EP1212414A2 - Polypeptides capable of interacting with the smad peptide and comprising the sequence pp(t/n)k - Google Patents
Polypeptides capable of interacting with the smad peptide and comprising the sequence pp(t/n)kInfo
- Publication number
- EP1212414A2 EP1212414A2 EP00954771A EP00954771A EP1212414A2 EP 1212414 A2 EP1212414 A2 EP 1212414A2 EP 00954771 A EP00954771 A EP 00954771A EP 00954771 A EP00954771 A EP 00954771A EP 1212414 A2 EP1212414 A2 EP 1212414A2
- Authority
- EP
- European Patent Office
- Prior art keywords
- polypeptide
- smad
- mixer
- cell
- amino acid
- Prior art date
- Legal status (The legal status 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 status listed.)
- Withdrawn
Links
Classifications
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- C07K14/46—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
- C07K14/47—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
- C07K14/4701—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
- C07K14/4702—Regulators; Modulating activity
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
Definitions
- the present invention relates to polypeptides and polynucleotides and their use in medicine and screening methods.
- TGF- ⁇ superfamily are secreted signalling molecules produced by cells to influence the behaviour of their neighbours, by regulating cell proliferation, survival, adhesion, differentiation and specification of developmental fate (Hogan et al 1994; Kingsley, 1994; Massague, 1998). These ligands bind a type II receptor which allows transphosphorylation of the type I receptor (Massague, 1998). This in turn leads to phosphorylation and activation of the receptor-activated class of Smads (R-Smads; Massague, 1998), which are responsible for transducing signals from the activated receptors to the nucleus.
- Smad proteins are a family of highly conserved, intracellular proteins that signal cellular responses downstream of transforming growth factor-beta (TGF-beta) family serine/threonine kinase receptors.
- R-Smads 2 and 3 are phosphorylated by TGF- ⁇ or activin type I receptors, whilst R-Smads 1, 5 and 8 are substrates for BMP type I receptors (Massague, 1998).
- Phosphorylation relieves an auto- inhibitory interaction of the C-terminal MH2 domain with the N-terminal MH1 domain (Hata et al 1997), allowing the R-Smads to form heteromeric complexes via their MH2 domains with members of the Smad4 class (Lagna et al 1996; Zhang et al 1997; Masuyama et al 1999; Howell et al 1999). These activated complexes translocate to the nucleus to regulate transcription of target genes (Whitman, 1998).
- Smads bind DNA very weakly alone (Shi et al 1998) and are primarily recruited to DNA by other DNA-binding transcription factors (Derynck et al 1998; Whitman, 1998), the prototype being the winged- helix/forkhead transcription factor, Fast-1 (Chen et al 1996; Chen et al 1997).
- These co-operating transcription factors are likely to be key determinants of cell type specificity of TGF- ⁇ signaling, but are mostly still poorly characterized.
- the Xenopus embryo provides an excellent system in which to elucidate the basis of specificity in TGF- ⁇ signaling pathways.
- TGF- ⁇ family members act as morphogens, playing key roles in the patterning of different tissues (Green and Smith, 1990; Gurdon et al 1994; Hogan, 1996; Whitman, 1998).
- an activin-like signal which requires the maternal transcription factor VegT for its production, is released by the vegetal hemisphere of the embryos to induce mesoderm in the overlying equatorial cells (Harland and Gerhart, 1997; Kimelman and Griffin, 1998; Zhang et al 1998).
- TGF ⁇ The TGF ⁇ superfamily, signalling pathways and likely functions have been extensively researched and reviewed.
- TGF ⁇ appears to be involved in the modulation of many biological processes and may be implicated in pathogenic conditions including tumour growth, inflammation, wound healing, scarring, fibrosis, kidney damage, for example in diabetes, and atherosclerosis.
- Proteins related to TGF ⁇ include activins, inhibins and bone morphogenetic proteins (BMPs).
- BMPs bone morphogenetic proteins
- enhancement of TGF ⁇ signalling may be beneficial, whilst in others, inhibition may be useful.
- a lack of specific small-molecule agonists or antagonists of TGF ⁇ signalling has impeded investigations, particularly in vivo.
- TGF ⁇ appears to play two contradictory roles in tumorigenesis (reviewed in Akhurst & Balmain (1999) "Genetic events and the role of TGF ⁇ in epithelial tumour progression. " J Pathol 187, 82-90). At early stages of tumorigenesis it acts as a tumour supressor through its ability to growth arrest epithelial cells, from which approximately 90% of human tumours are derived. However, at late stages of tumorigenesis, TGF ⁇ is a powerful tumour promoter, acting directly on the tumour cells themselves, promoting malignant conversion and tumour invasion, and acting indirectly by promoting angiogenesis and immunosuppression. Inhibition of the ability of TGF ⁇ to act as a tumour promoter without affecting its antiproliferative responses may therefore be desirable.
- TGF ⁇ is a dominant mediator of the pathologic extracellular matrix accumulation that characterises progression of tissue injury to end-stage organ failure.
- TGF ⁇ structure-function relationships and their relevance to models of tissue injury/ wound repair are also discussed.
- Eur Cytokine Netw 7(3), 363-74 reviews the roles of TGF- ⁇ 1, ⁇ 2 and ⁇ 3 in mammals. The author comments that they play critical roles in growth regulation and development. All three of these growth factors are secreted by most cell types, generally in a latent form, requiring activation before they can exert biological activity. This activation of latent TGF- ⁇ , which may involve plasmin, thrombospondin and possibly acidic microenvironments, appears to be a crucial regulatory step in controlling their effects.
- the TGF- ⁇ s possess three major activities: they inhibit proliferation of most cells, but can stimulate the growth of some mesenchymal cells; they exert immunosuppressive effects; and they enhance the formation of extracellular matrix.
- Two types of membrane receptors (type I and type II) possessing a serine/threonine kinase activity within their cytoplasmic domains are involved in signal transduction. Inhibition of growth by the TGF- ⁇ s stems from a blockage of the cell cycle in late Gl phase.
- the molecular participants concerned in Gl -arrest are the Retinoblastoma (Rb) protein and members of the Cyclin/Cyclin-dependent kinase/Cyclin dependent kinase inhibitor families.
- TGF- ⁇ s are involved in wound repair processes and in starting inflammatory reactions and then in their resolution.
- the latter effects of the TGF- ⁇ s derive in part from their chemotactic attraction of inflammatory cells and of fibroblasts. From gene knockout and from overexpression studies it has been shown that precise regulation of each isoform is essential for survival, at least in the long term. Several clinical applications for certain isoforms have already shown their efficacy and they have been implicated in numerous other pathological situations.
- TGF ⁇ is known to be an inhibitor of inflammation (as reviewed, for example, in Lawrence (1996) and Grande (1997), both cited above) for example from studies in which massive inflammatory lesions are seen in mice in which a TGF ⁇ gene is inactivated.
- Smad2 and Smad3 may be activated by phosphorylation near the C-terminus of the polypeptide, which induces a conformational change which exposes a binding site in the MH2 domain for transcription factors such as FASTI, FAST2 or the newly-identified partners.
- a Smad polypeptide in which the N-terminal domain is not present or is truncated may not require phosphorylation in order to expose this binding site.
- a first aspect of the invention provides a polypeptide (interacting polypeptide) capable of interacting with a Smad polypeptide wherein the interacting polypeptide comprises a Smad Interaction Motif (SIM) and is less than 32, 31, or 30 amino acids in length.
- a polypeptide interacting polypeptide
- SIM Smad Interaction Motif
- the interacting polypeptide/SIM preferably comprises the amino acid sequence PP(T/N)K or three out of four residues thereof.
- the three residues may be any three residues; ie the three residues need not be consecutive residues.
- the interacting polypeptide/SIM may comprise the amino acid sequence PPSK or PPQK ie a residue with an aliphatic hydroxyl side chain or an amide side chain may be present between the PP and K residues. It is strongly preferred that an alanine residue is not present instead of the T/N residue, ie between the PP and K residues.
- residues of the putative PP(N/T)K motif are not essential for all residues of the putative PP(N/T)K motif to be correct, as noted above and discussed further in Example 2 and the legend to Figure 15.
- either of the proline residues may be replaced, for example by an alanine residue, or the lysine residue may be replaced, for example by an alanine.
