interleukine-11 is 50 times more potent than the combination of
`interleukine-11 and the soluble interleukine-11 receptor KK-
`subunit. Thus, our findings support the concept that covalent
`fusion of two soluble proteins required for receptor activation
`dramatically increases their bioactivity.
`z 1999 Federation of European Biochemical Societies.
`
`Key words: Fusion protein; Interleukin-11; Superagonist;
`Gp130
`
`1. Introduction
`
`Interleukin-11 (IL-11) is a hematopoietic cytokine that ex-
`erts various biological responses on di¡erent cells and tissues
`(for review see [1]). IL-11 acts synergistically with other
`growth factors to stimulate various stages of hematopoiesis
`both in vitro and in vivo [2^4]. In a phase II clinical trial, it
`has been shown that due to its thrombopoietic activity, re-
`combinant human IL-11 ameliorates severe chemotherapy-in-
`duced thrombocytopenia therefore representing a valuable
`therapeutical protein [4]. More recently, anti-in£ammatory
`acitivities of IL-11 have been described [5,6]. Female mice
`that do not respond to IL-11 due to targeted deletion of a
`critical receptor component are infertile as a consequence of
`impaired trophoblast implantation [7].
`
`*Corresponding author. Fax: (49) (241) 88 88428.
`E-mail: mueller-newen@rwth-aachen.de
`
`Abbreviations: IL, interleukin; s, soluble; R, receptor; IL-11/R-FP,
`interleukin-11/receptor fusion protein; Jak, Janus kinase; STAT, sig-
`nal transducer and activator of transcription; CBM, cytokine binding
`module; D, domain
`
`2.1. Enzymes, antibodies and cell culture
`Enzymes were purchased from Boehringer Mannheim (Mannheim,
`Germany). The hIL-11 polyclonal antibody used in this study was
`obtained from R and D systems (Wiesbaden-Nordenstadt, Germany).
`K[32P]dATP was purchased from Amersham International (Amer-
`sham, UK) and tran[35S]-label metabolic labelling reagent from ICN
`(Meckenheim, Germany). Other chemicals were obtained from Roth
`(Karlsruhe, Germany) and media components for the cultivation of
`Pichia pastoris were purchased from Sigma-Aldrich (Deisenhofen,
`Germany) or Gibco (Eggenstein, Germany). HepG2 cells were main-
`tained in DMEM/F12 (Life Technologies, Eggenstein, Germany). Re-
`
`Singapore Exhibit 2009
`Lassen v. Singapore et al.
`PGR2019-00053
`
`0014-5793 / 99 / $20.00 ß 1999 Federation of European Biochemical Societies. All rights reserved.
`PII: S 0 0 1 4 - 5 7 9 3 ( 9 9 ) 0 0 4 7 7 - 9
`
`

`

`FEBS 21942
`
`FEBS Letters 450 (1999) 117^122
`
`A fusion protein of interleukin-11 and soluble interleukin-11 receptor acts
`as a superagonist on cells expressing gp130
`
`Stefan P£anza, Ingrid Tackena, Joachim Gro«tzingera, Yannick Jacquesb, Heike Dahmena,
`Peter C. Heinricha, Gerhard Mu«ller-Newena;*
`aInstitut fu«r Biochemie, Rheinisch-Westfa«lische Technische Hochschule Aachen, PauwelsstraMe 30, D-52057 Aachen, Germany
`bGroupe de Recherche Cytokines et Re¨cepteurs, Unite¨ INSERM 463, 9 Quai Moncousu, 44035 Nantes Cedex 1, France
`
`Received 22 March 1999
`
`Interleukin-11 is a hematopoietic cytokine that
`Abstract
`signals via the signal transducer gp130. Although gp130 is
`ubiquitously expressed,
`interleukine-11 responsiveness is re-
`stricted to cells that express the interleukine-11 receptor KK-
`subunit. The interleukine-11 receptor KK-subunit can be function-
`ally replaced by its soluble form indicating that the transmem-
`brane and cytoplasmic parts are not required for signal
`transduction. Here, we show that a recombinant fusion protein
`of a fragment of the human interleukine-11 receptor KK-subunit
`ectodomain linked to human interleukine-11 acts as a super-
`agonist on cells expressing gp130 but lacking the membrane-
`bound interleukine-11 receptor KK-subunit. It induces acute phase
`protein synthesis in hepatoma cells and efficiently promotes
`proliferation of Ba/F3 cells stably, transfected with gp130. In
`these bioassays, the fusion protein of a fragment of the human
`interleukine-11 receptor KK-subunit ectodomain linked to human
`interleukine-11 is 50 times more potent than the combination of
`interleukine-11 and the soluble interleukine-11 receptor KK-
`subunit. Thus, our findings support the concept that covalent
`fusion of two soluble proteins required for receptor activation
`dramatically increases their bioactivity.
