WO2010108824A1 - L-plastin antibodies and their use - Google Patents

L-plastin antibodies and their use Download PDF

Info

Publication number
WO2010108824A1
WO2010108824A1 PCT/EP2010/053387 EP2010053387W WO2010108824A1 WO 2010108824 A1 WO2010108824 A1 WO 2010108824A1 EP 2010053387 W EP2010053387 W EP 2010053387W WO 2010108824 A1 WO2010108824 A1 WO 2010108824A1
Authority
WO
WIPO (PCT)
Prior art keywords
plastin
nanobodies
antibody
binding
cells
Prior art date
Application number
PCT/EP2010/053387
Other languages
French (fr)
Inventor
Jan Gettemans
Veerle Delanote
Evelyne Friederich
Original Assignee
Vib Vzw
Universiteit Gent
Universite Du Luxembourg
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Vib Vzw, Universiteit Gent, Universite Du Luxembourg filed Critical Vib Vzw
Publication of WO2010108824A1 publication Critical patent/WO2010108824A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/22Immunoglobulins specific features characterized by taxonomic origin from camelids, e.g. camel, llama or dromedary
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/569Single domain, e.g. dAb, sdAb, VHH, VNAR or nanobody®
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/77Internalization into the cell
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value

Definitions

  • the present invention relates to antibodies, especially camelid antibodies, binding to the tandem actin binding domain in L-plastin, but not to the single actin binding domains. It further relates to the use of those antibodies to limit cell invasion and metastasis in cancer. Reorganization of the actin cytoskeleton accompanies several physiological processes including cell motility, differentiation and endo-/exocytosis, but is also more and more recognized as being essential for cancer cell motility and invasion (Vignjevic & Montagnac, 2008; Yamaguchi & Condeelis, 2007).
  • Actin remodeling is controlled by activation of signaling pathways in which small-GTPases are involved resulting in activaton of downstream actin binding proteins (ABPs), of which many are expressed in mammalian (cancer) cells.
  • ABPs actin binding proteins
  • Aberrant expression of ABPs in cancer cells, as well as their direct involvement in malignant cell behavior, have been demonstrated in many studies (Hara et al, 2007; Thompson et al, 2007; Wang et al, 2004; Wang et al, 2006).
  • L-plastin also called L-fimbrin
  • the Ca2+ binding modules are made up of 2 EF-hand structures, whereas each actin binding domain is built up of two calponin homology (CH) domains (reviewed in Delanote et al, 2005b; Hara et al, 2007).
  • CH calponin homology
  • L-plastin F-actin binding and bundling activities of L-plastin are negatively regulated by Ca2+ (Namba et al, 1992).
  • L-plastin is also localized in cell structures such as microspikes and podosomes that are involved in motility, adhesion and invasion (Evans et al, 2003; Janji et al, 2006; Messier et al, 1993).
  • L-plastin shuttles between the nucleus and cytoplasm of cells in contrast to its close relative, T-plastin (Delanote et al, 2005a). In adults, L-plastin is exclusively found in hematopoietic cells.
  • L-plastin expression is significantly correlated with the progression of colorectal cancer staging (Otsuka et al, 2001 ), and has been suggested as a potential metastatic marker. Futhermore, a clear causal relationship between plastin expression modulation, either by down-regulation or overexpression, and invasion/metastasis is well established (Janji et al, 2006; Klemke et al, 2007; Zheng et al, 1997; Zheng et al, 1999).
  • RNA interference RNA interference
  • RNAi RNA interference
  • RNAi is limited by incomplete RNA cleavage, inaccessible RNA sequences, unspecific targeting and difficult to achieve for target proteins with a long half-life (Cao & Heng, 2005; Persengiev et al, 2004).
  • lntrabodies can be directed against unique protein domains. This great selectivity allows modulation of the function of one domain by the specific intrabody at any time without interfering with other activities of the target protein.
  • VHHs variable domains of heavy chain antibodies
  • VHH antigen binding regions are limited to single immunoglobulin (IgG) domains, which makes them relatively easy to clone. These robust proteins represent the smallest in vivo matured functional antigen binding domains.
  • VHHs recognize unique conformational epitopes due to their enlarged complementarity determining region 3 (CDR3) (Desmyter et al, 2001 ).
  • CDR3 complementarity determining region 3
  • Fluorescent protein-tagged nanobodies can be used as tracers in living cells (Rothbauer et al, 2006), VHHs can inhibit the proapoptotic protein bax in living cells (Gueorguieva et al, 2006) and Jobling et al., (2003) showed that VHHs can inhibit enzyme function more efficient than anti-sense approaches.
  • a first aspect of the invention is an anti-L-plastin antibody, binding both actin binding domains of said L-plastin.
  • the antibody is not binding the N-terminal end of L-Plastin, more preferably it is not binding to the EF-hand structures.
  • said antibody recognizes a conformational epitope.
  • a preferred embodiment is an antibody according to the invention whereby the binding of the antibody to L-plastin is Ca 2+ independent.
  • said antibody is a nanobody.
  • said nanobody comprises SEQ ID N°1 (Table 3), even more preferably the CDR1 loop of said nanobody consists of SEQ ID N°1.
  • said nanobody comprises, preferably consists of SEQ ID N° 2 (Table 3; Figure 1 : Nb 5).
  • a still further preferred embodiment is an antibody according to the invention whereby said antibody is expressed as an intrabody.
  • Intrabody as used here, is an antibody that is present as a functional antibody within a living cell and exerts its function and binding in said cell. Said intrabody can be obtained by expressing the coding sequence of the antibody in the cell, whereby said expression results in a functional antibody.
  • the antibody may be internalized in the cell, preferably as a fusion product, such as a fusion of said antibody with a protein transduction domain, whereby said antibody is functional after internalization in the living cell.
  • Another aspect of the invention is the use of an antibody according to the invention to suppress cell invasion and/or metastasis. Suppressing, as used here, means that there is significantly less cell invasion and/or metastasis with the use of the antibody according to the invention compared with a control where no antibody or an irrelevant antibody is used. Suppressing of cell invasion and/or metastasis can be realized in vitro or in vivo. Therefore, another aspect of the invention is the use of an antibody to treat cancer. In vivo treatment of cancer implies that the antibody can be active in the cancer cells.
  • the antibody according to the invention is fused to a protein transduction domain, and internalized in the cancer cells.
  • Protein transduction domains are known to the person skilled in the art and comprise, but are not limited to TAT (disclosed in WO0119393), pep-1 (disclosed in US2003119725) and penetratin (disclosed in US5888762).
  • the antibody according to the invention may be fused to an EGFR1 binding protein, such as an antibody directed to EGFR1. By binding to the receptor, the construct will be internalized and the antibody according to the invention will bind to L-plastin and suppress cell invasion and/or metastasis.
  • FIG. 1 Amino acid sequences of anti-human L-plastin Nanobodies. All Nanobodies originate from VHH germline sequences. The complementarity determining regions (CDRs) are shown in bold. The gaps were introduced in order to align the sequences. The differences between some of the Nanobodies are limited to only one or few amino acids.
  • Figure 2. lmmunoprecipitation of endogenous L-plastin with specific nanobodies in PC-3 cells. PC-3 cells were transfected with V5-tagged nanobodies which were recovered from the lysate by binding on anti-V5 agarose. Endogenous L-plastin co-immunoprecipitates with all L- plastin nanobodies but to varying extent (upper panel). Lower panel; western blot with mouse anti-V5 antibody showing expression of the VHHs in PC-3 cells.
  • Nb Nanobody). Members of the same nanobody family are boxed.
  • FIG. 1 Nanobodies bind different conformations of L-plastin.
  • A SDS-PAGE and Coomassie staining of purified recombinant His6-tagged L-plastin nanobodies used in gelfiltration and ITC analysis.
  • B Schematic representation summarizing thermodynamical parameters for different nanobodies used in the ITC experiments.
  • C Superdex gelfiltration of
  • L-plastin-Nb complexes Black line: L-plastin (75 ⁇ g) without Ca2+ or nanobody; light grey line:
  • Left panel addition of recombinant nanobody 5.
  • Ca2+ has no influence on binding of nanobody 5 to L-plastin.
  • Right panel addition of recombinant nanobody 9.
  • Nanobody 9 only binds L-plastin when Ca2+is included.
  • AU absorbance units.
  • Nanobodies inhibit F-actin bundling by L-plastin.
  • A Low speed co-sedimentation assay: G-actin (7 ⁇ M) was copolymerized with recombinant L-plastin (3 ⁇ M) and different recombinant nanobodies at a concentration of 30 ⁇ M. Supernatants and pellets were analyzed by SDS-PAGE. Coomassie-staining patterns of pellets and supernatants in the absence or presence of nanobodies are shown. Notice that Nbs 5-8 reverse the pattern of the amount of actin in the pellet (p) and supernatant (s).
  • B Quantification of bundling of F-actin by L-plastin and nanobodies. Amounts of actin in pellet and supernatant were quantified by densitometry of
  • FIG. 6 Binding of VHHs to L-plastin EF-hands or CH domains (epitope mapping).
  • A Schematic diagram of the different recombinant GST-tagged L-plastin fragments used in the GST-pull down assay. GST was included as a control.
  • GST glutathione S- transferase.
  • B SDS-PAGE of GST pull down assay using different recombinant GST-tagged L-plastin fragments with addition of recombinant nanobody. Nb 5 binds only with the combination of the two ABDs.
  • Nbs 2 and 9 bind with the EF-hand motifs.
  • Last lane nanobody input.
  • C SDS-PAGE analysis of chemical crosslinking (X-link) of nanobodies with GST-EF. Cross-linking was carried out with 8 ⁇ M plastin construct and 32 ⁇ M nanobody. Reaction products were analyzed by SDS-PAGE showing that nanobodies 2 and 9 bind to the EF-hands of L-plastin.
  • FIG. 7 MOM recruitment assay and effect of nanobodies on filopodia. Through binding with the MOM-tagged Nb, L-plastin localizes to mitochondria.
  • A-D Fluorescence images of fixed PC-3 cells expressing different MOM-tagged nanobodies. Staining of the nanobodies was performed with anti-V5 antibody (red). Endogenous L-plastin was visualized with anti-Lplastin antibodies (green). Expression of the negative control anti-GFP nanobody does not direct endogenous L-plastin to the mitochondria (A).
  • Figure 8 Effect of L-plastin siRNA or L-plastin intrabody expression on cell migration of PC-3 cells in a woundhealing assay.
  • Cells were transfected with (A) negative control siRNA or L- plastin siRNA. Downregulation of L-plastin by siRNA compared with the control siRNA is shown in the upper part (B) Transfection of PC-3 cells with different nanobodies. Anti-GFP nanobody is the negative control. Efficiency in wound closure as a function of time is shown. Motility as measured by the rate of migration of cells into a wound was lower in L-plastin siRNA (A) or L-plastin nanobody transfected cells (B) in comparison with the corresponding negative controls.