- the two prolines may not be of equal importance in that mutation of the second P to A appears to have a larger impact on the ability to bind Smad2C and activate transcription.
- An order of preference for these residues may therefore be PP>A(for example)P> > PA(for example).
- At least one P may be important in this position ie pair of residues; in the case of mixer, the PP-AA mutant does not appear to bind to Smad2C.
- the residue immediately before (ie N-terminal of) the amino acids corresponding to the sequence motif PP(T/N)K may preferably be a hydrophobic residue, for example F, M or V.
- the residue immediately after (ie C-terminal of) the amino acid sequence corresponding to the sequence motif PP(T/N)K ie at position + 1 may preferably be an S or T, which may be immediately followed by an I or V residue.
- An acidic residue (for example glutamate or aspartate) may be present at position about +3 to about + 10, preferably +4 or +5 and may be immediately followed by a hydrophobic residue, for example M, V or I.
- a proline residue may be present at a position from 5 to about 20 residues C-terminal of the amino acid sequence corresponding to the sequence motif PP(T/N)K.
- An acidic residue (for example glutamate or aspartate) immediately followed by a hydrophobic residue (for example F, Y, L) may be present at position starting about -20 or -17 to -2 relative to the amino acids corresponding to the PP(T/N)K sequence motif, preferably at -9 to -8 or -5 to -4 or -2 to -1 (ie immediately N-terminal of the PP(T/N)K sequence motif.
- a leucine residue may be present at position about -2 to -15, preferably about -5 to -10.
- the leucine residue may be the hydrophobic residue that is immediately preceded by an acidic residue, as noted above.
- this motif may extend over a stretch of more than 32, 31 or 30 amino acids. It is preferred that a SIM, for example conforming to this motif, extends over a stretch of 32, 31, 30, 29, 28, 27, 26, 25 or fewer amino acids but it will be appreciated that this is not essential.
- a polypeptide of less than 32 amino acids in length may comprise a SIM and be capable of interacting with a Smad polypeptide without comprising all elements of the D/E-Hyd-(X) n -P-P-(N/T)-K-(T/S)-(I/V)-(X) m -(D/E)-(M/V/I)-(X) k -P motif; for example the polypeptide may be a polypeptide as defined below and in claim 12 which does not have residues corresponding to the D/E-Hyd-(X) ⁇ residues, because the N-terminal amino acids of the polypeptide correspond to the PPNK motif (for example a polypeptide consisting of the amino acid sequence PPNKTITPDMN VRIPPI) .
- the polypeptide may be a polypeptide as defined below and in claim 12 which does not have residues corresponding to the D/E-Hyd-(X) ⁇ residues, because the N-terminal
- the leucine residue may be the hydrophobic residue that is immediately preceded by an acidic residue (ie D/E).
- a residue which does not match the consensus sequence indicated above is an alanine residue.
- polypeptide in connection with the interacting polypeptide includes peptides as small as the peptide PPNK or PPTK.
- the invention includes a polypeptide of less than 32, 31 or 30 amino acids in length comprising the amino acid sequence PP(T/N)K.
- a further aspect of the invention provides a polypeptide (interacting polypeptide) capable of interacting with a Smad polypeptide wherein the interacting polypeptide comprises a SIM, for example the amino acid sequence PP(T/N)K, or three out of four residues thereof, and is not full-length Xenopus or human
- FASTI or a fragment thereof, mouse FAST2, Xenopus Milk, Xenopus Mixer,
- Bixl is not an interacting polypeptide. It may be preferred that the interacting polypeptide is not Zebrafish
- the interacting polypeptide may be Xenopus
- FASTI FAST2, Milk, Mixer, Bix3, Bix2 and Bixl are well known to those skilled in the art. Mixer may also be known as Mix3 (see Mead et al (1998)).
- FAST2 is given, for example, in Liu et al (1999) Mol Cell Biol 19, 424-430 or Labbe et al (1998) Mol Cell 2, 109-120.
- the sequence for FASTI is given, for example, in Chen et al (1996) Nature 383, 691-696 and Chen et al (1997) Nature 389, 85-89.
- the sequence of human Fasti is given in Zhou et al (1998) Mol Cell 2, 121-127 and in WO98/5380.
- Xenopus Bix3 AF079561 (Tada et al (1998))
- Xenopus Bix4 AF079562 (Tada et al (1998))
- Zebrafish Mixer AF121771 (Alexander et al (1999) Dev. Biol. 215,
- the interacting polypeptide may be a transcription factor or a fragment thereof.
- the interacting polypeptide may comprise a domain that is capable of binding to a nucleic acid, preferably DNA, still more preferably double-stranded DNA, yet more preferably to DNA that forms part of a promoter region for a gene.
- the interacting polypeptide may be a fragment of a transcription factor wherein the transcription factor comprises a said domain that is capable of binding to a nucleic acid but the interacting polypeptide does not comprise the said domain.
- the interacting polypeptide may bind to the said nucleic acid with higher affinity when the interacting polypeptide is bound to one or more other polypeptides, for example one or more Smad polypeptides, than when it is not so bound.
- the interacting polypeptide may bind to the said nucleic acid as a dimer or as a heterodimer with another transcription factor ie with another polypeptide comprising a domain that is capable of binding to a nucleic acid.
- the interacting polypeptide may be capable of promoting transcription of DNA; additional polypeptides may be required for transcription to take place.
- the interacting polypeptide may comprise, for example, a winged-helix DNA binding domain or a Paired DNA binding domain or a homeodomain, for example a Paired-like homeodomain. It will be appreciated that the interacting polypeptide may comprise more than one domain that is capable of binding to a nucleic acid.
- a promoter is an expression control element formed by a DNA sequence that permits binding of RNA polymerase and transcription to occur.
- a promoter may be a region of DNA capable of controlling transcription of neighbouring DNA.
- a transcription factor that is capable of interacting with a Smad polypeptide may not be capable of binding to DNA unless it is in a complex with the Smad polypeptide, for example Smad2 or Smad3 and Smad4.
- the transcription factor may be capable of interacting (directly or indirectly) with an RNA polymerase. It is preferred that the transcription factor is capable of interacting directly with an RNA polymerase.
- FASTI and FAST2 comprise a winged-helix (also known as a Forkhead) DNA binding domain.
- Members of the Mix family which may include the chicken CMIX polypeptide (Peale et al (1998) Mech of Dev 75, 167-170 and Stein et al (1998) Mech of Dev 75, 163-165), comprise a Paired-like homeodomain (see, for example Wilson et al (l993)Genes Dev 7, 2120-2134).
- paired homeodomain transcription factor is well known to those skilled in the art. Paired homeodomain transcription factors are reviewed in, for example, Galliot et al (1999) Evolution of homeobox genes: Q50 Paired-like genes founded the Paired class Dev Genes Evol 209, 186-197, Wright et al (1989) Vertebrate homeodomain proteins: families of region-specific transcription factors Trends Biochem Sci 14, 52-56 and Dora et al (1994) Homeodomain proteins in development and therapy Pharmacol Ther 61, 155- 184.
- the homeobox domain has about 60 amino acids and consists of a helix-turn- helix motif that binds DNA by inserting the recognition helix into the major groove of the DNA and its amino-terminal arm into the adjacent minor groove.
- Representative homeobox domain are found in the Drosophila Antennapedia polypeptide and the Drosophila Paired polypeptide.
- Mammalian members of this sub-class include the Pax genes (see, for example, Adams et al (1992) Genes & Dev 6, 1589-1607).
- a second sub-class has a lysine at position 50 (K 50 type) and a third sub-class has a glutamine residue (Q 50 type) at position 50.
- K 50 type lysine at position 50
- Q 50 type glutamine residue
- the K 50 and Q 50 sub-classes do not have the prd domain.
- the Mix family of polypeptides belongs to the Q 50 class.
- the paired domain motif is a domain of 128 amino acids identified as a secondary homology region in the homeobox-containing proteins of the Drosophila paired and gooseberry genes (Bopp et al (1986) Cell 47, 1033-1040; Baumgartner et al (1987) Genes & Dev 1, 1247-1267).
- the paired domain motif encodes a DNA-binding motif (Goulding et al (1991) EMBO J 10, 1135-1147; Treisman et al (1991) Genes & Dev 5, 594-604; Chalepakis et al (1991) Cell 66, 873-884).