`z 1999 Federation of European Biochemical Societies.
`
`Key words: Fusion protein; Interleukin-11; Superagonist;
`Gp130
`
`1. Introduction
`
`Interleukin-11 (IL-11) is a hematopoietic cytokine that ex-
`erts various biological responses on di¡erent cells and tissues
`(for review see [1]). IL-11 acts synergistically with other
`growth factors to stimulate various stages of hematopoiesis
`both in vitro and in vivo [2^4]. In a phase II clinical trial, it
`has been shown that due to its thrombopoietic activity, re-
`combinant human IL-11 ameliorates severe chemotherapy-in-
`duced thrombocytopenia therefore representing a valuable
`therapeutical protein [4]. More recently, anti-in£ammatory
`acitivities of IL-11 have been described [5,6]. Female mice
`that do not respond to IL-11 due to targeted deletion of a
`critical receptor component are infertile as a consequence of
`impaired trophoblast implantation [7].
`
`*Corresponding author. Fax: (49) (241) 88 88428.
`E-mail: mueller-newen@rwth-aachen.de
`
`Abbreviations: IL, interleukin; s, soluble; R, receptor; IL-11/R-FP,
`interleukin-11/receptor fusion protein; Jak, Janus kinase; STAT, sig-
`nal transducer and activator of transcription; CBM, cytokine binding
`module; D, domain
`
`IL-11 belongs to a subfamily of the 4-helix bundle cytokines
`including interleukin-6 (IL-6),
`leukemia inhibitory factor
`(LIF), oncostatin M (OSM), ciliary neurotrophic factor
`(CNTF) and cardiotrophin-1 (CT-1) that signal via the com-
`mon signal transducing receptor protein gp130 (for review see
`[8]). IL-11 ¢rst binds to its speci¢c K-receptor subunit (IL-
`11R) [9,10] and subsequently, the complex of IL-11 and IL-
`11R triggers the dimerization of gp130 [11] leading to activa-
`tion of the Janus kinase (Jak)/signal transducer and activator
`of transcription (STAT) signal transduction pathway [12].
`Both receptor components belong to the family of hemato-
`poietic cytokine receptors which are characterized by the pres-
`ence of at least one cytokine binding module (CBM) [13]. A
`CBM consists of two ¢bronectin type III-like domains. The
`N-terminal domain is characterized by four conserved cysteine
`residues, the C-terminal domain contains a WSXWS motif.
`The extracellular part of the IL-11R is proposed to consist
`of an Ig-like domain (D1) followed by a single CBM (D2 and
`D3) [9]. Whereas gp130 is expressed ubiquitously [14], IL-11R
`expression is more restricted [9,10]. Cells expressing gp130 but
`lacking membrane-bound IL-11R can be stimulated by the
`combination of IL-11 and soluble IL-11R (sIL-11R) indicat-
`ing that the cytoplasmic and transmembrane parts of IL-11R
`are not required for signal transduction [15].
`In this study, we show that a fusion protein of human IL-11
`and a fragment of human sIL-11R acts as a superagonist to
`which gp130 expressing cells respond more sensitive than to
`the combination of IL-11 and sIL-11R. Superagonistic activ-
`ity has also been described for a recombinant fusion protein
`consisting of a section of sIL-6R connected to IL-6 by a
`polypeptide linker [16]. The covalent linkage of two soluble
`receptor activating components exists naturally as well: the
`two separately encoded subunits of IL-12 (p35 and p40)
`undergo disul¢de-stabilized heterodimerization to constitute
`its biologically active form [17]. Our results support the idea
`that the covalent linkage of two soluble components required
`for receptor activation potentiates their bioactivity.
`
`2. Materials and methods
`
`2.1. Enzymes, antibodies and cell culture
`Enzymes were purchased from Boehringer Mannheim (Mannheim,
`Germany). The hIL-11 polyclonal antibody used in this study was
`obtained from R and D systems (Wiesbaden-Nordenstadt, Germany).