  • Figure 9 lmmunomodulation of L-plastin on matrigel invasion of PC-3 cells.
  • PC-3 cells were transfected with a nanobody. Different types of L-plastin intrabodies decrease invasion of PC-3 cells into matrigel.
  • Anti-GFP nanobody was used as a negative control.
  • RFU relative fluorescence units are shown in the y-axis. Each bar is the mean of 3 independent experiments ⁇ s.d..
  • Monoclonal anti-V5 antibody and HiFi Platinum Taq polymerase were purchased from Invitrogen (Merelbeke, Belgium). Polyclonal anti-V5 antibody was purchased from Sigma (St Louis, USA). An L-plastin-specific antibody was purchased from Neomarkers (Fremont, CA, USA); rabbit anti-GFP was from Santa Cruz, goat anti-GST from Amersham Biosciences. ⁇ -G- actin was purchased from Cytoskeleton (Boechout, Belgium). Lipofectamine reagent was purchased from Invitrogen. All commercial antibodies were used at the dilution recommended by the manufacturer.
  • PC-3 cells were maintained at 37 °C in a humidified 10% CO2 incubator and grown in RPMI 1640 (Gibco-BRL Life Technologies, Grand Island, NY, USA) supplemented with 10% fetal bovine serum, 100 ⁇ g/ml streptomycin and 100 IU/ml penicillin. The cells were transfected using Lipofectamine 2000 (Invitrogen) according to the manufacturer's protocol.
  • VHH antibodies were obtained in collaboration with the VIB nanobody service facility (VUB, Brussels, Belgium). An alpaca was injected subcutaneously on days 0, 7, 14, 21 , 28, 35 with -500 ⁇ g human L-plastin per injection. L-plastin was purified as described (Arpin et al., 1994). On day 39, anti-coagulated blood was collected for the analysis of the immune response and for the preparation of lymphocytes. IgG subclasses were obtained by successive affinity chromatography on protein A and protein G columns. Total serum and three purified IgG subclasses were tested by ELISA to assess the immune response to human L-plastin. IgGI and lgG2 responses were similar, while lgG3 response was the highest.
  • RNA from peripheral blood lymphocytes was used as template for first strand cDNA synthesis with oligodT primer.
  • VHH encoding sequences were amplified by PCR, digested with Pstl and Notl, and cloned into the Pstl and Notl sites of the phagemid vector pHEN4.
  • a VHH library of about 7 x 107 independent transformants was obtained. About 86% of these transformants harbored the vector with the right insert size.
  • a vector was constructed containing the transmembrane sequence of the mitochondrial outer membrane protein TOM 70 (Pfanner and Meijer, 1997; Hill et al., 1998; Schatz, 1998) according to Kaufmann et al., 2000.
  • Primers used are: fw ⁇ "l : CTC GAG ATG AAG TCA TTC ATC ACT CGT AAC AAG ACT GCA ATC CTA GC; fwd2: TAC GGT CGC TGC AAC TGG TAC CGC TAT CGG AGC TTA CTA TTA CTA TA; rev1 : GAT TGC AGT CTT GTT ACG AGT GAT GAA TGA CTT CAT CTC GAG A; rev2: ATA GTA ATA GTA AGC TCC GAT AGC GGT ACC AGT TGC AGC GAC CGT AGC TAG.
  • Nanobody expression was induced by addition of IPTG to a final concentration of 1 mM and incubated at 28°C with shaking overnight.
  • Recombinant His6 - tagged nanobodies were purified from E. coli WK6 cells by binding to Ni2+-chelating beads (Probound nickel resin, Invitrogen) and were recovered from the beads by elution with 50OmM imidazole pH 8.25. The nanobodies were further purified by gel filtration on a Superdex 75 column equilibrated in 20 mM Hepes pH 7.5, 100 mM KCI.
  • Wild-type human L-plastin, as well as its subdomains were produced in E. coli BL21 pLysS Star cells as GST-fusion proteins.
  • the L-plastin fragments were subcloned in the pGEX-2T vector and fusion proteins were cleaved with factor Xa.
  • L-plastin (75 ⁇ g) alone, or with a nanobody (50 ⁇ g) was analyzed by gelfiltration on a Superdex 200 HR 10/30 column at a constant flow rate of 0.3 ml/min.
  • the column was equilibrated in 50 mM sodium phosphate buffer pH 7.0, 100 mM KCI, 1 mM MgCI2, 1 mM ATP with 0.5 mM EGTA or 1 mM Ca2+. Proteins eluting from the column were detected by fluorescence (280 nm emission; 330 nm excitation).
  • samples were analyzed with an ApoTome Zeiss Axiovert 200 epifluorescence microscope (63 ⁇ objective) equipped with an Axiocam cooled CCD camera and processed using Axiovision software (Zeiss, Gottingen,, Germany).
  • ITC Isothermal titration calorimetry
  • L-plastin 5-1 0 ⁇ M of L-plastin was titrated with 50-80 ⁇ M of L-plastin nanobodies in Hepes buffer (2OmM) with 7OmM KCI with or without 100 ⁇ M Ca2+ in a Microcal VP-ITC MicroCalorimeter (Microcal Inc., Northampton, MA). Time between injections was set at 4 min to allow for reaching the baseline. Data were integrated and fitted using the Microcal Origin software, assuming a single class of site.
  • Nanobodies and L-plastin were subjected to high speed centrifugation at 200,000 g before initiating the experiment.
  • G-actin was polymerized overnight at 4°C or for 4 hours at room temperature in the presence of L-plastin and different nanobodies in polymerization buffer (100 mM KCI, 1 mM MgCI2, 1 mM ATP, 0.5 mM EGTA, 50 mM sodium phosphate buffer, pH 7.0) as indicated in figure legends.
  • Sedimentation of actin filaments and L-plastin was achieved by high-speed centrifugation at 200,000 xg for 30 minutes. To sediment actin bundles, samples were centrifuged for 15 minutes at 12,000 xg. Proteins in pellets and supernatants were separated by SDS-PAGE. Coomassie stained protein bands were scanned and densities were quantified with ImageJ.
  • PC-3 cells (1 x 106) were seeded into 6-well cell culture plates. The following day, cells were transfected either with pcDNA3.1 V5/His vector containing a nanobody or with an L-plastin siRNA. Fourty eight hours after transfection, a wound was made by scratching a line in a confluent monolayer. Cell debris was removed by washing the cells with serum-free medium. Migration of cells into the wound was then observed at different time points. At hourly intervals, the width of the wound was measured at the same location. Cells were followed for 24 h.
  • Example 1 L-plastin single domain VHHs (nanobodies) bind their target in vivo
  • Nanobodies were retrieved from cell extracts by use of anti-V5 antibody conjugated to agarose. Endogenous L-plastin coimmunoprecipitated with all different nanobodies, although in varying amounts, possibly due to differences in their stability and/or affinity (Fig. 2A top panel). Nanobodies 5, 6, 8 and 9 were particularly efficient in binding endogenous L-plastin. By contrast, nanobodies 1 , 3 and 4 bound with lower affinity, and they were excluded for this reason from further research. All nanobodies were expressed at comparable levels (Fig. 2B), but they did not detect endogenous or overexpressed L-plastin on western blot, suggesting that they recognize conformational epitopes.
  • Example 2 L-plastin nanobodies recognize different L-plastin conformations with different affinities
  • L-plastin harbours two closely spaced F-actin binding sites allowing the protein to crosslink actin filaments into thight bundles.
  • the effect of nanobodies on the bundling activity of L-plastin was assessed in a low-speed centrifugation assay (12,000 xg) allowing sedimentation of F- actin bundles but not single filaments (Glenney et al, 1981 ).
  • L-plastin promotes actin bundle formation resulting in high actin concentrations in the pellet fraction following sedimentation (Fig. 5A-B). For nanobodies 2 and 9, no difference in bundling of F-actin by L-plastin was detected.
  • L-plastin is characterized by a modular structure consisting of 2 amino-terminal EFhands and two tandem actinbinding domains (ABD), each divided into two calponin homology (CH) domains (de Arruda et al, 1990).
  • ABS tandem actinbinding domains
  • CH calponin homology domains
  • Nanobody 5 was found to bind to GST-full length L-plastin (GST-LPL) as well as to both ABDs combined (GST-ABD1 +2), but not to ABD1 or 2 alone or to the EFhands (GST-EF) (Fig. 6B).
  • GST-LPL GST-full length L-plastin
  • GST-EF EFhands
  • Fig. 6B nanobodies 2 and 9 both bind the EF-hands of Lplastin and full length L- plastin, but not to any other domain.
  • Other VHHs were not investigated since their similar CDRs indicate that they all presumably share the same epitope.
  • Example 6 Nanobody 5 obstructs filopodia formation of PC-3 cells
  • MOM mitochondrial outer membrane
  • a nanobody was tagged with a V5 epitope and the mitochondrial outer membrane (MOM) anchor sequence from TOM70 (McBride et al, 1992). This sequence targets proteins to the MOM, and binding partners can piggyback with the protein to this compartment.
  • Tagged nanobodies were expressed in PC-3 cells which were subsequently stained for V5 and for endogenous L-plastin.
  • a GFP-specific VHH (Rothbauer et al, 2006) was included as a negative control. No colocalization was observed between the negative control and endogenous L-plastin (Fig. 7A) which stains homogenously in the cytoplasm and nucleus as shown earlier (Delanote et al, 2005a). However, L-plastin nanobodies 2, 5 and 9 were able to recruit L-plastin to mitochondria as evidenced by their colocalization, thereby confirming a genuine intracellular interaction (Fig. 7B-D). No significant difference in efficacy was seen for nanobodies 2 and 9 (Ca2+-dependent binding to L-plastin) as compared to Nb5 (Ca2+ -independent binding).
  • Nb2 and 9 not only recognize the Ca2+-loaded form of L-plastin, but also the actin-bound conformation of L-plastin (Fig. 5A).
  • L- plastin Apart from its localization in the cytoplasm and nucleus, L- plastin is usually located at peripheral cell membrane protusions. Immunofluorescence was used to examine the effect of L-plastin nanobodies on cell morphology. We expressed nanobodies as GFP-fusion proteins in PC-3 cells and stained the cells for endogenous L- plastin. Colocalization between L-plastin and different nanobodies can be seen in specialized surface structures such as membrane ruffles and microvilli as well as in and filopodia (Fig. 7F- H).
  • Example 7 L-plastin nanobodies counteract cell migration and Matrigel invasion of PC- 3 cells As L-plastin has been linked to cell migration (Foran et al, 2006; Klemke et al, 2007; Zheng et al, 1999), we investigated if nanobodies could affect cell migration of PC-3 cells in a woundhealing assay. We first investigated if downregulation of L-plastin expression by siRNA transfection negatively regulates migration of these cells. PC-3 cells were transiently transfected with a control siRNA or an L-plastin siRNA. Fig. 8A shows that downregulation of L-plastin expression retards cell motility. We next repeated these experiments by using plastin nanobodies instead of siRNAs.
  • Fimbrin is a homolog of the cytoplasmic phosphoprotein plastin and has domains homologous with calmodulin and actin gelation proteins. Journal of Cell Biology 111 (3): 1069-1079.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Oncology (AREA)
  • Public Health (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Immunology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Peptides Or Proteins (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)