- paired domain proteins of vertebrates are encoded by a multigene family that has been conserved in evolution, termed the Pax gene family, as mentioned above.
- Forkhead or winged helix polypeptide is well known to those skilled in the art. Forkhead/winged helix polypeptides are reviewed, for example, in Kaufmann & Knochel (1996) Mech Dev 57, 3-20. A polypeptide may be identified as a Forkhead or winged-helix polypeptide if it comprises a domain with features of a Forkhead/winged-helix DNA binding domain.
- the Forkhead/winged-helix domain is a variant of the helix-mrn-helix motif (Brennan (1993) The winged-helix DNA-binding motif: Another helix-turn-helix takeoff Cell 74, 773-776; Clark et al (1993) Co-crystal structure of the HNF- 3/forkhead DNA-recognition motif resembles histone H5 Nature 364, 412-420).
- the forkhead/winged-helix domain is responsible for DNA-binding specificity and binds to DNA as a monomer, with two loops or wings on the C-terminal side of the helix-turn-helix.
- the forkhead domain is about 111 amino acids in length. Based on the degree of homology within the forkhead domain, the forkhead family is further split into subgroups. Over 80 genes with the conserved wing-helix forkhead motif have been identified from yeast to mammalian sources, as reviewed in Kaufmann & Knochel (1996) Mech Dev 57, 3-20. Sequence identity in the 111 amino acid domain may be more than about 50%, for example between about 70% and 95% identity; sequence identity outside this domain between forkhead family members may be much less.
- a Forkhead protein may have at least 30, 40, 50, 60, 75, 80, 85, 90 or 95% amino acid sequence identity with the FKHR Forkhead domain (Davis et al (1995) Hum Mol Genet 4, 2355-2362).
- the Forkhead domains of FASTI and FAST2 are about 40% identical to that of HNF-3 ⁇ and several other family members (Liu et al (1999).
- FASTI and FAST2 are highly homologous in the Forkhead domain and have sequence similarity in other domains. No homology to other Forkhead proteins is observed outside the Forkhead domain. FAST polypeptides therefore appear to form a sub-family of the Forkhead family.
- Xenopus FAST 3 The sequence of a novel FAST polypeptide, termed Xenopus FAST 3 is shown in Figure 13.
- the nucleotide sequence is shown in Figure 18.
- the interacting polypeptide may bind Smad2 and/or Smad3 MH2 domains but may not bind Smadl or Smad4 directly.
- the interaction may require the ⁇ -helix 2 of the MH2 domain, though the interaction may not be with the ⁇ -helix 2.
- the interaction may require regions equivalent to the regions of Smad2 indicated in Table 1 to be required for the interactions investigated.
- the Smad polypeptide with which the interacting polypeptide interacts is Smad2 or Smad3, more preferably human Smad2 or human Smad3, most preferably human Smad2.
- Smad, Smad2 and Smad3 are well know to those skilled in the art; see, for example Massague (1998); Macias-Silva et al (1996) Cell 87, 1215-1224 (human Smad2); Graff s al (1996) Cell 85, 479- 487 (Xenopus Smad2); Zhang et al (1996) Nature 383, 168-172 (human Smad3).
- Xenopus Smad3 a novel Smad polypeptide
- Figure 12 The sequence of Xenopus Smad3, a novel Smad polypeptide, is shown in Figure 12 with the sequences of human Smads 2 and 3 and Xenopus Smad2. Accession numbers for further Smad2 and Smad3 polypeptides are indicated below. Note that the ⁇ -helix2 region of these Smad2 and Smad3s, which is the region required for interaction with the SIM, is absolutely conserved with the previously characterized Xenopus Smad2 and human Smad2.
- a Smad polypeptide may have a domain recognisable as an MH2 domain.
- a tryptophan residue may be present at the residue equivalent to W274 of Xenopus Smad2 (see, for example, W097/22697).
- Smads 1, 2, 3, 4, 5 and 8 may further have a conserved domain recognisable as a MH1 domain, whilst Smads 6 and 7 may have a divergent MH1 domain.
- Smads 2 and 3 may be activated by TGF ⁇ or activin by phosphoryation at two serine residues near the C-terminus of the polypeptide. Smads 2 and 3 may be cytoplasmic until activated and then translocate to the nucleus. Smads 2 and 3 may also form a complex with Smad4 in response to ligand.
- Smad2 and 3 may be defined by the sequence in the L3 loop, which may dictate their binding to the activin and TGF ⁇ type I receptors and the sequence of the ⁇ -helix 2 that is required to bind to Fasti (see Shi et al (1997) Nature 388, 87-93 and WO99/01765), Milk and Mixer (see below).
- the Smad polypeptide may be a variant, fragment, derivative or fusion of human Smad2 or human Smad3.
- the Smad polypeptide has a greater amino acid identity with the C-terminal MH2 region, particularly the ⁇ -helix2 region (see Chen et al (1998)), of Smad2 or Smad3, for example human Smad2 or Smad3, than with the C-terminal MH2 region, particularly the ⁇ -helix2 region, of Smadl or Smad4, for example human Smadl or human Smad4.
- the MH2 domain of Xenopus Smad2 starts at amino acid W-274.
- variants of a polypeptide for example of Smad2 or Smad3, we include insertions, deletions and substitutions, either conservative or non-conservative.
- variants of the polypeptide where such changes do not substantially alter the activity of the said polypeptide, for example the ability of the Smad polypeptide to bind to an interacting polypeptide, for example a transcription factor such as FASTI, FAST2, Mixer or Milk, or another Smad polypeptide, for example Smad4.
- substitutions is intended combinations such as Gly, Ala; Val, He, Leu; Asp, Glu; Asn, Gin; Ser, Thr; Lys, Arg; and Phe, Tyr.
- the Smad polypeptide variant has an amino acid sequence which has at least 65 % identity with the amino acid sequence of Smad2 or Smad3, for example the amino acid sequence of Smad2 or Smad3 shown in Macias-Silva et al (1996) Cell 87, 1215-1224 (human Smad2); Graff et al (1996) Cell 85, 479-487 (Xenopus Smad2); Zhang et al (1996) Nature 383, 168-172 (human Smad3) or Figure 12 (Xenopus Smad3), more preferably at least 50%,
- the Smad polypeptide variant has an amino acid sequence which has at least 65 % identity with the amino acid sequence of the ⁇ - helix2 domain of Smad2 or Smad3 shown in Macias-Silva et al (1996) Cell 87, 1215-1224 (human Smad2); Graff et al (1996) Cell 85, 479-487 (Xenopus Smad2); Zhang et al (1996) Nature 383, 168-172 (human Smad3) or Figure 12 (Xenopus Smad3), more preferably at least 70% or 73%, still more preferably at least 75%, yet still more preferably at least 80%, in further preference at least 83% or 85% , in still further preference at least 90% and most preferably at least 95% or 97% identity with the amino acid sequence defined above.
- the ⁇ -helix2 domain of a Smad polypeptide may be readily identified by a person skilled in the art and as described in Chen et al (1998), for example using sequence comparisons as
- the percent sequence identity between two polypeptides may be determined using suitable computer programs, for example the GAP program of the University of Wisconsin Genetic Computing Group and it will be appreciated that percent identity is calculated in relation to polypeptides whose sequence has been aligned optimally.
- the alignment may alternatively be carried out using the Clustal W program
- Fast pairwise alignment parameters K-tuple(word) size; 1, window size; 5, gap penalty; 3, number of top diagonals; 5. Scoring method: x percent.
- “Variations” of the polypeptide also include a polypeptide in which relatively short stretches (for example 5 to 20 amino acids) have a high degree of homology (at least 80% and preferably at least 90 or 95%) with equivalent stretches of the polypeptide even though the overall homology between the two polypeptides may be much less. This is because important active or binding sites may be shared even when the general architecture of the protein is different.
- the Smad polypeptide for example Smad2 or Smad3 polypeptide is a polypeptide which consists of the amino acid sequence of the Smad2 or Smad3 polypeptide as shown in Macias-Silva et al (1996) Cell 87, 1215-1224 (human Smad2); Graff et al (1996) Cell 85, 479-487 (Xenopus Smad2); Zhang et al (1996) Nature 383, 168-172 (human Smad3) or Figure 12 (Xenopus Smad3), or naturally occurring allelic variants thereof and fusions thereof.