`K[32P]dATP was purchased from Amersham International (Amer-
`sham, UK) and tran[35S]-label metabolic labelling reagent from ICN
`(Meckenheim, Germany). Other chemicals were obtained from Roth
`(Karlsruhe, Germany) and media components for the cultivation of
`Pichia pastoris were purchased from Sigma-Aldrich (Deisenhofen,
`Germany) or Gibco (Eggenstein, Germany). HepG2 cells were main-
`tained in DMEM/F12 (Life Technologies, Eggenstein, Germany). Re-
`
`0014-5793 / 99 / $20.00 ß 1999 Federation of European Biochemical Societies. All rights reserved.
`PII: S 0 0 1 4 - 5 7 9 3 ( 9 9 ) 0 0 4 7 7 - 9
`
`

`

`118
`
`S. P£anz et al./FEBS Letters 450 (1999) 117^122
`
`combinant Trx/IL-11 and sIL-11R were prepared from bacteria or
`insect cells, respectively, as described previously [18]. Ba/F3-gp130
`cells [19] were cultured in DMEM containing 5% (v/v) conditioned
`medium from X63Ag8-653 BPV-mIL-3 myeloma cells (as a source of
`IL-3) in the presence of 1 mg/ml G418 in a water-saturated atmos-
`phere containing 5% CO2. Both media were supplemented with 10%
`(v/v) fetal calf serum, streptomycin (100 Wg/ml) and penicillin (60 Wg/
`ml).
`
`2.2. Plasmid construction
`First, a section of the hIL-11R cDNA, encoding domains 2 and 3
`(amino acids (aa) 109^318), was ampli¢ed by standard PCR methods.
`EcoRI/S¢I restriction sites (underlined) were introduced using the
`IL-11R-sense 5P-ACTCTCGGATCCCT-
`following primer pair:
`GGGCTACCCTCCAGC-3P and IL-11R-antisense 5P-TGAGAGGG-
`CCGGCTGGGCCCCCAGTGCTCGGAGTTCC-3P. Second, a frag-
`ment of hIL-11 cDNA encoding the proposed functional part of hIL-
`11 (aa 29^199) was ampli¢ed. Asp718/SacII restriction sites were in-
`troduced using the following primer pair: IL-11-sense 5P-AGA-
`CACGGTACCCCTCGAGTTTCCCCAGAC-3P and IL-11-antisense
`5P-ACTGTGCCGCGGTCACAGCCGAGTCTTCAG-3P. The plas-
`mid pPICZKA (Invitrogen) for secreted expression of recombinant
`proteins in the methylotrophic yeast P. pastoris was digested with
`the restriction endonucleases EcoRI and S¢I. Then, the ¢rst amplicon
`was ligated (pPICZKA/IL-11R). The resulting plasmid was digested
`with Asp718 and SacII and the second amplicon was ligated, which
`led to a plasmid encoding both proteins but lacking a peptide linker
`(pPICZKA/IL-11R/IL-11). The two following oligonucleotides, encod-
`ing the linker sequence, were annealed and subsequently ligated into
`the S¢I/Asp718-digested pPICZKA/IL-11R/IL-11 plasmid:
`linker-
`sense 5P-CGGCCAATCAGGAGGTGGAGGAGGCTCCGGAGG-
`AGGTTCTGGTGGAGGATCG-3P and linker-antisense 5P-GTAC-
`CGATCCTCCACCAGAACCTCCTCCGGAGCCTCCTCCACCT-
`CCTGATTGGCCGGCT-3P
`(pPICZKAIL-11/R-FP). All plasmids
`were sequenced using an ABI Prism automated sequencer (Perkin
`Elmer).
`
`2.3. Transfection of P. pastoris GS115 and expression of IL-11/R-FP
`Competent P. pastoris GS115 were transfected with pPICZKA (con-
`
`trol cells) or with pPICZKAIL-11/R-FP applying the LiCl method
`according to the manufacturer's instructions. Transfected cells were
`selected in YPD medium supplied with 0.1 mg/ml zeocin. Before in-
`duction of protein synthesis by supplementation of methanol, cells
`were grown in standard BMGY medium until A600 s 1.5 was reached.