Abstract

The present invention relates to antibodies, especially camelid antibodies, binding to the tandem actin binding domain in L-plastin, but not to the single actin binding domains. It further relates to the use of those antibodies to limit cell invasion and metastasis in cancer.

Description

L-PLASTIN ANTIBODIES AND THEIR USE
The present invention relates to antibodies, especially camelid antibodies, binding to the tandem actin binding domain in L-plastin, but not to the single actin binding domains. It further relates to the use of those antibodies to limit cell invasion and metastasis in cancer. Reorganization of the actin cytoskeleton accompanies several physiological processes including cell motility, differentiation and endo-/exocytosis, but is also more and more recognized as being essential for cancer cell motility and invasion (Vignjevic & Montagnac, 2008; Yamaguchi & Condeelis, 2007). Actin remodeling is controlled by activation of signaling pathways in which small-GTPases are involved resulting in activaton of downstream actin binding proteins (ABPs), of which many are expressed in mammalian (cancer) cells. Aberrant expression of ABPs in cancer cells, as well as their direct involvement in malignant cell behavior, have been demonstrated in many studies (Hara et al, 2007; Thompson et al, 2007; Wang et al, 2004; Wang et al, 2006). L-plastin (also called L-fimbrin) is an actin bundling protein consisting of an N-terminal Ca2+ binding module, and two C-terminal actin binding domains. The Ca2+ binding modules are made up of 2 EF-hand structures, whereas each actin binding domain is built up of two calponin homology (CH) domains (reviewed in Delanote et al, 2005b; Hara et al, 2007).
F-actin binding and bundling activities of L-plastin are negatively regulated by Ca2+ (Namba et al, 1992). L-plastin is also localized in cell structures such as microspikes and podosomes that are involved in motility, adhesion and invasion (Evans et al, 2003; Janji et al, 2006; Messier et al, 1993). In addition, L-plastin shuttles between the nucleus and cytoplasm of cells in contrast to its close relative, T-plastin (Delanote et al, 2005a). In adults, L-plastin is exclusively found in hematopoietic cells. However, 'ectopic' expression of the protein is observed in epithelial and mesenchyme-derived cancer cells of solid tumors (Park et al, 1994). L-plastin expression is significantly correlated with the progression of colorectal cancer staging (Otsuka et al, 2001 ), and has been suggested as a potential metastatic marker. Futhermore, a clear causal relationship between plastin expression modulation, either by down-regulation or overexpression, and invasion/metastasis is well established (Janji et al, 2006; Klemke et al, 2007; Zheng et al, 1997; Zheng et al, 1999). However, the exact mechanism by which L-plastin interaction with the actin cytoskeleton contributes to malignant cell behavior remains unclear. Conventional antibodies have proven useful in the development of diagnostics and drugs for treatment of various diseases including cancers. By immunomodulation, the in vivo function of selected proteins can be regulated by intracellular expression of antibodies (intrabodies). Immunomodulation has advantages over RNA interference (RNAi) since it is possible to specifically target individual protein -protein interactions (Visintin et al, 2008). Furthermore, RNAi is limited by incomplete RNA cleavage, inaccessible RNA sequences, unspecific targeting and difficult to achieve for target proteins with a long half-life (Cao & Heng, 2005; Persengiev et al, 2004). lntrabodies can be directed against unique protein domains. This great selectivity allows modulation of the function of one domain by the specific intrabody at any time without interfering with other activities of the target protein.
Such in vivo "protein domain knockout" has obvious advantages over depleting the cell of the whole protein (Lichtlen et al, 2002). Intracellular expression of multidomain antibodies (single chain variable fragments or scFv) has proven problematic due to incorrect assembly of the VH and VL chains, low solubility and proteolytic degradation of the linkers (Cattaneo & Biocca, 1999; Worn et al, 2000; Worn & Pluckthun, 2001 ). A promising alternative are the naturally occurring heavy chain antibodies, which are devoid of light chains, first discovered in Camelidae (Hamers-Casterman et al, 1993). Their antigen-binding domains, called variable domains of heavy chain antibodies, VHHs or nanobodies, are extremely stable and soluble entities (Dumoulin et al, 2002; Perez et al, 2001 ). VHH antigen binding regions are limited to single immunoglobulin (IgG) domains, which makes them relatively easy to clone. These robust proteins represent the smallest in vivo matured functional antigen binding domains. In addition, VHHs recognize unique conformational epitopes due to their enlarged complementarity determining region 3 (CDR3) (Desmyter et al, 2001 ). Several applications of VHHs as intrabodies have been reported. Fluorescent protein-tagged nanobodies can be used as tracers in living cells (Rothbauer et al, 2006), VHHs can inhibit the proapoptotic protein bax in living cells (Gueorguieva et al, 2006) and Jobling et al., (2003) showed that VHHs can inhibit enzyme function more efficient than anti-sense approaches.
Classical antibodies - but no nanobodies - against L-plastin have been described (US65310653, Frederick et al., 1996; Shinomya et al., 2003; Toyooka et al., 2006). Said antibodies can be used for diagnostic purposes. However, none of those antibodies has been expressed or was used as intrabody. Moreover, none of these antibodies are capable of inhibiting actin bundling. Indeed, as these antibodies have been developed for diagnostic purposes, the fact that they can be used in Westerns blot indicates that these antibodies recognize linear epitopes, whereas the recognition of a conformational epitope is essential for the inhibition of actin bundling and repression of the biological function of L-plastin, and/or its role in cell invasion, metastasis and/or cancer. In this study, VHHs were generated against human L-plastin. Surprisingly the antibodies generated recognized conformational epitopes, and several of those antibodies could modulate cancer cell morphology, migration and invasion when expressed as intrabodies. These results substantiate not only the role of L-plastin in cancer cell motility and invasion, but point to single domain VHHs as a new instrument to probe and curb the activity of pro-invasive proteins.
A first aspect of the invention is an anti-L-plastin antibody, binding both actin binding domains of said L-plastin. Preferably, the antibody is not binding the N-terminal end of L-Plastin, more preferably it is not binding to the EF-hand structures. Preferably, said antibody recognizes a conformational epitope. A preferred embodiment is an antibody according to the invention whereby the binding of the antibody to L-plastin is Ca2+ independent.
Even more preferably, said antibody is a nanobody. Preferably, said nanobody comprises SEQ ID N°1 (Table 3), even more preferably the CDR1 loop of said nanobody consists of SEQ ID N°1. Most preferably, said nanobody comprises, preferably consists of SEQ ID N° 2 (Table 3; Figure 1 : Nb 5). A still further preferred embodiment is an antibody according to the invention whereby said antibody is expressed as an intrabody. Intrabody, as used here, is an antibody that is present as a functional antibody within a living cell and exerts its function and binding in said cell. Said intrabody can be obtained by expressing the coding sequence of the antibody in the cell, whereby said expression results in a functional antibody. Alternatively, the antibody may be internalized in the cell, preferably as a fusion product, such as a fusion of said antibody with a protein transduction domain, whereby said antibody is functional after internalization in the living cell. Another aspect of the invention is the use of an antibody according to the invention to suppress cell invasion and/or metastasis. Suppressing, as used here, means that there is significantly less cell invasion and/or metastasis with the use of the antibody according to the invention compared with a control where no antibody or an irrelevant antibody is used. Suppressing of cell invasion and/or metastasis can be realized in vitro or in vivo. Therefore, another aspect of the invention is the use of an antibody to treat cancer. In vivo treatment of cancer implies that the antibody can be active in the cancer cells. Expression of the antibody can be realized by gene therapy; methods for gene therapy are known to the person skilled in the art. Alternatively the antibody according to the invention is fused to a protein transduction domain, and internalized in the cancer cells. Protein transduction domains are known to the person skilled in the art and comprise, but are not limited to TAT (disclosed in WO0119393), pep-1 (disclosed in US2003119725) and penetratin (disclosed in US5888762). Alternatively, as L-plastin is mainly overexpressed in epithelial derived tumors, the antibody according to the invention may be fused to an EGFR1 binding protein, such as an antibody directed to EGFR1. By binding to the receptor, the construct will be internalized and the antibody according to the invention will bind to L-plastin and suppress cell invasion and/or metastasis.
Brief description of the figures
Figure 1. Amino acid sequences of anti-human L-plastin Nanobodies. All Nanobodies originate from VHH germline sequences. The complementarity determining regions (CDRs) are shown in bold. The gaps were introduced in order to align the sequences. The differences between some of the Nanobodies are limited to only one or few amino acids. Figure 2. lmmunoprecipitation of endogenous L-plastin with specific nanobodies in PC-3 cells. PC-3 cells were transfected with V5-tagged nanobodies which were recovered from the lysate by binding on anti-V5 agarose. Endogenous L-plastin co-immunoprecipitates with all L- plastin nanobodies but to varying extent (upper panel). Lower panel; western blot with mouse anti-V5 antibody showing expression of the VHHs in PC-3 cells. (Nb = Nanobody). Members of the same nanobody family are boxed.
Figure 3. Nanobodies bind different conformations of L-plastin. (A) SDS-PAGE and Coomassie staining of purified recombinant His6-tagged L-plastin nanobodies used in gelfiltration and ITC analysis. (B) Schematic representation summarizing thermodynamical parameters for different nanobodies used in the ITC experiments. (C) Superdex gelfiltration of
L-plastin-Nb complexes. Black line: L-plastin (75 μg) without Ca2+ or nanobody; light grey line:
L-plastin (75 μg) with nanobody (50 μg), without Ca2+; dark grey line: L-plastin (75 μg) including nanobody (50 μg) and Ca2+. Left panel: addition of recombinant nanobody 5. Ca2+ has no influence on binding of nanobody 5 to L-plastin. Right panel: addition of recombinant nanobody 9. Nanobody 9 only binds L-plastin when Ca2+is included. AU = absorbance units.
Figure 4. Nanobodies do not affect F-actin-L-plastin interaction in vitro. (A) SDS-PAGE analysis of a high speed co-sedimentation assay. Binding of L-plastin to F-actin: G-actin (7 μM) was copolymerized with recombinant L-plastin (3μM) and different recombinant nanobodies up to a concentration of 30 μM. Coomassie-staining patterns of pellets and supernatants in the absence or presence of nanobodies are shown for comparison. p= pellet, s=supernatant. (B)
Quantification of binding of L-plastin to F-actin in the presence or absence of nanobodies. Amou nts of L-plastin in pellets and supernatants were quantified by densitometry of
Coomassie-stained protein bands. Each bar is the mean of three independent experiments ± s.d.
Figure 5. Nanobodies inhibit F-actin bundling by L-plastin. (A) Low speed co-sedimentation assay: G-actin (7 μM) was copolymerized with recombinant L-plastin (3μM) and different recombinant nanobodies at a concentration of 30 μM. Supernatants and pellets were analyzed by SDS-PAGE. Coomassie-staining patterns of pellets and supernatants in the absence or presence of nanobodies are shown. Notice that Nbs 5-8 reverse the pattern of the amount of actin in the pellet (p) and supernatant (s). (B) Quantification of bundling of F-actin by L-plastin and nanobodies. Amounts of actin in pellet and supernatant were quantified by densitometry of