- a preferred fusion may be a GST fusion, for example as described in Example 1 or any other fusion described in Example 1 or a Myc fusion as described, for example, in Chen et al (1997).
- a further preferred fusion may have the tag Glu-Phe-Met-Pro-Met-Glu (termed EE-tag) or a His, HA or FLAG tag, as well known to those skilled in the art.
- the Smad polypeptide is a fragment or a fusion of a fragment of a Smad2 or Smad3 polypeptide, as shown in Macias-Silva et al (1996) (human Smad2); Graff et al (1996) (Xenopus Smad2); Zhang et al (1996) (human Smad3) or Figure 12 (Xenopus Smad3), or naturally occurring allelic variants thereof.
- the said fragment or fusion of a fragment comprises the MH2 domain, in particular the ⁇ -helix 2 domain of the said Smad2 or Smad3 polypeptide, as shown in the references indicated above, or naturally occurring allelic variants thereof.
- Particularly preferred fragments or fusions include the fragments indicated in Table 1 as capable of binding to the endogenous activity, Mixer, Milk or Fast-1, and fusions of those fragments, for example with GST.
- the Smad polypeptide is a polypeptide that is capable of binding to FASTI, FAST2, FAST3, Mixer, Milk, or Bix3.
- the capability of the said Smad polypeptide with regard to binding FASTI, FAST2, FAST3, Mixer, Milk, or Bix3 may be measured by any method of detecting/measuring a protein/protein interaction, as discussed further below and in Example 1. Suitable methods include yeast two-hybrid interactions, co-purification (for example co-immunoprecipitation or GST-pulldown assays), ELISA, co- immunoprecipitation methods and bandshift assays.
- Smad polypeptide may be phosphorylated in order for FASTI, FAST2, Mixer, Milk, or Bix3 or the said interacting polypeptide, for example FAST3, to be capable of binding to the Smad polypeptide ie for the Smad polypeptide to be activated.
- Phosphorylation of a full-length Smad polypeptide may be necessary to relieve an auto-inhibitory interaction of the C-terminal MH2 domain with the N-terminal MH1 domain, as discussed above.
- Smad fragments in which the N-terminal MH1 domain is absent, disrupted or truncated may not require phosphorylation in order for the interacting polypeptide to interact with the fragment.
- the relevant phosphorylation of Smad2 takes place on residues Ser465 and Ser467 (see, for example, Souchelnytskyi et al (1997) J Biol Chem 272, 28107-28115).
- Smad polypeptide may be the preferred fragments and fusions capable of binding to the endogenous activity, Mixer, Milk or FASTI listed in Table 1.
- the interacting polypeptide may be capable of interacting with a portion of the Smad polypeptide that is equivalent to ⁇ -helix 2 or part thereof of a full length Smad polypeptide, for example Smad2 or Smad3.
- the interacting polypeptide or PP(T/N)K- containing polypeptide is less (in order of preference) than 150, 100, 80, 70, 55, 50, 45, 40, 35, 32, 31, 30, 28 or 26 amino acids in length. It is further preferred that the interacting polypeptide is at least (in order to preference) 4, 5, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 25, 26, 28 or 30 amino acids in length or any combination of these maximum and minimum lengths. It is particularly preferred that the interacting polypeptide is between 4 and about 30, 33 or 35 amino acids in length; in further preference the interacting polypeptide is between 25 and about 30, 33 or 35 amino acids in length.
- the interacting polypeptide consists of a fragment of a naturally occurring protein such as those described below or a fusion thereof.
- the fragment of a naturally occurring protein is less than (in order of preference) 150, 100, 80, 70, 55, 50, 45, 40, 35, 33, 32, 31, 30, 28 or 26 amino acids in length.
- the fragment of a naturally occurring protein is at least (in order of preference) 4, 5, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 25, 26, 28, 30 or 33 amino acids in length.
- the interacting polypeptide further has an acidic (ie negatively charged) amino acid residue present at a position from 3 to 10, preferably 4 to 5 residues C-terminal of the amino acid sequence corresponding to the PP(T/N)K motif (and may be immediately followed by a hydrophobic residue, for example M, V or I), and/or a proline residue present at a position from 5 to 20 residues C-terminal of the amino acid sequence corresponding to the PP(T/N)K motif, as discussed above.
- the acidic (negatively charged) amino acid residue is typically a glutamate or aspartate residue.
- At least one of the two proline residues within the PP(T/N)K motif are believed to be essential for interaction with the Smad polypeptide, as discussed above and further in relation to Figure 15 below.
- Polypeptides with the sequences AANK or QTNK in place of PP(T/N)K appear not to bind Smad2.
- the downstream proline and acid (for example aspartate) residues as described above may also be important for binding.
- the residue immediately before (ie N- terminal of) the amino acid sequence PP(T/N)K may preferably be a hydrophobic residue, for example F, M or V.
- the amino acid sequence PP(T/N)K ie at position + 1 may preferably be an S or T, which may be immediately followed by an I or V residue.
- An acidic residue (for example glutamate or aspartate) immediately followed by a hydrophobic residue (for example F, Y, L) may be present at position starting about -20 to -2 relative to the amino acids corresponding to the PP(T/N)K sequence motif, preferably at -9 to -8 or -5 to -4 or -2 to -1 (ie immediately N- terminal of the PP(T/N)K sequence motif.
- a leucine residue may be present at position about -2 to -15, preferably about -5 to -10.
- the leucine residue may be the hydrophobic residue that is immediately preceded by an acidic residue, as noted above.
- the interacting polypeptide consists of or comprises the amino acid sequence PPNKTITPDMNVRIPPI or PPNKTITPDMNTIIPQI or PPNKSVFDVLTSHPGD or
- the interacting polypeptide may consist of or comprise the amino acid sequence of residues 283 to 307 of Xenopus Mixer, residues 316 to 340 of Xenopus Milk, residues 470 to 493 of Xenopus FASTI, residues 363 to 386 of mouse FAST2, residues 316 to 341 of Xenopus Bix2, resiudes 305 to 319 of Xenopus Bix 3, residues 327 to 350 of human FASTI, residues 363 to 386 of human FASTI, residues 245 to 269 of Xenopus FAST3, or the equivalent residues of the equivalent mammalian, preferably human, Mixer, Milk, Bix2/3, FASTI, FAST2 or FAST3 polypeptides or zebrafish polypeptides, for example zebrafish FASTI or Mixer.
- the interacting polypeptide or PP(T/N)K-containing polypeptide typically comprises the amino acid sequence X n [SIM; for example PP(T/N)K] Z m wherein X n represents the amino acid sequence of the consecutive n amino acids immediately N terminal to the SIM (for example amino acid sequence PP(T/N)K) in a naturally occurring polypeptide comprising a SIM, for example the amino acid sequence PP(T/N)K, for example a said naturally occurring polypeptide described above, and wherein Z m represents the amino acid sequence of the consecutive m amino acids immediately C terminal to the SIM, for example immediately C terminal to the amino acid sequence PP(T/N)K, in a naturally occurring polypeptide comprising the SIM, for example comprising the amino acid sequence PP(T/N)K, for example a said naturally occurring polypeptide described above, wherein n and m may independently be any number between 0 and 1, 5, 10, 15, 20, 25, 30, 50, 80, 100, 150, 200, 300 or 500 amino acids
- amino acid sequences X n and Z m are immediately N and C terminal, respectively, to the SIM, for example the amino acid sequence PP(T/N)K, in the same naturally occurring polypeptide.
- residue equivalent to a particular residue, for example the residue Pro291 of full-length Xenopus Mixer, is included the meaning that the amino acid residue occupies a position in the native two or three dimensional structure of a polypeptide, for example a transcription factor comprising a Paired-like homeodomain, corresponding to the position occupied by the said particular residue, for example Pro291, in the native two or three dimensional structure of full-length Xenopus Mixer. It will be appreciated that Pro291 of Xenopus full- length Mixer is located outside the Paired-like homeodomain, towards the C- terminus of the polypeptide.
- the residue equivalent to a particular residue may be identified by alignment of the sequence of the polypeptide with that of full-length Xenopus Mixer in such a way as to maximise the match between the sequences.