`The expression conditions for IL-11/R-FP were optimized in a stand-
`ard BMGY medium containing 2% v/v methanol and 1% w/v cas-
`amino acids. For protein expression, the GS115 culture was incubated
`at 29³C for 36 h.
`
`2.4. Immunoblotting and enhanced chemiluminescence (ECL) detection
`Proteins separated by SDS-PAGE were transferred to a PVDF
`membrane by a semi-dry electroblotting procedure. PVDF mem-
`branes were blocked in a solution of 20 mM Tris-HCl (pH 7.6), 137
`mM NaCl, 0.1% Nonidet-P40 containing 10% bovine serum albumin
`and probed with antibody, followed by incubation with horseradish
`peroxidase-conjugated secondary antibody. Immunoreactive proteins
`were detected by chemiluminescence using the ECL-kit (Amersham,
`UK) following the manufacturer's instructions.
`
`2.5. Induction of K1-antichymotrypsin synthesis in HepG2 cells
`HepG2 cells were stimulated with Trx/IL-11 and sIL-11R or IL-11/
`R-FP as indicated in Fig. 3. After 18 h, cells were metabolically pulse-
`labelled with [35S]methionine/cysteine for 4 h. Induction of K1-anti-
`chymotrypsin was measured in cell culture supernatants by immuno-
`precipitation. Immunocomplexes were separated on 10% SDS poly-
`acrylamide gels, visualized by autoradiography and quanti¢ed using a
`Phosphorimager.
`
`2.6. Proliferation assay
`Ba/F3-gp130 cells (20 000 cells/well) were plated on 96 well plates
`and stimulated as indicated in Fig. 4. After 72 h of incubation, viable
`and metabolically active cells were quanti¢ed using a colorimetric
`assay based on the Cell Proliferation kit II (XTT) (Boehringer Mann-
`heim, Germany).
`
`2.7. Electrophoretic mobility shift assay (EMSA)
`Ba/F3-gp130 cells were incubated at 37³C in the presence of 0.5 nM
`IL-11/R-FP or 5 nM of each Trx/IL-11 and sIL-11R for periods of
`
`Fig. 1. Schematic representations of the fusion protein IL-11/R-FP. (A) Schematic representation of the IL-11/R-FP primary structure. The pro-
`tein starts with domains 2 and 3 of human IL-11R (Leu-109^Gly-318), followed by a 21 amino acid linker and human IL-11 (Pro-29^Leu-199).
`Due to the cloning strategy, the fusion protein contains two additional N-terminal amino acids, shown in italic letters. (B) Schematic represen-
`tation of the IL-11/R-FP tertiary structure. The linker (black line) should allow binding of IL-11 to domains 2 (D2) and 3 (D3) of IL-11R. N-
`and C-termini are indicated by N and C, respectively.
`
`

`

`S. P£anz et al./FEBS Letters 450 (1999) 117^122
`
`119
`
`time as indicated in Fig. 5. Preparation of nuclear extracts and EM-
`SAs were performed as described [20]. A double-stranded oligonucleo-
`tide derived from the c-fos promoter (m67SIE: 5P-GGGAGGGATT-
`TACGGGGAAATGCTG-3P) was used as 32P-labelled probe [21].
`The protein-DNA complexes were separated on a 4.5% polyacryl-
`amide gel containing 7.5% glycerol. The electrophoresis was per-
`formed using 0.25-fold TBE bu¡er at 260 V.
`
`3. Results
`
`3.1. Expression of IL-11/R-FP in the methylotrophic yeast
`P. pastoris
`Using a PCR-based methodology, we constructed an ex-
`pression vector encoding a fusion protein of the human IL-
`11R CBM (domains 2 and 3) and human IL-11. In order to
`allow binding of IL-11 to the binding epitope of IL-11R, both
`proteins were connected by a £exible polypeptide linker (Fig.