Coomassie-stained protein bands. Each bar is the mean of three independent experiments ± s.d. (C) Quantification of the amount of nanobody in the pellet fraction and supernatant by densitometry of Coomassie-stained protein bands. Co-sedimentation of nanobodies 2 and 9 is significantly higher compared to the other nanobodies, implicating that there is binding between these nanobodies and L-plastin in the absence of Ca2+ but presence of actin. (D) The low speed co-sedimentation assay as in (A) was repeated with increasing molar concentration ratios of L-plastin to nanobody as indicated in the horizontal axis. p=pellet; s=supernatant.
Figure 6. Binding of VHHs to L-plastin EF-hands or CH domains (epitope mapping). (A) Schematic diagram of the different recombinant GST-tagged L-plastin fragments used in the GST-pull down assay. GST was included as a control. LPL= L-plastin, ABD= actin binding domain, CH= calponin homology domain EF= EF-hand domain. GST=glutathione S- transferase. N= NH2-terminus; C= COOH-terminus (B) SDS-PAGE of GST pull down assay using different recombinant GST-tagged L-plastin fragments with addition of recombinant nanobody. Nb 5 binds only with the combination of the two ABDs. Nbs 2 and 9 bind with the EF-hand motifs. Last lane = nanobody input. (C). SDS-PAGE analysis of chemical crosslinking (X-link) of nanobodies with GST-EF. Cross-linking was carried out with 8μM plastin construct and 32 μM nanobody. Reaction products were analyzed by SDS-PAGE showing that nanobodies 2 and 9 bind to the EF-hands of L-plastin.
Figure 7. MOM recruitment assay and effect of nanobodies on filopodia. Through binding with the MOM-tagged Nb, L-plastin localizes to mitochondria. (A-D) Fluorescence images of fixed PC-3 cells expressing different MOM-tagged nanobodies. Staining of the nanobodies was performed with anti-V5 antibody (red). Endogenous L-plastin was visualized with anti-Lplastin antibodies (green). Expression of the negative control anti-GFP nanobody does not direct endogenous L-plastin to the mitochondria (A). (B-D) Colocalization of MOM-tagged nanobody (MOM Nb 2, 5 and 9) and endogenous L-plastin at mitochondria. Bar = 10 μm. (EH) Intracellular distribution of L-plastin and intrabodies in PC-3 cells. PC-3 cells were transfected with the empty GFP vector or with GFP-tagged L-plastin nanobodies. White arrows point at specialized surface structures. Endogenous L-plastin was visualized with anti-L-plastin antibody (red). GFP tagged nanobody is shown in green. There is colocalization in filopodia and in membrane ruffles (F-H). There is no specific localization between empty GFP and L- plastin (E). Bar = 10 μm. (I) Cells expressing nanobody 5 have less filopodia compared to cells transfected with nb 2 or nb 9. Cells with long filopodia were counted for GFP expression. Each bar is the mean of three independent experiments ± s.d.
Figure 8. Effect of L-plastin siRNA or L-plastin intrabody expression on cell migration of PC-3 cells in a woundhealing assay. Cells were transfected with (A) negative control siRNA or L- plastin siRNA. Downregulation of L-plastin by siRNA compared with the control siRNA is shown in the upper part (B) Transfection of PC-3 cells with different nanobodies. Anti-GFP nanobody is the negative control. Efficiency in wound closure as a function of time is shown. Motility as measured by the rate of migration of cells into a wound was lower in L-plastin siRNA (A) or L-plastin nanobody transfected cells (B) in comparison with the corresponding negative controls.
Figure 9. lmmunomodulation of L-plastin on matrigel invasion of PC-3 cells. PC-3 cells were transfected with a nanobody. Different types of L-plastin intrabodies decrease invasion of PC-3 cells into matrigel. Anti-GFP nanobody was used as a negative control. RFU, relative fluorescence units are shown in the y-axis. Each bar is the mean of 3 independent experiments ± s.d..
Examples
Materials and methods to the examples
Reagents and antibodies
Monoclonal anti-V5 antibody and HiFi Platinum Taq polymerase were purchased from Invitrogen (Merelbeke, Belgium). Polyclonal anti-V5 antibody was purchased from Sigma (St Louis, USA). An L-plastin-specific antibody was purchased from Neomarkers (Fremont, CA, USA); rabbit anti-GFP was from Santa Cruz, goat anti-GST from Amersham Biosciences. β-G- actin was purchased from Cytoskeleton (Boechout, Belgium). Lipofectamine reagent was purchased from Invitrogen. All commercial antibodies were used at the dilution recommended by the manufacturer.
Cell culture
PC-3 cells were maintained at 37 °C in a humidified 10% CO2 incubator and grown in RPMI 1640 (Gibco-BRL Life Technologies, Grand Island, NY, USA) supplemented with 10% fetal bovine serum, 100 μg/ml streptomycin and 100 IU/ml penicillin. The cells were transfected using Lipofectamine 2000 (Invitrogen) according to the manufacturer's protocol.
Generation of VHHs and purification
Immunization. VHH antibodies were obtained in collaboration with the VIB nanobody service facility (VUB, Brussels, Belgium). An alpaca was injected subcutaneously on days 0, 7, 14, 21 , 28, 35 with -500 μg human L-plastin per injection. L-plastin was purified as described (Arpin et al., 1994). On day 39, anti-coagulated blood was collected for the analysis of the immune response and for the preparation of lymphocytes. IgG subclasses were obtained by successive affinity chromatography on protein A and protein G columns. Total serum and three purified IgG subclasses were tested by ELISA to assess the immune response to human L-plastin. IgGI and lgG2 responses were similar, while lgG3 response was the highest.
Construction of a nanobody library. Total RNA from peripheral blood lymphocytes was used as template for first strand cDNA synthesis with oligodT primer. Using this cDNA, the VHH encoding sequences were amplified by PCR, digested with Pstl and Notl, and cloned into the Pstl and Notl sites of the phagemid vector pHEN4. A VHH library of about 7 x 107 independent transformants was obtained. About 86% of these transformants harbored the vector with the right insert size.
Isolation of anti-plastin nanobodies. Three consecutive rounds of panning were performed on solid-phase coated recombinant plastin (10 μg/well). The enrichment for antigen-specific phages was further evaluated by polyclonal phage ELISA. Antigen-specific phages were enriched after each round of panning. In total, 144 individual colonies were obtained and analyzed by ELISA for the presence of antigen-specific VHHs in their periplasmic extracts. Out of these 144 colonies, 42 colonies scored positive in this assay. Based of Hinfl RFLP analysis, 17 clones were selected for sequence analysis. These 17 clones represented 9 different nanobody cDNAs. The specificities of these VHHs were again confirmed by ELISA in an independent experiment using soluble nanobodies. cDNA cloning. The expression plasmids pHEN6c (kindly provided by Dr. Gholamreza Hassanzadeh Ghassabeh, VIB nanobody service facility), pEGFP-N1 (clontech) and pcDNA3.1A/5His/TOPO (invitrogen) were used to subclone nanobodies, which were amplified by PCR with the following primers: GFP Nb Fwd : 5' TCG AAT TCG CCA CCA TGC AGG TGC AGC TGC AGG AG 3'; GFP Nb Rev : 5' ATG ACT GAT GGA TCC TTG CTG GAG ACG GTG ACC TG 3'; pHEN6 Nb Fwd: 5' GAT GTG CAG CTG CAG GAG TCT GGA/G GGA GG 3'; pH EN6 Nb Rev: 5' GGA CTA GTG CGG CCG CTG GAG ACG GTG ACC TGG GT 3'; pcDNA3.1 V5/His Nb Fwd: 5' GGT TTA AAC GCC ACC ATG GCC CAG GTG CAG CTG CAG GAG TCT GGG 3'; pcDNA3.1 V5/His Nb Rev: 5' GCC ACT AGT TGC TCG GCC GGA ACC GTA GTC CGG 3'. The cDNA of the anti-GFP nanobody was a kind gift of Dr. Gholamreza Hassanzadeh Ghassabeh, VIB nanobody service facility.
For the analysis of intracellular recruitment of L-plastin by VHHs, a vector was constructed containing the transmembrane sequence of the mitochondrial outer membrane protein TOM 70 (Pfanner and Meijer, 1997; Hill et al., 1998; Schatz, 1998) according to Kaufmann et al., 2000. Two pairs of nucleotide sequences coding for the 29 amino acids of the TOM70 (EMBL X05585) peptide identified earlier as the mitochondrial anchor sequence (McBride et al., 1992) were annealed, phosphorylated with T4 kinase (Invitrogen), ligated with T4 ligase and inserted into the pcDNA3.1A/5His/TOPO vector to create the vector pMOM. Primers used are: fwα"l : CTC GAG ATG AAG TCA TTC ATC ACT CGT AAC AAG ACT GCA ATC CTA GC; fwd2: TAC GGT CGC TGC AAC TGG TAC CGC TAT CGG AGC TTA CTA TTA CTA TA; rev1 : GAT TGC AGT CTT GTT ACG AGT GAT GAA TGA CTT CAT CTC GAG A; rev2: ATA GTA ATA GTA AGC TCC GAT AGC GGT ACC AGT TGC AGC GAC CGT AGC TAG. This vector was then used to clone the different VHHs after amplification with the following primers: MOM V5 pcDNA3.1 Nb Fwd: 5' CGT ACC GGT GCC CAG GTG CAG CTG CAG GAG TCT GG 3'; MOM V5 pcDNA3.1 Nb Rev: 5' CGT ACC GGT CTA GCT GGA GAC GGT GAC CTG GGT C 3'. Nanobody purification. pHEN6c-nanobody His6 plasmids were transformed in E. coli WK6 and a freshly transformed colony was grown overnight in LB medium containing ampicilin (100μg/ml) and 1 % glucose. The next day, 1 ml of pre-culture was added to 330 ml of TB medium supplemented with ampicillin, 2mM MgCI2 and 0.1% glucose and grown at 37°C with shaking until an OD600 of 0.6-0.9 was reached. Nanobody expression was induced by addition of IPTG to a final concentration of 1 mM and incubated at 28°C with shaking overnight. Recombinant His6 - tagged nanobodies were purified from E. coli WK6 cells by binding to Ni2+-chelating beads (Probound nickel resin, Invitrogen) and were recovered from the beads by elution with 50OmM imidazole pH 8.25. The nanobodies were further purified by gel filtration on a Superdex 75 column equilibrated in 20 mM Hepes pH 7.5, 100 mM KCI.
Expression and purification L-plastin(domains).
Wild-type human L-plastin, as well as its subdomains were produced in E. coli BL21 pLysS Star cells as GST-fusion proteins. The L-plastin fragments were subcloned in the pGEX-2T vector and fusion proteins were cleaved with factor Xa.
lmmunoprecipitation and immunoblotting.
Cells were disrupted in ice-cold lysis buffer (20 mM Tris-HCI, pH 7.5, 137 mM NaCI, 0,5 % Triton X-100, 1 mM PMSF, and a protease inhibitor cocktail mix) followed by centrifugation (20,000 xg for 10 min at 4 °C). Protein concentrations were determined by the method of Bradford (Bradford, 1976) using bovine serum albumin as a standard. 1 mg of total protein was incubated with 15 μl V5 agarose at 4°C. The beads were washed 4 times with lysis buffer, boiled for 5 min in Laemmli sample buffer and proteins were fractionated by SDS-PAGE. Western blotting was performed as described (Towbin et al, 1992).
Gelfiltration.
L-plastin (75 μg) alone, or with a nanobody (50 μg) was analyzed by gelfiltration on a Superdex 200 HR 10/30 column at a constant flow rate of 0.3 ml/min. The column was equilibrated in 50 mM sodium phosphate buffer pH 7.0, 100 mM KCI, 1 mM MgCI2, 1 mM ATP with 0.5 mM EGTA or 1 mM Ca2+. Proteins eluting from the column were detected by fluorescence (280 nm emission; 330 nm excitation).
lmmunostaining and microscopy.
Cells were washed with PBS, fixed with 3% paraformaldehyde for 25 min at room temperature and permeabilized with 0.1 % Triton X-100 in PBS. Cells were then blocked in PBS-1% BSA for 10 min at room temperature and incubated with primary antibody for 1 h at 37 °C (anti-L-plastin antibody or anti-V5 antibody). Cells were washed in PBS, incubated with secondary antibody (Alexa 488-conjugated goat anti-mouse) and Alexa Fluor 594 phalloidin for 30 min at room temperature. Following immunostaining, samples were analyzed with an ApoTome Zeiss Axiovert 200 epifluorescence microscope (63χ objective) equipped with an Axiocam cooled CCD camera and processed using Axiovision software (Zeiss, Gottingen,, Germany).
Isothermal titration calorimetry (ITC) experiments.
5-1 0 μ M of L-plastin was titrated with 50-80 μ M of L-plastin nanobodies in Hepes buffer (2OmM) with 7OmM KCI with or without 100 μM Ca2+ in a Microcal VP-ITC MicroCalorimeter (Microcal Inc., Northampton, MA). Time between injections was set at 4 min to allow for reaching the baseline. Data were integrated and fitted using the Microcal Origin software, assuming a single class of site.
Actin binding and bundling assays.
Nanobodies and L-plastin were subjected to high speed centrifugation at 200,000 g before initiating the experiment. G-actin was polymerized overnight at 4°C or for 4 hours at room temperature in the presence of L-plastin and different nanobodies in polymerization buffer (100 mM KCI, 1 mM MgCI2, 1 mM ATP, 0.5 mM EGTA, 50 mM sodium phosphate buffer, pH 7.0) as indicated in figure legends. Sedimentation of actin filaments and L-plastin was achieved by high-speed centrifugation at 200,000 xg for 30 minutes. To sediment actin bundles, samples were centrifuged for 15 minutes at 12,000 xg. Proteins in pellets and supernatants were separated by SDS-PAGE. Coomassie stained protein bands were scanned and densities were quantified with ImageJ.