- the alignment may be carried out by visual inspection and/or by the use of suitable computer programs, for example the GAP program of the University of Wisconsin Genetic Computing Group, which will also allow the percent identity of the polypeptides to be calculated.
- the Align program (Pearson (1994) in: Methods in Molecular Biology, Computer Analysis of Sequence Data, Part II (Griffin, AM and Griffin, HG eds) pp 365- 389, Humana Press, Clifton).
- residues identified in this manner are also "equivalent residues”.
- the three-letter and one-letter amino acid code of the IUPAC-IUB Biochemical Nomenclature Commission is used herein.
- the sequence of polypeptides are given N-terminal to C-terminal as is conventional.
- Xaa represents any amino acid.
- the amino acids are L-amino acids, in particular it is strongly preferred that the SIM, for example a PP(T/N)K motif, consists of L-amino acid residues.
- the amino acid residues immediately flanking (such as those within 10 to 20 residues) of the SIM, for example flanking the PP(T/N)K motif are L-amino acids residues, but they may be D-amino acid residues.
- polypeptides or peptide may be made by methods well known in the art and as described below and in Example 1, for example using molecular biology methods or automated chemical peptide synthesis methods.
- Peptides may be synthesised by the Fmoc-polyamide mode of solid-phase peptide synthesis as disclosed by Lu et al (1981) J. Org. Chem. 46, 3433 and references therein. Temporary N-amino group protection is afforded by the 9- fluorenylmethyloxycarbonyl (Fmoc) group. Repetitive cleavage of this highly base-labile protecting group is effected using 20% piperidine in N,N- dimethylformamide.
- Side-chain functionalities may be protected as their butyl ethers (in the case of serine threonine and tyrosine), butyl esters (in the case of glutamic acid and aspartic acid), butyloxycarbonyl derivative (in the case of lysine and histidine), trityl derivative (in the case of cysteine) and 4-methoxy-2,3,6- trimethylbenzenesulphonyl derivative (in the case of arginine).
- glutamine or asparagine are C-terminal residues, use is made of the 4,4'- dimethoxybenzhydryl group for protection of the side chain amido functionalities.
- the solid-phase support is based on a polydimethyl-acrylamide polymer constituted from the three monomers dimethylacrylamide (backbone-monomer), bisacryloylethylene diamine (cross linker) and acryloylsarcosine methyl ester (functionalising agent).
- the peptide-to-resin cleavable linked agent used is the acid-labile 4-hydroxymethyl-phenoxy acetic acid derivative. All amino acid derivatives are added as their preformed symmetrical anhydride derivatives with the exception of asparagine and glutamine, which are added using a reversed N,N- dicyclohexyl-carbodiimide/1-hydroxybenzotriazole mediated coupling procedure.
- peptides are cleaved from the resin support with concomitant removal of side-chain protecting groups by treatment with 95 % trifluoroacetic acid containing a 50% scavenger mix.
- Scavengers commonly used are ethanedithiol, phenol, anisole and water, the exact choice depending on the constituent amino acids of the peptide being synthesised.
- Trifluoroacetic acid is removed by evaporation in vacuo, with subsequent trituration with diethyl ether affording the crude peptide.
- Any scavengers present are removed by a simple extraction procedure which on lyophilisation of the aqueous phase affords the crude peptide free of scavengers.
- Reagents for peptide synthesis are generally available from Calbiochem- Novabiochem (UK) Ltd, Nottingham NG7 2QJ, UK. Purification may be effected by any one, or a combination of, techniques such as size exclusion chromatography, ion-exchange chromatography and (principally) reverse-phase high performance liquid chromatography. Analysis of peptides may be carried out using thin layer chromatography, reverse-phase high performance liquid chromatography, amino-acid analysis after acid hydrolysis and by fast atom bombardment (FAB) mass spectrometric analysis.
- FAB fast atom bombardment
- peptidomimetic compounds may also be useful.
- polypeptide or “peptide” we include not only molecules in which amino acid residues are joined by peptide (-CO-NH-) linkages but also molecules in which the peptide bond is reversed.
- Such retro-inverso peptidomimetics may be made using methods known in the art, for example such as those described in Meziere et al (1997) J. Immunol. 159, 3230-3237, incorporated herein by reference. This approach involves making pseudopeptides containing changes involving the backbone, and not the orientation of side chains. Meziere et al (1997) show that, at least for MHC class II and T helper cell responses, these pseudopeptides are useful.
- Retro-inverse peptides which contain NH-CO bonds instead of CO-NH peptide bonds, are much more resistant to proteolysis.
- the peptide bond may be dispensed with altogether provided that an appropriate linker moiety which retains the spacing between the C ⁇ atoms of the amino acid residues is used; it is particularly preferred if the linker moiety has substantially the same charge distribution and substantially the same planarity as a peptide bond.
- the peptide may conveniently be blocked at its N- or C-terminus so as to help reduce susceptibility to exoproteolytic digestion.
- the interacting polypeptide for example which comprises the amino acid sequence PP(T/N)K may be a peptidomimetic compound, as described above.
- a further aspect of the invention provides a molecule comprising an interacting polypeptide of the invention and a further portion, wherein the said molecule is not full-length Xenopus FASTI or human FASTI or a fragment thereof, mouse
- the said further portion confers a desirable feature on the said molecule; for example, the portion may useful in detecting or isolating the molecule, or promoting cellular uptake of the molecule or the interacting polypeptide.
- the portion may be, for example, a biotin moiety, a radioactive moiety, a fluorescent moiety, for example a small fluorophore or a green fluorescent protein (GFP) fluorophore, as well known to those skilled in the art.
- GFP green fluorescent protein
- the moiety may be an immunogenic tag, for example a Myc tag, as known to those skilled in the art or may be a lipophilic molecule or polypeptide domain that is capable of promoting cellular uptake of the molecule or the interacting polypeptide, as known to those skilled in the art, for example as characterised for a Drosophila polypeptide.
- the moiety may derivable from the Antennapedia helix 3 (Derossi et al (1998) Trends Cell Biol 8, 84-87).
- a particularly preferred molecule of the invention is Biotin.Aminohexanoicacid- RQIKIWFQNRRMKWKKLLMDFNNFPPNKTITPDMNVRIPPI, discussed in Example 1.
- the first 16 amino acids are from the helix 3 of Antennapedia which allows internalization of these peptides into live cells (Derossi et al 1998); the last 25 amino acids are codons 283-307 of Mixer.
- the Antennapedia third helix is underlined.
- a further aspect of the invention provides a nucleic acid (or polynucleotide) encoding or capable of expressing an interacting polypeptide or polypeptide containing PP(T/N)K of the invention.
- a still further aspect of the invention provides a nucleic acid complementary to a nucleic acid encoding or capable of expressing a polypeptide of the invention. Methods of preparing or isolating such a nucleic acid are well known to those skilled in the art.
- the following methods of isolating a nucleic acid encoding an interacting polypeptide or polypeptide containing PP(T/N)K of the invention are given for purposes of illustration and are not considered to be exhaustive.
- the polypeptide may be cleaved, for example using trypsin, cyanogen bromide,
- V8 protease formic acid or another specific cleavage reagent.
- the digest may be chromatographed on a Vydac C18 column or subjected to SDS-PAGE to resolve the peptides.
- the N-terminal sequence of the peptides may then be determined using standard methods.
- sequences are used to isolate a nucleic acid encoding the peptide sequences using standard PCR-based strategies.
- Degenerate oligonucleotide mixtures each comprising a mixture of all possible sequences encoding a part of the peptide sequences, are designed and used as PCR primers or probes for hybridisation analysis of PCR products after Southern blotting.
- mRNA prepared from cells in which the polypeptide may be expressed is used as the template for reverse transcriptase, to prepare cDNA, which is then used as the template for the PCR reactions.
- Positive PCR fragments are subcloned and used to screen cDNA libraries to isolate a full length clone for the polypeptide.
- sequences of initial subcloned PCR fragments may be determined, and the sequence may then be extended by known PCR-based techniques to obtain a full length sequence.
- the initial PCR sequence may be used to screen electronic databases of expressed sequence tags (ESTs) or other known sequences.