`1A). In Fig. 1B, the anticipated ternary structure of the fusion
`protein IL-11/R-FP is schematically represented in which the
`CBM of IL-11R accommodates IL-11. This arrangement of
`the two fusion partners is based on the analogy to the struc-
`ture of the growth hormone/growth hormone receptor com-
`plex [22]. Similarly, fusion proteins of the two IL-12 subunits
`p35 and p40 [23] as well as IL-6 and sIL-6R [16] were con-
`structed and shown to be biologically active. The methylotro-
`phic yeast P. pastoris was transfected with the vector pPICZ-
`KAIL-11/R-FP allowing the expression and secretion of the
`fusion protein (for details see Section 2). Supernatants of
`mock- or pPICZKAIL-11/R-FP-transfected cells were ana-
`lyzed by SDS-PAGE, followed by electroblotting and immu-
`nostaining with a hIL-11 polyclonal antibody (Fig. 2). A
`prominent band corresponding to a protein of approximately
`50 kDa was detected in the IL-11/R-FP supernatants but not
`
`Fig. 2. Detection of IL-11/R-FP in the supernatants of transfected
`yeasts. Supernatants of mock-transfected (control SN) and pPICZ-
`KAIL-11/R-FP-transfected yeasts (IL-11/R-FP SN) were concen-
`trated, treated with endoglycosidase H as indicated and, after sepa-
`ration of proteins on a 10% polyacrylamide SDS gel, analyzed by
`immunoblotting using a human IL-11 polyclonal antibody. Electro-
`phoretic mobilities of molecular mass marker proteins are indicated
`on the left.
`
`in the supernatants of mock-transfected cells. The electropho-
`retic mobility of the 50 kDa protein was increased after pre-
`treatment with endoglycosidase H resulting in a 44 kDa pro-
`tein. This is in good agreement with the calculated molecular
`
`Fig. 3. Induction of the K1-ACT synthesis in HepG2 cells. HepG2 cells were stimulated with equilibrated supernatants containing IL-11/R-FP
`or the combination of Trx/IL-11 and sIL-11R, each protein at molar concentrations as indicated. Cells incubated with supernatants of mock-
`transfected yeast cells served as a control. Newly synthesized proteins were metabolically labelled using [35S]methionine/cysteine. After immuno-
`precipitation from cell supernatants and separation by SDS-PAGE, radioactively-labelled K1-ACT was visualized by autoradiography (upper
`panel). Quanti¢cation of radioactivity was performed using a Phosphorimager (lower panel). The S.D.s were calculated from two independent
`experiments.
`
`

`

`120
`
`S. P£anz et al./FEBS Letters 450 (1999) 117^122
`
`mass of 43.5 kDa. Thus, the glycoprotein detected by the
`antibody can be envisaged as IL-11/R-FP. Band intensities
`of IL-11/R-FP and serial dilutions of an IL-11 preparation
`of a known concentration were quanti¢ed using a Lumi-Im-
`ager (not shown). From these data, the concentration of
`IL-11/R-FP in the yeast supernatant was calculated to be
`200 ng/ml. Yeast supernatants were equilibrated with PBS
`previous to application in the following bioassays.
`
`3.2. IL-11/R-FP e¤ciently induces acute phase protein
`synthesis in hepatoma cells
`The combination of IL-11 and sIL-11R induces the acute
`phase protein synthesis in hepatoma cells [24]. In order to
`evaluate the bioactivity of IL-11/R-FP, HepG2 cells were
`stimulated with either the combination of Trx/IL-11 and
`sIL-11R or IL-11/R-FP. Supernatants from mock-transfected
`cells served as a control. Trx/IL-11 is a well-characterized
`recombinant fusion protein of thioredoxin and IL-11 which
`has been shown to have biological properties indistinguishable
`from IL-11 wild-type [18,25]. After metabolic labelling of the
`proteins with [35S]methionine and [35S]cysteine, secreted K1-
`antichymotrypsin (K1-ACT) was precipitated from superna-
`
`Fig. 5. STAT activation in response to IL-11/R-FP and the combi-
`nation of Trx/IL-11 and sIL-11R. Ba/F3-gp130 cells were stimulated
`either with IL-11/R-FP (upper panel) at a concentration of 0.5 nM
`or the combination of Trx/IL-11 and sIL-11R (lower panel), each at
`a concentration of 5 nM. After di¡erent time points (as indicated),
`nuclear extracts were prepared and the protein concentrations were
`quanti¢ed. 10 Wg of nuclear protein was analyzed by EMSA using a
`32P-labelled m67SIE probe derived from the c-fos promotor provid-
`ing a binding site for STAT1 and STAT3. Protein-DNA complexes
`were separated by PAGE and visualized by autoradiography.
`STAT3 and STAT1 homodimers as well as STAT1/3 heterodimers
`are indicated by arrows.