GST-pull down assay.
To 20 μl of 50% glutathione-sepharose (Amersham-biosciences), equilibrated in PBS with 0.5 % Triton, 10 μg GST-fusion protein was added. After incubation at 4 °C for 1 h, 5 μg nanobody were added and incubated for two more hours at 4 °C. Glutathione beads were then recovered by centrifugation and washed four times with binding buffer. Proteins bound to the beads were eluted by SDS sample buffer and resolved by SDSPAGE followed by Coomassie Blue staining.
Chemical crosslinking experiments. Eight μM of GST-EF hand domain was incubated with a 4* molar excess of recombinant nanobodies together in K-phosphate buffer including 75 mM NaCI and 100 μM Ca2+ during 20 min. Then, 5 mM EDC (zero cross linker) and 5 mM sulfo-NHS (stabilizer) were added and incubated during 20-40 min at RT. The mixture was then analyzed by SDS-PAGE.
Wound healing assays.
PC-3 cells (1 x 106) were seeded into 6-well cell culture plates. The following day, cells were transfected either with pcDNA3.1 V5/His vector containing a nanobody or with an L-plastin siRNA. Fourty eight hours after transfection, a wound was made by scratching a line in a confluent monolayer. Cell debris was removed by washing the cells with serum-free medium. Migration of cells into the wound was then observed at different time points. At hourly intervals, the width of the wound was measured at the same location. Cells were followed for 24 h.
Invasion assays.
Cell invasion assays were performed with the QCM™ 24-well Matrigel Invasion assay (Chemicon® International, Temecula, CA) as described (van den Abbeele et al, 2007). Data are expressed as relative fluorescence units (RFU). Assays were performed in triplicate.
Example 1: L-plastin single domain VHHs (nanobodies) bind their target in vivo
Panning of a phage display library with recombinant L-plastin yielded 9 different nanobody cDNAs. The sequences of the nanobodies are shown in Figure 1. The characteristics of the nanobodies are summarized in Table 1 and Table 2. Six of these evolved from the same B-cell clone since few amino acid substitutions were found (Nb 1 , 4, 5, 6, 7, 8). They presumably have the same epitope. The three remaining VHH cDNAs (2, 3 and 9) have different CDRs and are clonally independent. These nanobodies were subsequently evaluated in their ability to function as intrabodies. To this end, pcDNA3.1A/5-nanobody cDNAs were transfected into
PC-3 prostate cancer cells. Following expression, nanobodies were retrieved from cell extracts by use of anti-V5 antibody conjugated to agarose. Endogenous L-plastin coimmunoprecipitated with all different nanobodies, although in varying amounts, possibly due to differences in their stability and/or affinity (Fig. 2A top panel). Nanobodies 5, 6, 8 and 9 were particularly efficient in binding endogenous L-plastin. By contrast, nanobodies 1 , 3 and 4 bound with lower affinity, and they were excluded for this reason from further research. All nanobodies were expressed at comparable levels (Fig. 2B), but they did not detect endogenous or overexpressed L-plastin on western blot, suggesting that they recognize conformational epitopes.
Example 2: L-plastin nanobodies recognize different L-plastin conformations with different affinities
Selected nanobodies were recombinantly produced and purified (Fig. 2A) by immobilized metal ion affinity chromatography (IMAC) and gelfiltration. Next, we analyzed the binding characteristics with L-plastin in more detail using isothermal titration calorimetry (ITC). Table 1 summarizes all parameters obtained from ITC measurements. Surprisingly, for VHH 2 and 9 no significant binding was found although both nanobodies bind to and immunoprecipiate endogenous L-plastin (Fig. 3A). However, as L-plastin is known to bind Ca2+ through its EF- hand structures (Namba et al, 1992), we included 100 μM Ca2+ in the buffer. In both cases strong binding was observed in the presence of Ca2+, indicating that VHH 2 and 9 recognize a conformational epitope in L-plastin. For other nanobodies, the effect of Ca2+ on binding to L- plastin was only minor. The dissociation constant (Kd) ranges from -1.04 μM (Nb2) to -40 nM (Nb5). Of note, the high negative entropy (parameters for different nanobodies are summarized in Figure 3B) observed for Ca2+-loaded L-plastin and nanobodies 2 and 9 suggested that binding involves a conformational change. We observed a high change in enthalpy upon binding of these nanobodies which counteracts an unfavorable change in entropy. Large entropy variations are typical for induced fit mechanisms (Boniface et al, 1999). On the other hand, reaction parameters for binding of Nb5 to L-plastin implies a key and lock mechanism consistent with recognition of two rigid molecules that do not undergo large conformational changes upon binding (Karlsson & FaIt, 1997). The higher entropy observed in binding between nanobodies 7 and 8 to their epitope might originate from the exclusion of water molecules surrounding the hydrophobic surfaces, thus omitting the unfavorable interaction between hydrophobic surfaces and water.
Complexes between L-plastin and Nb5 or Nb9 were gel filtered on a superdex 200 HR column with or without Ca2+. Two peaks were observed for L-plastin at 85 kDa and 160 kDa, indicating that L-plastin may form dimers in vitro (Fig. 3C). For Nb5 we observed an increase in relative fluorescence of the L-plastin peaks in the presence of nanobody, irrespective of Ca2+. In addition the L-plastin-Nb5 complex eluted as a larger protein as compared to Lplastin alone (Fig. 3C, asterisks). A significant increase in relative fluorescence applied also to binding of Nb9 to L-plastin, but this occured only in the presence of Ca2+ (Fig. 3C, right). Here L-plastin also eluted as a slightly higher MW protein when Ca2+and Nb9 were present (Fig. 3C). The protein peaks were analyzed by SDS-PAGE confirming the presence of nanobody and/or L- plastin. Example 3: Nanobodies do not significantly inhibit F-actin-L-plastin binding in vitro
To study a possible effect of nanobodies on L-plastin function we first investigated if nanobodies have an effect on F-actin-L-plastin binding. Therefore we used a high speed cosedimentation assay with F-actin. G-Actin was allowed to polymerize in the presence of recombinant L-plastin, with or without different nanobodies. By centrifugation at high speed (200,000 xg), only filaments are precipitated. Pellet and supernatans were then separated and analyzed by SDS-PAGE followed by Coomassie blue staining (Fig. 4A) and band intensity was quantified. While some variation was observed in the amount of L-plastin retrieved in the pellet fraction in the presence of a nanobody (Fig. 4B), these changes were not drastic. Up to a molar ratio of 10:1 nanobody versus L-plastin, no significant inhibition in binding of Lplastin to F-actin was observed.
Example 4: Nanobodies inhibit actin bundling by L-plastin
L-plastin harbours two closely spaced F-actin binding sites allowing the protein to crosslink actin filaments into thight bundles. The effect of nanobodies on the bundling activity of L-plastin was assessed in a low-speed centrifugation assay (12,000 xg) allowing sedimentation of F- actin bundles but not single filaments (Glenney et al, 1981 ). L-plastin promotes actin bundle formation resulting in high actin concentrations in the pellet fraction following sedimentation (Fig. 5A-B). For nanobodies 2 and 9, no difference in bundling of F-actin by L-plastin was detected. However, in the case of nanobodies 5, 6, 7 and 8, originating from the same B-cell clone, a strong inhibition of bundling by L-pastin was observed (Fig. 5A-B) when the molar ratio of L-plastin:nanobody was 1 :10. The amount of nanobody cosedimenting with L-plastin in the pellet fraction was much lower for Nbs 5-8 as compared to Nb2 and 9 (Fig. 5C). As these experiments were carried out in the presence of EGTA, no binding of Nb2 and Nb9 to L-plastin was expected (Table 1 ) because L-plastin activity is inhibited at elevated Ca2+ concentrations (Namba et al, 1992). As nanobodies 2 and 9 cosedimented with L-plastin under these conditions we deduce that they can also bind to Ca2+-deprived L-plastin when L-plastin is bound to actin. Lower molar ratios of L-plastin/Nb induced similar effects. In fact, nearly complete inhibition of L-plastin bundling activity was also noticed at a plastin to nanobody molar ratio of 1 :1 (Nbs 5-8, as compared to Nbs 2 and 9). (Fig. 5D).
Example 5: Nanobodies bind different epitopes in L-plastin
L-plastin is characterized by a modular structure consisting of 2 amino-terminal EFhands and two tandem actinbinding domains (ABD), each divided into two calponin homology (CH) domains (de Arruda et al, 1990). In order to map the epitopes of selected nanobodies in L- plastin we performed an in vitro GST pull down assay. Recombinant GST-Lplastin and L- plastin modular domains (Fig. 6A) were purified and incubated with a nanobody. Following immobilization on glutathione-Sepharose beads complex formation was analyzed by SDS- PAGE. Nanobody 5 was found to bind to GST-full length L-plastin (GST-LPL) as well as to both ABDs combined (GST-ABD1 +2), but not to ABD1 or 2 alone or to the EFhands (GST-EF) (Fig. 6B). By contrast, nanobodies 2 and 9 both bind the EF-hands of Lplastin and full length L- plastin, but not to any other domain. Other VHHs were not investigated since their similar CDRs indicate that they all presumably share the same epitope. These findings were further confirmed by in vitro cross-linking experiments with the zero cross linker EDC. When either Nb2 or 9 was cross-linked to GST-EF (-35 kDa), a unique cross-linked product of -50 kDa could be observed (Fig. 6C, arrowheads), in agreement with the predicted molecular mass of the cross-linked product. This band was not observed in the GST-EF/Nb5 cross link. Thus nanobodies 2 and 9 target the same epitope. In addition we investigated whether these L- plastin subdomains encompass the entire epitope. We therefore compared the affinities of the VHHs for full length GST-tagged L-plastin with the affinity for GST-tagged L-plastin domains. This was done by ITC, and the values obtained (3.2 ± 0.2 μM for Nb2, 98.0 ± 9.4 nM for Nb9, and 46.5 ± 3.7 nM for Nb5) are very close to those obtained for full length L-plastin. These findings suggest that the entire epitope of the different nanobodies is present in the L-plastin fragments.
Example 6: Nanobody 5 obstructs filopodia formation of PC-3 cells To convincingly demonstrate an interaction in PC-3 cells between the VHH intrabodies and endogenous L-plastin, we performed a mitochondrial outer membrane (MOM) recruitment assay (Kaufmann et al, 2000). A nanobody was tagged with a V5 epitope and the mitochondrial outer membrane (MOM) anchor sequence from TOM70 (McBride et al, 1992). This sequence targets proteins to the MOM, and binding partners can piggyback with the protein to this compartment. Tagged nanobodies were expressed in PC-3 cells which were subsequently stained for V5 and for endogenous L-plastin. A GFP-specific VHH (Rothbauer et al, 2006) was included as a negative control. No colocalization was observed between the negative control and endogenous L-plastin (Fig. 7A) which stains homogenously in the cytoplasm and nucleus as shown earlier (Delanote et al, 2005a). However, L-plastin nanobodies 2, 5 and 9 were able to recruit L-plastin to mitochondria as evidenced by their colocalization, thereby confirming a genuine intracellular interaction (Fig. 7B-D). No significant difference in efficacy was seen for nanobodies 2 and 9 (Ca2+-dependent binding to L-plastin) as compared to Nb5 (Ca2+ -independent binding). This might be explained by the fact that Nb2 and 9 not only recognize the Ca2+-loaded form of L-plastin, but also the actin-bound conformation of L-plastin (Fig. 5A). Apart from its localization in the cytoplasm and nucleus, L- plastin is usually located at peripheral cell membrane protusions. Immunofluorescence was used to examine the effect of L-plastin nanobodies on cell morphology. We expressed nanobodies as GFP-fusion proteins in PC-3 cells and stained the cells for endogenous L- plastin. Colocalization between L-plastin and different nanobodies can be seen in specialized surface structures such as membrane ruffles and microvilli as well as in and filopodia (Fig. 7F- H). Similar observations were made with V5-tagged intrabodies. Strikingly, a large subpopulation of cells, particularly in the case of nanobody 5, were deprived of long filopodia (Fig. 7H). Conversely, cells with long plasma membrane cell surface projections never showed expression of nanobody. Thus Nb5 impedes formation of filopodia in PC-3 cells, which was not or much less observed with the empty GFP control vector or Nb 2 and 9, respectively. Quantification of this phenomenon by counting cells with long filopodia clearly showed a very strong effect exerted by Nb5 (Fig. 7I) which, unlike to Nb2 and 9, inhibits L- plastin mediated bundling of actin filaments.