- ESTs expressed sequence tags
- related sequences may be identified which may be useful in isolating a full length sequence using the two approaches described above. Sequences are determined using the Sanger dideoxy method. The encoded amino acid sequences may be deduced by routine methods.
- antibodies may be raised against the polypeptide.
- the antibodies are used to screen a ⁇ gtll expression library made from cDNA copied from mRNA from cells in which the polypeptide may be expressed.
- polypeptide encoded by the isolated nucleic acid may be desirable to express in order to determine that the polypeptide has the expected properties, for example that it is capable of interacting with a Smad polypeptide, for example Smad2 or Smad3.
- the invention also includes a polynucleotide comprising a fragment of the recombinant polynucleotide of the second aspect of the invention.
- the polynucleotide comprises a fragment which is at least 10 nucleotides in length, more preferably at least 14 nucleotides in length and still more preferably at least 18 nucleotides in length.
- Such polynucleotides are useful as PCR primers.
- the polynucleotide or recombinant polynucleotide may be DNA or RNA, preferably DNA.
- the polynucleotide may or may not contain introns in the coding sequence; preferably the polynucleotide is a cDNA.
- a “variation" of the polynucleotide includes one which is (i) usable to produce a protein or a fragment thereof which is in turn usable to prepare antibodies which specifically bind to the protein encoded by the said polynucleotide or (ii) an antisense sequence corresponding to the gene or to a variation of type (i) as just defined.
- different codons can be substituted which code for the same amino acid(s) as the original codons.
- the substitute codons may code for a different amino acid that will not affect the activity or immunogenicity of the protein or which may improve or otherwise modulate its activity or immunogenicity.
- site-directed mutagenesis or other techniques can be employed to create single or multiple mutations, such as replacements, insertions, deletions, and transpositions, as described in Botstein and Shortle, "Strategies and Applications of In Vitro Mutagenesis," Science, 229: 193-210 (1985), which is incorporated herein by reference. Since such modified polynucleotides can be obtained by the application of known techniques to the teachings contained herein, such modified polynucleotides are within the scope of the claimed invention.
- polynucleotide sequence (or fragments thereof) of the invention can be used to obtain other polynucleotide sequences that hybridise with it under conditions of high stringency.
- Such polynucleotides includes any genomic DNA.
- the polynucleotide of the invention includes polynucleotide that shows at least 55 per cent, preferably 60 per cent, and more preferably at least 70 per cent and most preferably at least 90 per cent homology with the polynucleotide identified in the method of the invention, provided that such homologous polynucleotide encodes a polypeptide which is usable in at least some of the methods described below or is otherwise useful.
- Per cent homology can be determined by, for example, the GAP program of the University of Wisconsin Genetic Computer Group.
- DNA-DNA, DNA-RNA and RNA-RNA hybridisation may be performed in aqueous solution containing between 0. IXSSC and 6XSSC and at temperatures of between 55 °C and 70 °C. It is well known in the art that the higher the temperature or the lower the SSC concentration the more stringent the hybridisation conditions. By “high stringency” we mean 2XSSC and 65 °C. IXSSC is 0.15M NaCl/0.015M sodium citrate. Polynucleotides which hybridise at high stringency are included within the scope of the claimed invention.
- “Variations" of the polynucleotide also include polynucleotide in which relatively short stretches (for example 20 to 50 nucleotides) have a high degree of homology (at least 80% and preferably at least 90 or 95%) with equivalent stretches of the polynucleotide of the invention even though the overall homology between the two polynucleotides may be much less. This is because important active or binding sites may be shared even when the general architecture of the protein is different.
- a further aspect of the invention provides a replicable vector comprising a recombinant polynucleotide encoding an interacting polypeptide or a polypeptide containing PP(T/N)K of the invention.
- the said recombinant polynucleotide may encode an interacting polypeptide or polypeptide containing PP(T/N)K of the invention that is a fusion of an interacting polypeptide or polypeptide containing PP(T/N)K.
- complementary homopolymer tracts can be added to the DNA segment to be inserted to the vector DNA.
- the vector and DNA segment are then joined by hydrogen bonding between the complementary homopolymeric tails to form recombinant DNA molecules.
- Synthetic linkers containing one or more restriction sites provide an alternative method of joining the DNA segment to vectors.
- the DNA segment generated by endonuclease restriction digestion as described earlier, is treated with bacteriophage T4 DNA polymerase or E. coli DNA polymerase I, enzymes that remove protruding, 3' -single-stranded termini with their 3'-5'-exonucleolytic activities, and fill in recessed 3 '-ends with their polymerizing activities.
- the combination of these activities therefore generates blunt-ended DNA segments.
- the blunt-ended segments are then incubated with a large molar excess of linker molecules in the presence of an enzyme that is able to catalyze the ligation of blunt-ended DNA molecules, such as bacteriophage T4 DNA ligase.
- an enzyme that is able to catalyze the ligation of blunt-ended DNA molecules, such as bacteriophage T4 DNA ligase.
- the products of the reaction are DNA segments carrying polymeric linker sequences at their ends.
- These DNA segments are then cleaved with the appropriate restriction enzyme and ligated to an expression vector that has been cleaved with an enzyme that produces termini compatible with those of the DNA segment.
- Synthetic linkers containing a variety of restriction endonuclease sites are commercially available from a number of sources including International Biotechnologies Inc, New Haven, CN, USA.
- a desirable way to modify the DNA encoding the polypeptide of the invention is to use the polymerase chain reaction as disclosed by Saiki et al (1988) Science 239, 487-491. This method may be used for introducing the DNA into a suitable vector, for example by engineering in suitable restriction sites, or it may be used to modify the DNA in other useful ways as is known in the art.
- the DNA to be enzymatically amplified is flanked by two specific primers which themselves become incorporated into the amplified DNA.
- the said specific primers may contain restriction endonuclease recognition sites which can be used for cloning into expression vectors using methods known in the art.
- the DNA (or in the case of retroviral vectors, RNA) is then expressed in a suitable host to produce a polypeptide comprising the compound of the invention.
- the DNA encoding the polypeptide of the invention may be used in accordance with known techniques, appropriately modified in view of the teachings contained herein, to construct an expression vector, which is then used to transform an appropriate host cell for the expression and production of the polypeptide of the invention.
- Such techniques include those disclosed in US Patent Nos.
- DNA (or in the case of retroviral vectors, RNA) encoding the polypeptide constituting the compound of the invention may be joined to a wide variety of other DNA sequences for introduction into an appropriate host.
- the companion DNA will depend upon the nature of the host, the manner of the introduction of the DNA into the host, and whether episomal maintenance or integration is desired.
- DNA may be linked to the appropriate transcriptional and translational regulatory control nucleotide sequences recognised by the desired host, although such controls are generally available in the expression vector.
- the vector is then introduced into the host through standard techniques. Generally, not all of the hosts will be transformed by the vector. Therefore, it will be necessary to select for transformed host cells.
- One selection technique involves incorporating into the expression vector a DNA sequence, with any necessary control elements, that codes for a selectable trait in the transformed cell, such as antibiotic resistance. Alternatively, the gene for such selectable trait can be on another vector, which is used to co-transform the desired host cell.
- Host cells that have been transformed by the recombinant DNA of the invention are then cultured for a sufficient time and under appropriate conditions known to those skilled in the art in view of the teachings disclosed herein to permit the expression of the polypeptide, which can then be recovered.
- bacteria for example E. coli and Bacillus subtilis
- yeasts for example Saccharomyces cerevisiae
- filamentous fungi for example Aspergillus
- plant cells animal cells and insect cells.
- the vectors include a prokaryotic replicon, such as the ColEl ori, for propagation in a prokaryote, even if the vector is to be used for expression in other, non-prokaryotic, cell types.
- the vectors can also include an appropriate promoter such as a prokaryotic promoter capable of directing the expression (transcription and translation) of the genes in a bacterial host cell, such as E. coli, transformed therewith.
- a promoter is an expression control element formed by a DNA sequence that permits binding of RNA polymerase and transcription to occur.
- Promoter sequences compatible with exemplary bacterial hosts are typically provided in plasmid vectors containing convenient restriction sites for insertion of a DNA segment of the present invention.
- Typical prokaryotic vector plasmids are pUC18, pUC19, pBR322 and pBR329 available from Biorad Laboratories, (Richmond, CA, USA) and p7Vc99A and pKK223-3 available from Pharmacia, Piscataway, NJ, USA.