`
`tants, transferred to SDS-PAGE and quanti¢ed using a Phos-
`phorimager (Fig. 3). Whereas no stimulation of the K1-ACT
`synthesis was evident at a concentration of 400 pM Trx/IL-11
`in the presence of equimolar amounts of sIL-11R, the hepa-
`toma cells responded signi¢cantly to 8 pM IL-11/R-FP. Ob-
`viously, IL-11/R-FP is a more potent agonist than the combi-
`nation of Trx/IL-11 and sIL-11R.
`
`3.3. IL-11/R-FP acts as a superagonist on Ba/F3-gp130 cells
`In order to quantify the agonistic activity of IL-11/R-FP
`more precisely, proliferation assays were performed. Ba/F3
`cells, which normally grow IL-3-dependently, are known to
`proliferate in response to various cytokines after transfection
`of the corresponding receptor chains. Ba/F3 cells transfected
`with gp130 (Ba/F3-gp130) proliferate after stimulation with
`Trx/IL-11 and sIL-11R [18]. Half-maximal proliferation was
`achieved with 6^7 nM Trx/IL-11 in the presence of equimolar
`amounts of sIL-11R (Fig. 4A). When Ba/F3-gp130 cells were
`stimulated with IL-11/R-FP, a concentration of 130^140 pM
`was su¤cient for half-maximal proliferation (Fig. 4B). Equil-
`ibrated supernatants from mock-transfected yeast cells did
`not
`induce proliferation. Thus, IL-11/R-FP is a 50-fold
`more potent agonist compared to the combination of Trx/
`IL-11 and sIL-11R. Moreover, an increased sensitivity of
`Ba/F3-gp130 cells to IL-11/R-FP cells is obvious from the
`response to low concentrations of agonist. Signi¢cant prolif-
`eration of Ba/F3-gp130 cells upon incubation with IL-11/R-
`FP is observed already at an 80-fold lower concentration
`
`Fig. 4. Proliferation of Ba/F3-gp130 cells. Ba/F3 cells stably trans-
`fected with gp130 were seeded in a 96 well plate (20 000 cells/well)
`and incubated with increasing amounts of (A) Trx/IL-11 in the pres-
`ence of sIL-11R or (B) IL-11/R-FP as indicated in the diagram.
`Open circles represent the response of cells stimulated with control
`supernatants. After 72 h, a tetrazolium compound was added as a
`substrate and incubated for 5 h at 37³C. Subsequently, the absorb-
`ance at 450 and 690 nm was measured. The di¡erence of absorban-
`ces corresponds to the number of metabolically active cells (XTT
`proliferation assay).
`
`

`

`S. P£anz et al./FEBS Letters 450 (1999) 117^122
`
`121
`
`(50 pM) than in response to Trx/IL-11 and sIL-11R (4 nM,
`Fig. 4). At high agonist concentrations, both stimuli led to an
`almost identical maximal response.
`
`3.4. STAT activation in response to IL-11/R-FP
`Signal transduction of IL-11 and related cytokines via the
`common signal transducer gp130 is mediated by the activation
`of STAT3 and to a lesser extent STAT1 [12,18]. In order to
`test in how far STAT activation is a¡ected by fusion of IL-11
`to the CBM of its receptor, Ba/F3-gp130 cells were stimulated
`either with IL-11/R-FP or the combination of Trx/IL-11 and
`sIL-11R. To obtain comparable levels of STAT activation,
`IL-11R/FP was applied at a 10-fold lower molar concentra-
`tion than Trx/IL-11 and sIL-11R. At di¡erent time points,
`nuclear extracts were prepared and analyzed for STAT bind-
`ing activity by an EMSA using a radioactively-labelled DNA
`probe that contains a binding site for STAT1 and STAT3
`homodimers as well as STAT1/STAT3 heterodimers (Fig. 5).
`Within a few minutes, both stimuli lead to a strong activation
`of STAT3 and to a much lesser extent STAT1 that peaks
`within 30 min and again after 4 h. No signi¢cant discrepancies
`in the pattern as well as the time course of STAT activation in
`response to both stimuli were observed. Thus, superagonism
`of IL-11/R-FP is not due to an altered mechanism of STAT
`activation.
`
`STAT3 that is comparable to the response to IL-11/R-FP.