Example 7: L-plastin nanobodies counteract cell migration and Matrigel invasion of PC- 3 cells As L-plastin has been linked to cell migration (Foran et al, 2006; Klemke et al, 2007; Zheng et al, 1999), we investigated if nanobodies could affect cell migration of PC-3 cells in a woundhealing assay. We first investigated if downregulation of L-plastin expression by siRNA transfection negatively regulates migration of these cells. PC-3 cells were transiently transfected with a control siRNA or an L-plastin siRNA. Fig. 8A shows that downregulation of L-plastin expression retards cell motility. We next repeated these experiments by using plastin nanobodies instead of siRNAs. Cells were transfected either with a negative control (anti-GFP nanobody) or one of the L-plastin nanobodies. From Fig. 8B it can be deduced that PC-3 cells expressing a nanobody showed reduced woundhealing efficiency as compared to the control (Fig. 8B). Together these findings demonstrate that depletion or immunomodulation of L- plastin slows down cell migration. It has been shown earlier that down-regulation of L-plastin reduces invasion of PC-3 cells through matrigel (Zheng et al, 1999). In view of our results we addressed the question if nanobodies could have a similar inhibitory effect on invasion of prostate cancer cells. In our setup however expression of the protein is not affected. Again, the anti-GFP nanobody was used as a negative control. PC-3 cells transiently transfected with one of the 3 different nanobodies showed a markedly diminished capacity to invade through matrigel compared to PC-3 cells transfected with the control GFP VHH (Fig. 9). This shows that intrabodies against L-plastin can counteract cell invasion similar to expression regulation of L-plastin. Table 1 : Thermodynamic parameters of Isothermal Titration Calorimetry (ITC) measurements between different nanobodies and L-plastin
Figure imgf000016_0001
* with T(Kelvin)=303,15 « ΔG = ΔH-TΔS
Table 2 €K eiΛ ievt suinmaiizing the diffeient piopeities of L-pϊasfni πaiiobodiεa
Figure imgf000017_0001
Table 3: Sequences
SEQ ID N°1: GGTFGRVGVG
SEQ ID N°2: QVQLQESGGGLVQAGDSLRLSCAVSGGTFGRVGVGWFRRAPGKEREFVAAVNWSGISTFY ADSVKGRFTISRDDNKHTVDLRMNSLKPEDSAWFCATDFRFNVPMNGTEYDYWGQGTQVT VSS References
- Arpin M, Friederich E, Algrain M, Vernel F, Louvard D (1994) Functional differences between L- and T-plastin isoforms. J Cell Biol 127(6 Pt 2): 1995-2008
Boniface JJ, Reich Z, Lyons DS, Davis MM (1999) Thermodynamics of T cell receptor binding to peptide-MHC: evidence for a general mechanism of molecular scanning.
Proc Natl Acad Sci U S A 96(20): 1 1446-11451.
Bradford MM (1976) A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding. Anal Biochem 72: 248-
254. - Cao T, Heng BC (2005) Intracellular antibodies (intrabodies) versus RNA interference for therapeutic applications. Ann CHn Lab Sci 35(3): 227-229.
Cattaneo A, Biocca S (1999) The selection of intracellular antibodies. Trends
Biotechnol 17(3): 1 15-121. de Arruda MV, Watson S, Lin CS, Leavitt J, Matsudaira P (1990) Fimbrin is a homolog of the cytoplasmic phosphoprotein plastin and has domains homologous with calmodulin and actin gelation proteins. Journal of Cell Biology 111 (3): 1069-1079.
Delanote V, Van lmpe K, De Corte V, Bruyneel E, Vetter G, Boucherie C, Mareel M,
Vandekerckhove J, Friederich E, Gettemans J (2005a) Molecular basis for dissimilar nuclear trafficking of the actin-bundling protein isoforms T- and L-plastin. Traffic 6(4): 335-345.
Delanote V, Vandekerckhove J, Gettemans J (2005b) Plastins: versatile modulators of actin organization in (patho)physiological cellular processes. Acta Pharmacol Sin 26(7):
769-779.
Desmyter A, Decanniere K, Muyldermans S, Wyns L (2001 ) Antigen specificity and high affinity binding provided by one single loop of a camel single-domain antibody. J
Biol Chem 276(28): 26285-26290.
Dumoulin M, Conrath K, Van Meirhaeghe A, Meersman F, Heremans K, Frenken LG,
Muyldermans S, Wyns L, Matagne A (2002) Single-domain antibody fragments with high conformational stability. Protein Sci 11 (3): 500-515. - Evans JG, Correia I, Krasavina O, Watson N, Matsudaira P (2003) Macrophage podosomes assemble at the leading lamella by growth and fragmentation. J Cell Biol
161 (4): 697-705.
- Foran E, McWilliam P, Kelleher D, Croke DT, Long A (2006) The leukocyte protein Lplastin induces proliferation, invasion and loss of E-cadherin expression in colon cancer cells, lnt J Cancer 118(8): 2098-2104.
- Frederick, MJ, Rodriguez, LV, Johnston, DA, Darnay BG, Grimm EA (1996) Characterization of the Mr 65,000 lymphokine-activated killer proteins phosphorylated after tumor target binding: evidence that pp65a and pp65b are phosphorylated forms of L-plastin. Cancer Res 56: 138-144.
Glenney JR, Jr., Kaulfus P, Matsudaira P, Weber K (1981 ) F-actin binding and bundling properties of fimbrin, a major cytoskeletal protein of microvillus core filaments. J Biol Chem 256(17): 9283-9288.
- Gueorguieva D, Li SH, Walsh N, Mukerji A, Tanha J, Pandey S (2006) Identification of single-domain, Bax-specific intrabodies that confer resistance to mammalian cells against oxidative-stress-induced apoptosis. Faseb Journal 20: 2636-+ Hamers-Casterman C, Atarhouch T, Muyldermans S, Robinson G, Hamers C, Songa EB, Bendahman N, Hamers R (1993) Naturally occurring antibodies devoid of light chains. Nature 363(6428): 446-448.
Hara T, Honda K, Shitashige M, Ono M, Matsuyama H, Naito K, Hirohashi S, Yamada
T (2007) Mass spectrometry analysis of the native protein complex containing actinin-4 in prostate cancer cells. MoI Cell Proteomics 6(3): 479-491. - Hill K, Model K, Ryan MT, Dietmeier K, Martin F, Wagner R, Pfanner N (1998) Tom40 forms the hydrophilic channel of the mitochondrial import pore for preproteins [see comment]. Nature 395(6701 ): 516-521
Janji B, Giganti A, De Corte V, Catillon M, Bruyneel E, Lentz D, Plastino J, Gettemans
J, Friederich E (2006) Phosphorylation on Ser5 increases the F-actin-binding activity of Lplastin and promotes its targeting to sites of actin assembly in cells. J Cell Sci 119(Pt
9): 1947-1960.
- Jobling SA, Jarman C, Teh MM, Holmberg N, Blake C, Verhoeyen ME (2003) lmmunomodulation of enzyme function in plants by single-domain antibody fragments. Nat Biotechnol 21 (1 ): 77-80. - Karlsson R, FaIt A (1997) Experimental design for kinetic analysis of protein-protein interactions with surface plasmon resonance biosensors. J Immunol Methods 200(1-2): 121-133.
Kaufmann U, Zuppinger C, Waibler Z, Rudiger M, Urbich C, Martin B, Jockusch BM, Eppenberger H, Starzinski-Powitz A (2000) The armadillo repeat region targets ARVCF to cadherin-based cellular junctions. J Ce// Sc/ 113 ( Pt 22): 4121-4135.
- Klemke M, Rafael MT, Wabnitz GH, Weschenfelder T, Konstandin MH, Garbi N, Autschbach F, Hartschuh W, Samstag Y (2007) Phosphorylation of ectopically expressed Lplastin enhances invasiveness of human melanoma cells, lnt J Cancer 120(12): 2590-2599. - Lichtlen P, Auf der Maur A, Barberis A (2002) Target validation through protein-domain knockout - applications of intracellular^ stable single-chain antibodies. TARGETS 1 (1 ): 37-44. McBride HM, Millar DG, Li JM, Shore GC (1992) A signal-anchor sequence selective for the mitochondrial outer-membrane. Journal of Cell Biology 119(6): 1451-1457. Messier JM, Shaw LM, Chafel M, Matsudaira P, Mercurio AM (1993) Fimbrin localized to an insoluble cytoskeletal fraction is constitutively phosphorylated on its headpiece domain in adherent macrophages. Cell Motil Cytoskeleton 25(3): 223-233.
Namba Y, lto M, Zu Y, Shigesada K, Maruyama K (1992) Human T cell L-plastin bundles actin filaments in a calcium-dependent manner. J Biochem 112(4): 503-507. Otsuka M, Kato M, Yoshikawa T, Chen H, Brown EJ, Masuho Y, Omata M, Seki N (2001 ) Differential expression of the L-plastin gene in human colorectal cancer progression and metastasis. Biochem Biophys Res Commun 289(4): 876-881.
Park T, Chen ZP, Leavitt J (1994) Activation of the leukocyte plastin gene occurs in most human cancer cells. Cancer Res 54(7): 1775-1781.
Perez JMJ, Renisio JG, Prompers JJ, van Platerink CJ, Cambillau C, Darbon H, Frenken LGJ (2001 ) Thermal unfolding of a llama antibody fragment: A two-state reversible process. Biochemistry 40(1 ): 74-83.
Persengiev SP, Zhu X, Green MR (2004) Nonspecific, concentration-dependent stimulation and repression of mammalian gene expression by small interfering RNAs (siRNAs). RNA 10(1 ): 12-18.
- Pfanner N, Meijer M (1997) The Tom and Tim machine. Curr Biol 7(2): R100-103 - Rothbauer U, Zolghadr K, Tillib S, Nowak D, Schermelleh L, Gahl A, Backmann N,
Conrath K, Muyldermans S, Cardoso MC, Leonhardt H (2006) Targeting and tracing antigens in live cells with fluorescent nanobodies. Nat Methods 3(11 ): 887-889.
- Schatz G (1998) Protein transport. The doors to organelles. Nature 395(6701 ): 439-440 Shinomiya H, Nagai K, Hirata H, Kobayashi N, Hasegawa H, Liu F, Sumita K, Asano Y (2003) Preparation and characterization of recombinant murine p65/L-plastin expressed in Escherichia coli and high titer antibodies against the protein. Biosci Biotechnol Biochem 67(6): 1368-1375.
- Thompson CC, Ashcroft FJ, Patel S, Saraga G, Vimalachandran D, Prime W, Campbell F, Dodson A, Jenkins RE, Lemoine NR, Crnogorac-Jurcevic T, Yin HL, Costello E (2007) Pancreatic cancer cells overexpress gelsolin family-capping proteins, which contribute to their cell motility. Gut 56(1 ): 95-106.
- Towbin H, Staehelin T, Gordon J (1992) Electrophoretic transfer of proteins from polyacrylamide gels to nitrocellulose sheets: procedure and some applications. 1979. Biotechnology 24: 145-149. - Toyooka K, Liu F, lshii M, Saito S, Kirikae T, Asano Y, Shinomiya H (2006) Generation and characterization of monoclonal antibodies that specifically recognize p65/L-plastin isoform but not T-plastin isoform. Biosci Biotechnol Biochem 70(6): 1402-1407. van den Abbeele A, De Corte V, Van lmpe K, Bruyneel E, Bottcherie C, Bracke M, Vandekerckhove J, Gettemans J (2007) Downregulation of gelsolin family proteins counteracts cancer cell invasion in vitro. Cancer Letters 255(1 ): 57-70.
- Vignjevic D, Montagnac G (2008) Reorganisation of the dendritic actin network during cancer cell migration and invasion. Semin Cancer Biol 18(1 ): 12-22.
- Visintin M, Melchionna T, Cannistraci I, Cattaneo A (2008) In vivo selection of intrabodies specifically targeting protein -protein interactions: a general platform for an "undruggable" class of disease targets. J Biotechnol 135(1 ): 1-15.
- Wang W, Goswami S, Lapidus K, Wells AL, Wyckoff JB, Sahai E, Singer RH, Segall JE, Condeelis JS (2004) Identification and testing of a gene expression signature of invasive carcinoma cells within primary mammary tumors. Cancer Res 64(23): 8585- 8594.
- Wang W, Mouneimne G, Sidani M, Wyckoff J, Chen X, Makris A, Goswami S, Bresnick AR, Condeelis JS (2006) The activity status of cofilin is directly related to invasion, intravasation, and metastasis of mammary tumors. J Cell Biol 173(3): 395-404.
- Worn A, Auf der Maur A, Escher D, Honegger A, Barberis A, Pluckthun A (2000) Correlation between in vitro stability and in vivo performance of anti-GCN4 intrabodies as cytoplasmic inhibitors. J Biol Chem 275(4): 2795-2803.
- Worn A, Pluckthun A (2001 ) Stability engineering of antibody single-chain Fv fragments. J MoI Biol 305(5): 989-1010.
- Yamaguchi H, Condeelis J (2007) Regulation of the actin cytoskeleton in cancer cell migration and invasion. Biochim Biophys Acta 1773(5): 642-652.
- Zheng J, Rudra-Ganguly N, Miller GJ, Moffatt KA, Cote RJ, Roy-Burman P (1997) Steroid hormone induction and expression patterns of L-plastin in normal and carcinomatous prostate tissues. Am J Pathol 150(6): 2009-2018.
- Zheng J, Rudra-Ganguly N, Powell WC, Roy-Burman P (1999) Suppression of prostate carcinoma cell invasion by expression of antisense L-plastin gene. Am J Pathol 155(1 ): 1 15-122.