- a typical mammalian cell vector plasmid is pSVL available from Pharmacia, Piscataway, NJ, USA. This vector uses the SV40 late promoter to drive expression of cloned genes, the highest level of expression being found in T antigen-producing cells, such as COS-1 cells.
- an inducible mammalian expression vector is pMSG, also available from Pharmacia. This vector uses the glucocorticoid-inducible promoter of the mouse mammary tumour virus long terminal repeat to drive expression of the cloned gene.
- Useful yeast plasmid vectors are pRS403-406 and pRS413-416 and are generally available from Stratagene Cloning Systems, La Jolla, CA 92037, USA.
- Plasmids pRS403, pRS404, pRS405 and pRS406 are Yeast Integrating plasmids (Yips) and incorporate the yeast selectable markers HIS3, TRPI, LEU2 and URA3.
- Plasmids pRS413-416 are Yeast Centromere plasmids (YCps).
- the present invention also relates to a host cell transformed with a polynucleotide vector construct of the present invention.
- the host cell can be either prokaryotic or eukaryotic.
- Bacterial cells are preferred prokaryotic host cells and typically are a strain of E. coli such as, for example, the E. coli strains DH5 available from Bethesda Research Laboratories Inc., Bethesda, MD, USA, and RRl available from the American Type Culture Collection (ATCC) of Rockville, MD, USA (No ATCC 31343).
- Preferred eukaryotic host cells include yeast, insect and mammalian cells, preferably vertebrate cells such as those from a mouse, rat, monkey or human fibroblastic and kidney cell lines.
- Yeast host cells include YPH499, YPH500 and YPH501 which are generally available from Stratagene Cloning Systems, La Jolla, CA 92037, USA.
- Preferred mammalian host cells include Chinese hamster ovary (CHO) cells available from the ATCC as CCL61, NIH Swiss mouse embryo cells NIH/3T3 available from the ATCC as CRL 1658, monkey kidney-derived COS-1 cells available from the ATCC as CRL 1650 and 293 cells which are human embryonic kidney cells.
- Preferred insect cells are Sf9 cells which can be transfected with baculovirus expression vectors.
- Transformation of appropriate cell hosts with a DNA construct of the present invention is accomplished by well known methods that typically depend on the type of vector used. With regard to transformation of prokaryotic host cells, see, for example, Cohen et al (1972) Proc. Natl Acad. Sci. USA 69, 2110 and Sambrook et al (1989) Molecular Cloning, A Laboratory Manual, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY. Transformation of yeast cells is described in Sherman et al (1986) Methods In Yeast Genetics, A Laboratory Manual, Cold Spring Harbor, NY. The method of Beggs (1978) Nature 275, 104-109 is also useful.
- reagents useful in transfecting such cells for example calcium phosphate and DEAE-dextran or liposome formulations, are available from Stratagene Cloning Systems, or Life Technologies Inc., Gaithersburg, MD 20877, USA.
- Electroporation is also useful for transforming and/or transfecting cells and is well known in the art for transforming yeast cell, bacterial cells, insect cells and vertebrate cells.
- bacterial species may be transformed by the methods described in Luchansky et al (1988) Mol Microbiol. 2, 637-646 incorporated herein by reference. The greatest number of transformants is consistently recovered following electroporation of the DNA-cell mixture suspended in 2.5X PEB using 6250V per cm at 25 ⁇ FD. Methods for transformation of yeast by electroporation are disclosed in Becker & Guarente (1990) Methods Enzymol 194, 182.
- Successfully transformed cells ie cells that contain a DNA construct of the present invention
- cells resulting from the introduction of an expression construct of the present invention can be grown to produce the polypeptide of the invention.
- Cells can be harvested and lysed and their DNA content examined for the presence of the DNA using a method such as that described by Southern (1975) J. Mol Biol 98, 503 or Berent et al (1985) Biotech. 3, 208.
- the presence of the protein in the supernatant can be detected using antibodies as described below.
- successful transformation can be confirmed by well known immunological methods when the recombinant DNA is capable of directing the expression of the protein.
- cells successfully transformed with an expression vector produce proteins displaying appropriate antigenicity. Samples of cells suspected of being transformed are harvested and assayed for the protein using suitable antibodies.
- the present invention also contemplates a culture of those cells, preferably a monoclonal (clonally homogeneous) culture, or a culture derived from a monoclonal culture, in a nutrient medium.
- a further aspect of the invention provides a method of making a polypeptide of the invention the method comprising culturing a host cell comprising a recombinant polynucleotide or a replicable vector which encodes said polypeptide, and isolating said polypeptide from said host cell.
- Methods of cultivating host cells and isolating recombinant proteins are well known in the art.
- a further aspect of the invention provides an antibody capable of reacting with a polypeptide of the invention, in particular an antibody capable of reacting with an epitope comprising the amino acid sequence PP(T/N)K.
- Antibodies reactive towards the said polypeptide of the invention may be made by methods well known in the art.
- the antibodies may be polyclonal or monoclonal.
- Suitable monoclonal antibodies may be prepared by known techniques, for example those disclosed in “Monoclonal Antibodies: A manual of techniques", H Zola (CRC Press, 1988) and in “Monoclonal Hybridoma Antibodies: Techniques and applications", J G R Hurrell (CRC Press, 1982), both of which are incorporated herein by reference.
- Other techniques for raising and purifying antibodies are well known in the art and any such techniques may be chosen to achieve the preparations useful in the methods claimed in this invention.
- Techniques for preparing antibodies are well known to those skilled in the art, for example as described in Harlow, ED & Lane, D “Antibodies: a laboratory manual” (1988) New York Cold Spring Harbor Laboratory.
- Polyclonal antibodoes may be prepared using methods well known in the art. In the case of both monoclonal and polyclonal antibodies, it is useful to use as immunogen any suitable polypeptide containing a SIM, for example containing the PP(T/N)K motif. In particular with respect to the production of polyclonal antibodies it is useful to use polypeptides of between 10 and 30 amino acid residues containing a SIM, for example containing the PP(T/N)K motif.
- an antibody of the invention is capable of preventing or disrupting the interaction between a Smad polypeptide and a polypeptide comprising a SIM, for example comprising the amino acid sequence PP(T/N)K.
- antibody-like molecules may be useful in the practice of the invention including, for example, antibody fragments or derivatives which retain their antigen-binding sites, synthetic antibody-like molecules such as single-chain Fv fragments (ScFv) and domain antibodies (dAbs), and other molecules with antibody-like antigen binding motifs.
- ScFv single-chain Fv fragments
- dAbs domain antibodies
- antibody-like molecules are included by the term antibody as used below.
- a further aspect of the invention provides a method of disrupting or preventing the interaction between a Smad polypeptide and a polypeptide (target polypeptide) that is (1) a transcription factor capable of interacting with the said Smad polypeptide and/or (2) a polypeptide capable of interacting with the said Smad polypeptide, the interaction requiring ⁇ -helix2 of the said Smad polypeptide, the method comprising exposing the Smad polypeptide to an interacting polypeptide of the invention or an antibody of the invention.
- the Smad polypeptide may be exposed to a compound of the invention, as described below.
- the said polypeptide capable of interacting with the said Smad polypeptide may interact with ⁇ -helix 2 of the said Smad polypeptide; alternatively, the interaction may require ⁇ -helix 2 but contact between the said polypeptide capable of interacting with the said Smad polypeptide and the said Smad polypeptide may occur at site in the said Smad polypeptide that is not part of ⁇ -helix 2.
- a further aspect of the invention provides a method of disrupting or preventing the interaction between a Smad polypeptide and a polypeptide (target polypeptide) which target polypeptide comprises a SIM, for example comprises the amino acid sequence PP(T/N)K, the method comprising exposing the Smad polypeptide to an interacting polypeptide of the invention or an antibody of the invention.
- the Smad polypeptide may be exposed to a compound of the invention, as described below.
- the Smad polypeptide is a naturally occurring Smad polypeptide, for example Smad2 or Smad3 or naturally occurring allelic variants thereof. It is still more preferred that the Smad polypeptide is a human Smad polypeptide, for example human Smad2 or human Smad3.