`Fusion of IL-11 to its K-receptor has no consequence for
`the quality of
`the STAT signal,
`i.e.
`the time course of
`STAT activation and the ratio of STAT3 and STAT1 activa-
`tion suggesting that signaling receptor complexes of similar
`conformation are formed in response to both stimuli.
`How can the superagonistic activity of IL-11/R-FP be ex-
`plained? Receptor activation of gp130 by IL-11 is a two-step
`process requiring two binding events with di¡erent a¤nities.
`First, IL-11 binds to either the membrane-bound or the solu-
`ble IL-11R with a low a¤nity. Subsequently, the complex of
`IL-11 and IL-11R binds gp130 with a high a¤nity (KD = 250
`pM) [30] leading to gp130 oligomerization and signal trans-
`duction. Covalent linkage of IL-11 to IL-11R circumvents the
`¢rst low a¤nity binding step. Thus, the fusion protein IL-11/
`R-FP can be envisaged as an activated cytokine that directly
`binds its signal transducing receptor gp130 via the high a¤n-
`ity interaction. Since IL-11, due to its hematopoietic and anti-
`in£ammatory properties, is of high interest for various medi-
`cal applications, this superagonistic fusion protein may be of
`great value.
`
`Acknowledgements: This work was supported by Grants from the
`European Community (BIO4 CT 972010), the Deutsche Forschungs-
`gemeinschaft (Bonn) and the Fonds der Chemischen Industrie (Frank-
`furt a. M). The authors thank Stephan Hellwig for helpful discussions.
`
`4. Discussion
`
`References
`
`In this study, we describe a fusion protein consisting of the
`human IL-11R CBM fused by a £exible 21 amino acid linker
`to human IL-11. The recombinant fusion protein IL-11/R-FP
`was expressed and secreted by the methylotrophic yeast P.
`pastoris. Since treatment of the fusion protein with endogly-
`cosidase H led to a decrease in the apparent molecular mass,
`we conclude that at least one of the two potential N-glycosyl-
`ation sites within the CBM of human IL-11R is glycosylated.
`In mammalian cells, it has been shown that both sites are used
`for N-linked glycosylation [26]. The cytokine itself does not
`contain potential N-glycosylation sites [27].
`For the related human IL-6R, it has been shown that the
`CBM is su¤cient for ligand binding and mediation of signal
`transduction via gp130 [28]. Therefore, IL-11/R-FP was con-
`structed without the Ig-like domain of the IL-11R. Our results
`demonstrate that in analogy to IL-6R, the Ig-like domain of
`IL-11R is not required for biological activity. To obtain su¤-
`cient £exibility, a glycine-rich sequence was chosen to connect
`the IL-11R CBM to IL-11. Since the N-terminal 28 amino
`acids of IL-6 were found to be dispensable for receptor acti-
`vation [29], we fused a truncated IL-11, lacking the sequence
`PGPPPGP at its N-terminus. This proline-rich sequence, at
`least in Escherichia coli, seems to suppress expression of the
`cytokine (unpublished observations). Again, the bioactivity of
`IL-11/R-FP in two di¡erent cellular assays proved that this
`sequence is indeed not required for functionality.
`On HepG2 cells, IL-11/R-FP turned out to be a more po-
`tent inducer of the acute phase protein K1-antichymotrypsin
`than the combination of IL-11 and sIL-11R. In a proliferation
`assay using Ba/F3-gp130 cells, a 50-fold lower concentration
`of IL-11/R-FP led to half-maximal stimulation compared to
`the two individual components. In line with these ¢ndings is
`the observation that a 10-fold higher molar concentration of
`IL-11 and sIL-11R is required to obtain an activation of
`
`[1] Du, X. and Williams, D.A. (1997) Blood 89, 3897^3908.
`[2] Yonemura, Y., Kawakita, M., Masuda, T., Fujimoto, K., Kato,
`K. and Takatsuki, K. (1992) Exp. Hematol. 20, 1011.
`[3] Neben, S., Donaldson, D., Sie¡, C., Mauch, P., Bodine, D.,
`Ferrara, J., Yetz-Aldape, J. and Turner, K. (1994) Exp. Hematol.
`22, 353.
`[4] Tepler, I. et al. (1996) Blood 87, 3607^3614.
`[5] Trepicchio, W.L., Bozza, M., Pedneault, G. and Dorner, A.J.