Claims

Claims
1. An anti-L plastin antibody binding both actin binding domains.
2. The antibody of claim 1 , whereby said antibody is not binding the EF-hand structures.
3. The antibody according to claim 1 or 2, whereby said binding is Ca2+ independent.
4. The antibody according to any of the claims 1-3, whereby said antibody is a nanobody.
5. The antibody according to claim 4, whereby said antibody comprises SEQ ID N° 1.
6. The antibody according to claim 4 or 5, whereby said antibody comprises SEQ ID
N° 2.
7. The antibody according to any of the claims 1-6, whereby said antibody is an intra body.
8. The use of an antibody according to any of the claims 1-7 to suppress cell invasion and/or metastasis.
9. The use of an antibody according to claim 8, whereby said antibody is coupled to a protein transduction domain.
PCT/EP2010/053387 2009-03-23 2010-03-16 L-plastin antibodies and their use WO2010108824A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US21081909P 2009-03-23 2009-03-23
US61/210,819 2009-03-23

Publications (1)

Publication Number Publication Date
WO2010108824A1 true WO2010108824A1 (en) 2010-09-30

Family

ID=42124364

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2010/053387 WO2010108824A1 (en) 2009-03-23 2010-03-16 L-plastin antibodies and their use

Country Status (1)

Country Link
WO (1) WO2010108824A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20140363434A1 (en) * 2012-01-06 2014-12-11 Complix Nv Binding agents to intracellular target molecules
WO2019035919A1 (en) * 2017-08-16 2019-02-21 University Of Maryland, Baltimore Compositions and methods for inhibition of l-plastin activity in osteoclasts to reduce bone loss

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5310653A (en) 1989-10-24 1994-05-10 Board Of Regents, The University Of Texas System Tumor marker protein and antibodies thereto for cancer risk assessment or diagnosis
US5888762A (en) 1990-06-05 1999-03-30 Centre National De La Recherche Scientifique (Cnrs) Neurotropic growth factors comprising a homeobox peptide
WO2001019393A1 (en) 1999-09-13 2001-03-22 Cornell Research Foundation, Inc. Delivering to eucaryotic cells bacterial proteins that are secreted via type iii secretion systems
US20030119725A1 (en) 2000-07-31 2003-06-26 Gilles Divita Peptide-mediated transfection agents and methods of use

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5310653A (en) 1989-10-24 1994-05-10 Board Of Regents, The University Of Texas System Tumor marker protein and antibodies thereto for cancer risk assessment or diagnosis
US5888762A (en) 1990-06-05 1999-03-30 Centre National De La Recherche Scientifique (Cnrs) Neurotropic growth factors comprising a homeobox peptide
WO2001019393A1 (en) 1999-09-13 2001-03-22 Cornell Research Foundation, Inc. Delivering to eucaryotic cells bacterial proteins that are secreted via type iii secretion systems
US20030119725A1 (en) 2000-07-31 2003-06-26 Gilles Divita Peptide-mediated transfection agents and methods of use

Non-Patent Citations (55)