`(1996) J. Immunol. 157, 3627.
`[6] Redlich, C.A., Gao, X., Rockwell, S., Kelley, M. and Elias, J.A.
`(1996) J. Immunol. 157, 1705.
`[7] Robb, L., Li, R., Hartley, L., Nandurkar, H.H., Koentgen, F.
`and Begley, C.G. (1998) Nat. Med. 4, 303^308.
`[8] Heinrich, P.C., Behrmann, I., Mu«ller-Newen, G., Schaper, F. and
`Graeve, L. (1998) Biochem. J. 334, 297^314.
`[9] Hilton, D.J. et al. (1994) EMBO J. 13, 4765^4775.
`[10] Cherel, M., Sorel, M., Lebeau, B., Dubois, S., Moreau, J.F.,
`Bataille, R., Minvielle, S. and Jacques, Y. (1995) Blood 86,
`2534^2540.
`[11] Yin, T., Taga, T., Tsang, M.L., Yasukawa, K., Kishimoto, T.
`and Yang, Y.C. (1993) J. Immunol. 151, 2555^2561.
`[12] Lu«tticken, C. et al. (1994) Science 263, 89^92.
`[13] Bazan, J.F. (1990) Proc. Natl. Acad. Sci. USA 87, 6934^6938.
`[14] Hibi, M., Murakami, M., Saito, M., Hirano, T., Taga, T. and
`Kishimoto, T. (1990) Cell 63, 1149^1157.
`[15] Karow, J., Hudson, K.R., Hall, M.A., Vernallis, A.B., Taylor,
`J.A., Gossler, A. and Heath, J.K. (1996) Biochem. J. 318, 489^
`495.
`[16] Fischer, M., Goldschmitt, J., Peschel, C., Brakenho¡, J.P.G.,
`Kallen, K.-J., Wollmer, A., Gro«tzinger, J. and Rose-John, S.
`(1997) Nat. Biotechnol. 15, 142^145.
`[17] Kobayashi, M. et al. (1989) J. Exp. Med 170, 827^845.
`[18] Dahmen, H. et al. (1998) Biochem. J. 331, 695^702.
`[19] Horsten, U., Mu«ller-Newen, G., Gerhartz, C., Wollmer, A., Wij-
`denes, J., Heinrich, P.C. and Gro«tzinger, J. (1997) J. Biol. Chem.
`272, 23748^23757.
`[20] Wegenka, U.M. et al. (1994) Mol. Cell. Biol. 14, 3186^3196.
`[21] Wagner, B.J., Hayes, T.E., Hoban, C.J. and Cochran, B.H.
`(1990) EMBO J. 9, 4477^4484.
`[22] de Vos, A.M., Ultsch, M. and Kossiako¡, A.A. (1992) Science
`255, 306^312.
`
`

`

`122
`
`S. P£anz et al./FEBS Letters 450 (1999) 117^122
`
`[23] Lieschke, G.J., Rao, P.K., Gately, M.K. and Mulligan, R.C.
`(1997) Nat. Biotechnol. 15, 35^40.
`[24] Baumann, H., Wang, Y., Morella, K.K., Lai, C.-F., Dams, H.,
`Hilton, D.J., Hawley, R.G. and Mackiewicz, A. (1996) J. Immu-
`nol. 157, 284^290.
`[25] LaVallie, E.R., DiBlasio, E.A., Kovacic, S., Grant, K.L., Schen-
`del, P.F. and McCoy, J.M. (1993) Biotechnology 11, 187^193.
`[26] Lebeau, B. et al. (1997) FEBS Lett. 407, 141^147.
`[27] Paul, S.R. et al. (1990) Proc. Natl. Acad. Sci. USA 87, 7512^
`7516.
`
`[28] Taga, T., Hibi, M., Hirata, Y., Yamasaki, K., Yasukawa, K.,
`Matsuda, T., Hirano, T. and Kishimoto, T. (1989) Cell 58,
`573^581.
`[29] Brakenho¡, J.P.J., Hart, M. and Aarden, L.A. (1989) J. Immu-
`nol. 143, 1175^1182.
`[30] Nandurkar, H.H., Hilton, D.J., Nathan, P., Wilson, T., Nicola,
`N. and Begley, C.G. (1996) Oncogene 12, 585^593.
`
`

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