* Cited by examiner, † Cited by third party
Title
ARPIN M; FRIEDERICH E; ALGRAIN M; VERNEL F; LOUVARD D: "Functional differences between L- and T-plastin isoforms", J CELL BIOL, vol. 127, 1994, pages 1995 - 2008
BONIFACE JJ; REICH Z; LYONS DS; DAVIS MM: "Thermodynamics of T cell receptor binding to peptide-MHC: evidence for a general mechanism of molecular scanning", PROC NATL ACAD SCI U S A, vol. 96, no. 20, 1999, pages 11446 - 11451
BRADFORD MM: "A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding", ANAL BIOCHEM, vol. 72, 1976, pages 248 - 254
CAO T; HENG BC: "Intracellular antibodies (intrabodies) versus RNA interference for therapeutic applications", ANN CLIN LAB SCI, vol. 35, no. 3, 2005, pages 227 - 229
CATTANEO A; BIOCCA S: "The selection of intracellular antibodies", TRENDS BIOTECHNOL, vol. 17, no. 3, 1999, pages 115 - 121
DAVIES J ET AL: "Affinity improvement of single antibody VH domains: residues in all three hypervariable regions affect antigen binding", IMMUNOTECHNOLOGY, ELSEVIER SCIENCE PUBLISHERS BV, NL LNKD- DOI:10.1016/S1380-2933(96)00045-0, vol. 2, no. 3, 1 September 1996 (1996-09-01), pages 169 - 179, XP004070292, ISSN: 1380-2933 *
DE ARRUDA MV; WATSON S; LIN CS; LEAVITT J; MATSUDAIRA P: "Fimbrin is a homolog of the cytoplasmic phosphoprotein plastin and has domains homologous with calmodulin and actin gelation proteins", JOURNAL OF CELL BIOLOGY, vol. 111, no. 3, 1990, pages 1069 - 1079
DELANOTE V; VAN IMPE K; DE CORTE V; BRUYNEEL E; VETTER G; BOUCHERIE C; MAREEL M; VANDEKERCKHOVE J; FRIEDERICH E; GETTEMANS J: "Molecular basis for dissimilar nuclear trafficking of the actin-bundling protein isoforms T- and L-plastin", TRAFFIC, vol. 6, no. 4, 2005, pages 335 - 345
DELANOTE V; VANDEKERCKHOVE J; GETTEMANS J: "Plastins: versatile modulators of actin organization in (patho)physiological cellular processes", ACTA PHARMACOL SIN, vol. 26, no. 7, 2005, pages 769 - 779
DELANOTE VEERLE ET AL: "An alpaca single-domain antibody blocks filopodia formation by obstructing L-plastin-mediated F-actin bundling", FASEB JOURNAL, vol. 24, no. 1, January 2010 (2010-01-01), pages 105 - 118, XP002581647, ISSN: 0892-6638 *
DELANOTE VEERLE ET AL: "IMMUNOMODULATION OF CANCER CELL MIGRATION AND INVASION USING RECOMBINANT CAMELID NANOBODIES AGAINST CYTOSKELETAL PROTEINS", ANTICANCER RESEARCH, INTERNATIONAL INSTITUTE OF ANTICANCER RESEARCH, GR, vol. 28, no. 5C, 22 October 2008 (2008-10-22), pages 3291 - 3292, XP009133270, ISSN: 0250-7005 *
DESMYTER A; DECANNIERE K; MUYLDERMANS S; WYNS L: "Antigen specificity and high affinity binding provided by one single loop of a camel single-domain antibody", J BIOL CHEM, vol. 276, no. 28, 2001, pages 26285 - 26290
DUMOULIN M; CONRATH K; VAN MEIRHAEGHE A; MEERSMAN F; HEREMANS K; FRENKEN LG; MUYLDERMANS S; WYNS L; MATAGNE A: "Single-domain antibody fragments with high conformational stability", PROTEIN SCI, vol. 11, no. 3, 2002, pages 500 - 515
EVANS JG; CORREIA; KRASAVINA O; WATSON N; MATSUDAIRA P: "Macrophage podosomes assemble at the leading lamella by growth and fragmentation", J CELL BIOL, vol. 161, no. 4, 2003, pages 697 - 705
FORAN E; MCWILLIAM P; KELLEHER D; CROKE DT; LONG A: "The leukocyte protein Lplastin induces proliferation, invasion and loss of E-cadherin expression in colon cancer cells", INT J CANCER, vol. 118, no. 8, 2006, pages 2098 - 2104
FREDERICK, MJ; RODRIGUEZ, LV; JOHNSTON, DA; DARNAY BG; GRIMM EA: "Characterization of the Mr 65,000 lymphokine-activated killer proteins phosphorylated after tumor target binding: evidence that pp65a and pp65b are phosphorylated forms of L-plastin", CANCER RES, vol. 56, 1996, pages 138 - 144
GLENNEY JR, JR.; KAULFUS P; MATSUDAIRA P; WEBER K: "F-actin binding and bundling properties of fimbrin, a major cytoskeletal protein of microvillus core filaments", J BIOL CHEM, vol. 256, no. 17, 1981, pages 9283 - 9288
GUEORGUIEVA D; LI SH; WALSH N; MUKERJI A; TANHA J; PANDEY S: "Identification of single-domain, Bax-specific intrabodies that confer resistance to mammalian cells against oxidative-stress-induced apoptosis", FASEB JOURNAL, vol. 20, 2006, pages 2636
HAMERS-CASTERMAN C; ATARHOUCH T; MUYLDERMANS S; ROBINSON G; HAMERS C; SONGA EB; BENDAHMAN N; HAMERS R: "Naturally occurring antibodies devoid of light chains", NATURE, vol. 363, no. 6428, 1993, pages 446 - 448
HARA T; HONDA K; SHITASHIGE M; ONO M; MATSUYAMA H; NAITO K; HIROHASHI S; YAMADA T: "Mass spectrometry analysis of the native protein complex containing actinin-4 in prostate cancer cells", MOL CELL PROTEOMICS, vol. 6, no. 3, 2007, pages 479 - 491
HILL K; MODEL K; RYAN MT; DIETMEIER K; MARTIN F; WAGNER R; PFANNER N: "Tom40 forms the hydrophilic channel of the mitochondrial import pore for preproteins", NATURE, vol. 395, no. 6701, 1998, pages 516 - 521
INTERNATIONAL INSTITUTE OF ANTICANCER RESEARCH, XP002581651, Retrieved from the Internet <URL:http://www.iiar-anticancer.org/images/Final_programme.pdf> [retrieved on 20100510] *
JANJI B; GIGANTI A; DE CORTE V; CATILLON M; BRUYNEEL E; LENTZ D; PLASTINO J; GETTEMANS J; FRIEDERICH E: "Phosphorylation on Ser5 increases the F-actin-binding activity of Lplastin and promotes its targeting to sites of actin assembly in cells", J CELL SCI, vol. 119, 2006, pages 1947 - 1960
JOBLING SA; JARMAN C; TEH MM; HOLMBERG N; BLAKE C; VERHOEYEN ME: "Immunomodulation of enzyme function in plants by single-domain antibody fragments", NAT BIOTECHNOL, vol. 21, no. L, 2003, pages 77 - 80
KARLSSON R; FALT A: "Experimental design for kinetic analysis of protein-protein interactions with surface plasmon resonance biosensors", J IMMUNOL METHODS, vol. 200, no. 1-2, 1997, pages 121 - 133
KAUFMANN U; ZUPPINGER C; WAIBLER Z; RUDIGER M; URBICH C; MARTIN B; JOCKUSCH BM; EPPENBERGER H; STARZINSKI-POWITZ A: "The armadillo repeat region targets ARVCF to cadherin-based cellular junctions", J CELL SCI, vol. 113, 2000, pages 4121 - 4135
KLEMKE M; RAFAEL MT; WABNITZ GH; WESCHENFELDER T; KONSTANDIN MH; GARBI N; AUTSCHBACH F; HARTSCHUH W; SAMSTAG Y: "Phosphorylation of ectopically expressed Lplastin enhances invasiveness of human melanoma cells", INT J CANCER, vol. 120, no. 12, 2007, pages 2590 - 2599
LEBART M -C ET AL: "Biochemical characterization of the L-plastin-actin interaction shows a resemblance with that of alpha-actinin and allows a distinction to be made between the two actin-binding domains of the molecule.", BIOCHEMISTRY, vol. 43, no. 9, 9 March 2004 (2004-03-09), pages 2428 - 2437, XP002581646, ISSN: 0006-2960 *
LICHTLEN P; AUF DER MAUR A; BARBERIS A: "Target validation through protein-domain knockout - applications of intracellularly stable single-chain antibodies", TARGETS, vol. 1, no. 1, 2002, pages 37 - 44
MCBRIDE HM; MILLAR DG; LI JM; SHORE GC: "A signal-anchor sequence selective for the mitochondrial outer-membrane", JOURNAL OF CELL BIOLOGY, vol. 119, no. 6, 1992, pages 1451 - 1457
MESSIER JM; SHAW LM; CHAFEL M; MATSUDAIRA P; MERCURIO AM: "Fimbrin localized to an insoluble cytoskeletal fraction is constitutively phosphorylated on its headpiece domain in adherent macrophages", CELL MOTIL CYTOSKELETON, vol. 25, no. 3, 1993, pages 223 - 233
NAMBA Y; ITO M; ZU Y; SHIGESADA K; MARUYAMA K: "Human T cell L-plastin bundles actin filaments in a calcium-dependent manner", J BIOCHEM, vol. 112, no. 4, 1992, pages 503 - 507
OTSUKA M; KATO M; YOSHIKAWA T; CHEN H; BROWN EJ; MASUHO Y; OMATA M; SEKI N: "Differential expression of the L-plastin gene in human colorectal cancer progression and metastasis", BIOCHEM BIOPHYS RES COMMUN, vol. 289, no. 4, 2001, pages 876 - 881
PARK T; CHEN ZP; LEAVITT J: "Activation of the leukocyte plastin gene occurs in most human cancer cells", CANCER RES, vol. 54, no. 7, 1994, pages 1775 - 1781
PEREZ JMJ; RENISIO JG; PROMPERS JJ; VAN PLATERINK CJ; CAMBILLAU C; DARBON H; FRENKEN LGJ: "Thermal unfolding of a llama antibody fragment: A two-state reversible process", BIOCHEMISTRY, vol. 40, no. 1, 2001, pages 74 - 83
PERSENGIEV SP; ZHU X; GREEN MR: "Nonspecific, concentration-dependent stimulation and repression of mammalian gene expression by small interfering RNAs (siRNAs)", RNA, vol. 10, no. 1, 2004, pages 12 - 18
PFANNER N; MEIJER M: "The Tom and Tim machine", CURR BIOL, vol. 7, no. 2, 1997, pages R100 - 103
ROTHBAUER U; ZOLGHADR K; TILLIB S; NOWAK D; SCHERMELLEH L; GAHL A; BACKMANN N; CONRATH K; MUYLDERMANS S; CARDOSO MC: "Targeting and tracing antigens in live cells with fluorescent nanobodies", NAT METHODS, vol. 3, no. 11, 2006, pages 887 - 889
RUDIKOFF S ET AL: "Single amino acid substitution altering antigen-binding specificity", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES (PNAS), NATIONAL ACADEMY OF SCIENCE, US LNKD- DOI:10.1073/PNAS.79.6.1979, vol. 79, no. 6, 1 March 1982 (1982-03-01), pages 1979 - 1983, XP002986051, ISSN: 0027-8424 *
SCHATZ G: "Protein transport. The doors to organelles", NATURE, vol. 395, no. 6701, 1998, pages 439 - 440
SHINOMIYA H; NAGAI K; HIRATA H; KOBAYASHI N; HASEGAWA H; LIU F; SUMITA K; ASANO Y: "Preparation and characterization of recombinant murine p65/L-plastin expressed in Escherichia coli and high titer antibodies against the protein", BIOSCI BIOTECHNOL BIOCHEM, vol. 67, no. 6, 2003, pages 1368 - 1375
THOMPSON CC; ASHCROFT FJ; PATEL S; SARAGA G; VIMALACHANDRAN D; PRIME W; CAMPBELL F; DODSON A; JENKINS RE; LEMOINE NR: "Pancreatic cancer cells overexpress gelsolin family-capping proteins, which contribute to their cell motility", GUT, vol. 56, no. 1, 2007, pages 95 - 106
TOWBIN H; STAEHELIN T; GORDON J: "Electrophoretic transfer of proteins from polyacrylamide gels to nitrocellulose sheets: procedure and some applications", BIOTECHNOLOGY, vol. 24, 1979, pages 145 - 149
TOYOOKA K; LIU F; ISHII M; SAITO S; KIRIKAE T; ASANO Y; SHINOMIYA H: "Generation and characterization of monoclonal antibodies that specifically recognize p65/L-plastin isoform but not T-plastin isoform", BIOSCI BIOTECHNOL BIOCHEM, vol. 70, no. 6, 2006, pages 1402 - 1407
TOYOOKA KIMINORI ET AL: "Generation and characterization of monoclonal antibodies that specifically recognize p65/L-plastin isoform but not T-plastin isoform", BIOSCIENCE BIOTECHNOLOGY AND BIOCHEMISTRY, vol. 70, no. 6, June 2006 (2006-06-01), pages 1402 - 1407, XP002581645, ISSN: 0916-8451 *
VAN DEN ABBEELE A; DE CORTE V; VAN IMPE K; BRUYNEEL E; BOTTCHERIE C; BRACKE M; VANDEKERCKHOVE J; GETTEMANS J: "Downregulation of gelsolin family proteins counteracts cancer cell invasion in vitro", CANCER LETTERS, vol. 255, no. 1, 2007, pages 57 - 70
VIGNJEVIC D; MONTAGNAC G: "Reorganisation of the dendritic actin network during cancer cell migration and invasion", SEMIN CANCER BIOL, vol. 18, no. 1, 2008, pages 12 - 22
VISINTIN M; MELCHIONNA T; CANNISTRACI; CATTANEO A: "In vivo selection of intrabodies specifically targeting protein-protein interactions: a general platform for an "undruggable" class of disease targets", J BIOTECHNOL, vol. 135, no. 1, 2008, pages 1 - 15
WANG W; GOSWAMI S; LAPIDUS K; WELLS AL; WYCKOFF JB; SAHAI E; SINGER RH; SEGALL JE; CONDEELIS JS: "Identification and testing of a gene expression signature of invasive carcinoma cells within primary mammary tumors", CANCER RES, vol. 64, no. 23, 2004, pages 8585 - 8594
WANG W; MOUNEIMNE G; SIDANI M; WYCKOFF J; CHEN X; MAKRIS A; GOSWAMI S; BRESNICK AR; CONDEELIS JS: "The activity status of cofilin is directly related to invasion, intravasation, and metastasis of mammary tumors", J CELL BIOL, vol. 173, no. 3, 2006, pages 395 - 404
WORN A; AUF DER MAUR A; ESCHER D; HONEGGER A; BARBERIS A; PLUCKTHUN A: "Correlation between in vitro stability and in vivo performance of anti-GCN4 intrabodies as cytoplasmic inhibitors", J BIOL CHEM, vol. 275, no. 4, 2000, pages 2795 - 2803
WORN A; PLUCKTHUN A: "Stability engineering of antibody single-chain Fv fragments", J MOL BIOL, vol. 305, no. 5, 2001, pages 989 - 1010
YAMAGUCHI H; CONDEELIS J: "Regulation of the actin cytoskeleton in cancer cell migration and invasion", BIOCHIM BIOPHYS ACTA, vol. 1773, no. 5, 2007, pages 642 - 652
ZHENG J; RUDRA-GANGULY N; MILLER GJ; MOFFATT KA; COTE RJ; ROY-BURMAN P: "Steroid hormone induction and expression patterns of L-plastin in normal and carcinomatous prostate tissues", AM J PATHOL, vol. 150, no. 6, 1997, pages 2009 - 2018
ZHENG J; RUDRA-GANGULY N; POWELL WC; ROY-BURMAN P: "Suppression of prostate carcinoma cell invasion by expression of antisense L-plastin gene", AM J PATHO, vol. 155, no. 1, 1999, pages 115 - 122

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20140363434A1 (en) * 2012-01-06 2014-12-11 Complix Nv Binding agents to intracellular target molecules
WO2019035919A1 (en) * 2017-08-16 2019-02-21 University Of Maryland, Baltimore Compositions and methods for inhibition of l-plastin activity in osteoclasts to reduce bone loss
US11485762B2 (en) 2017-08-16 2022-11-01 University Of Maryland, Baltimore Compositions and methods for inhibition of L-plastin activity in osteoclasts to reduce bone loss

Similar Documents

Publication Publication Date Title
Cornfine et al. The kinesin KIF9 and reggie/flotillin proteins regulate matrix degradation by macrophage podosomes
CA2660516A1 (en) Treating or preventing cancers over-expressing reg4 or kiaa0101
US9409978B2 (en) Modulation of midbody derivatives
TW200920406A (en) EBI3, DLX5, NPTX1 and CDKN3 for target genes of lung cancer therapy and diagnosis
Delanote et al. An alpaca single‐domain antibody blocks filopodia formation by obstructing L‐plastin‐mediated F‐actin bundling
KR20160139005A (en) Monoclonal antibodies to growth and differentiation factor 15 (gdf-15), and uses thereof for treating cancer cachexia and cancer
JP2010530735A (en) Antibody binding to intracellular PRL-1 polypeptide or PRL-3 polypeptide
US20230331836A1 (en) Anti-cldn18.2 antibodies and diagnostic uses thereof
JP2010524433A (en) Human anti-CD166 antibody that binds to human tumor cells
WO2010108824A1 (en) L-plastin antibodies and their use
US20160075752A1 (en) B-type plexin antagonists and uses thereof
US20240043557A1 (en) A platform to obtain monoclonal antibodies directed against processed tumor-specific antigens
KR102264305B1 (en) Phosphorylated beta-catenin specific antibody and uses thereof
US20220106406A1 (en) Polypeptide directed against protein tyrosine phosphatase 4a proteins, and compositions and methods for use thereof
JP2020128349A (en) Cancer metastasis-inhibiting agents and methods for selecting candidate substances of cancer metastasis-inhibiting agents
KR102125703B1 (en) A Epitope of EPB41L5 and monoclonal antibody
JP7442769B2 (en) Antibodies against filamin A and their therapeutic uses
US20220154153A1 (en) New inhibitors of lrrk2/pp1 interaction
Foran et al. L-plastin Expression in HCT 116 Colorectal Cells Increases Migration and ROS in an NADPH Oxidase-Dependent Manner
Chen Functions of the Unique Region of the Desmosomal Cadherin, Desmoglein 2, in Desmoglein Internalization

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 10708995

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 10708995

Country of ref document: EP

Kind code of ref document: A1