- the antibody of the invention is capable of reacting with an epitope comprising the amino acid sequence PP(T/N)K.
- the target polypeptide may be an interacting polypeptide of the invention, for example FAST3. It is preferred that the target polypeptide comprises a SIM, for example comprises the amino acid sequence PP(T/N)K.
- the target polypeptide may be FASTI, FAST2, Mixer, Milk or Bix 2 or 3 or a fragment, variant, derivative or fusion thereof. It is preferred that the target polypeptide is a naturally occurring polypeptide or a fusion thereof.
- the interaction between the Smad polypeptide and the target polypeptide and its disruption or prevention may be measured by any method of detecting/measuring a protein/protein interaction, as discussed further below and in Example 1. Suitable methods include yeast two-hybrid interactions, co-purification, ELISA, co-immunoprecipitation methods and bandshift assays.
- the methods may be performed in vitro, either in intact cells or tissues, with broken cell or tissue preparations or at least partially purified components. Alternatively, they may be performed in vivo.
- the cells tissues or organisms in/on which the use or methods are performed may be transgenic. In particular they may be transgenic for the Smad interacting protein under consideration or for a further Smad interacting protein or Smad.
- a further aspect of the invention provides a method of identifying a polypeptide (interacting polypeptide) that is capable of interacting with a Smad polypeptide, for example Smad2 or Smad3, comprising examining the sequence of a polypeptide and determining that the polypeptide comprises a SIM, for example comprises the amino acid sequence PP(T/N)K (or three out of four residues thereof). It is believed that the amino acid sequence PP(T/N)K (or at three out of four residues thereof) is necessary and may be sufficient for interaction of a polypeptide with a Smad polypeptide, for example Smad2 or Smad3. Preferences for the Smad polypeptide are as given above.
- the presence of further or other features of a SIM may be determined. It may be determined that an acidic amino acid residue is present at a position from 3 to 10, preferably 4 to 5 residues residues C-terminal of the amino acid sequence corresponding to the PP(T/N)K motif (which may of course be PP(T/N)K) and/or a proline residue is present at a position from 5 to 20 residues C-terminal of the amino acid sequence corresponding to the PP(T/N)K motif; these residues may also promote the interaction between the said interacting polypeptide and the Smad polypeptide.
- the acidic (negatively charged) amino acid residue is typically a glutamate or aspartate residue. It may further be determined that the said acidic residue (ie present at position about +3 to about + 10, preferably +4 or +5) is immediately followed by a hydrophobic residue, for example M, V or
- the downstream proline and acidic, for example aspartate residues as described above may also be important for binding. It may further be determined that the residue immediately after (ie C-terminal of) the amino acid sequence corresponding to the PP(T/N)K motif ie at position + 1 is an S or T, which may be immediately followed by an I or V residue. It may further be determined that the residue immediately before (ie N-terminal of) the amino acid sequence corresponding to the PP(T/N)K motif is a hydrophobic residue, for example F, M or V. It may further be determined that a proline residue is present at a position from 5 to 20 residues C-terminal of the amino acid sequence corresponding to the sequence motif PP(T/N)K.
- an acidic residue for example glutamate or aspartate
- a hydrophobic residue for example F, Y, L
- an acidic residue for example glutamate or aspartate
- a hydrophobic residue for example F, Y, L
- F, Y, L an acidic residue
- F, Y, L hydrophobic residue
- a leucine residue may be present at position about -2 to -15, preferably about -5 to -10.
- the leucine residue may be the hydrophobic residue that is immediately preceded by an acidic residue, as noted above.
- the method may comprise determining that the polypeptide comprises at least 8,
- the amino acid sequence of the said interacting polypeptide or the nucleotide sequence encoding the said interacting polypeptide may be determined by methods well known to those skilled in the art, for example PCR-based cloning methods, as indicated above. It may be desirable to confirm that the interacting polypeptide identified by the method is capable of interacting with a Smad polypeptide, for example Smad2 or Smad3, using methods of detecting or measuring protein/protein interactions, as described above, for example using the interacting polypeptide expressed as described above.
- the interacting polypeptide may also be useful in a screening assay for identifying a drug like compound that may inhibit the interaction between Smad2 or Smad3 and a polypeptide that interacts with Smad2 or Smad3 in vivo, for example a homologue of Milk, Mixer, other Mix family members, FASTI and FAST2. It will be appreciated that the polypeptide may only interact with Smad2 or Smad3 when the Smad2 or Smad3 is in an activated state, for example following activation and/or phosphorylation as a consequence of TGF ⁇ superfamily receptor activation, or wherein the N-terminal domain is not present or is truncated. It will be appreciated that the Smad2 or Smad3 may further interact with Smad4.
- Smad2 or Smad3 may interact or form a complex with more than one polypeptide that is not Smad4; for example, Smad2 or Smad3 may form a complex with Mixer, Milk and Smad4. Mixer and Milk may form a heterodimer.
- a further aspect of the invention thus provides a method of identifying a compound capable of disrupting or preventing the interaction between a Smad polypeptide and a polypeptide (target polypeptide) that is (1) a transcription factor capable of interacting with the said Smad polypeptide and/or (2) a polypeptide capable of interacting with a Smad polypeptide, the interaction requiring ⁇ -helix2 of the said Smad polypeptide and/or (3) a polypeptide comprising the amino acid sequence PP(T/N)K, the method comprising measuring the ability of the compound to disrupt or prevent the interaction between the Smad polypeptide and an interacting polypeptide of the invention.
- target polypeptide that is (1) a transcription factor capable of interacting with the said Smad polypeptide and/or (2) a polypeptide capable of interacting with a Smad polypeptide, the interaction requiring ⁇ -helix2 of the said Smad polypeptide and/or (3) a polypeptide comprising the amino acid sequence PP(T/N)K
- the interaction between the Smad polypeptide and the interacting polypeptide and its disruption or prevention may be measured by any method of detecting/measuring a protein/protein interaction, as discussed in Example 1. Suitable methods include yeast two-hybrid interactions, co-purification, ELISA, co-immunoprecipitation methods and bandshift assays. Further suitable methods may include Scintillation Proximity Assays, as well known to those skilled in the art.
- suitable methods may include bandshift assays looking for disruption of the endogenous FAST/Smad2/Smad4 ARF complex or disruption of the Mixer/GSTSmad2C interaction, as described in Example 1 and transcription assays in tissue culture cells in which expression of a reporter gene driven by a promoter with a binding site for (for example) Mixer is measured following treatment of the cells with TGF ⁇ . Disruption or prevention of TGF ⁇ - dependent transcription in the presence of the compound may be detected.
- the cells may be transiently transfected or may be a stable cell line capable of expressing Mixer (or other appropriate transcription factor) with an integrated reporter gene, as described in Example 2.
- the reporter gene may express luciferase or a green fluorescent protein (GFP) or secreted alkaline phosphatase (SEAP) or CAT, as well known to those skilled in the art.
- GFP green fluorescent protein
- SEAP secreted alkaline phosphatase
- chip screening methods may be used.
- arrays of cDNAs or oligonucleotides may be used in assessing expression of endogenous genes that are modulated by TGF ⁇ and therefore for assessing effects of compounds on such expression.
- a further aspect of the invention therefore provides a cell comprising 1) a recombinant polynucleotide suitable for expressing a transcription factor that is capable of interacting with a Smad polypeptide and 2) a recombinant polynucleotide comprising a reporter gene driven by a promoter with a binding site for the said transcription factor.
- a further aspect of the invention provides a stably-transformed cell line cell comprising a reporter gene driven by a promoter with a binding site for an activated Smad, wherein the Smad is activated in the cell by exposure of the cell to TGF ⁇ .
- the reporter gene may express luciferase or CAT or SEAP or a green fluorescent protein (GFP).
- a further aspect of the invention provides a method of identifying a compound capable of modulating TGF ⁇ -dependent transcription wherein the effect of the compound on expression of the reporter gene in a cell of the invention is measured, following treatment of the cell with TGF ⁇ .
- a further aspect of the invention provides a method of identifying a compound capable of modulating TGF ⁇ -dependent transcription wherein the effect of the compound on TGF ⁇ -signalling- dependent invasive behaviour of a stably- transformed cell line cell, for example in collagen gels, is measured and a compound that reduces invasive behaviour is selected.
- the stably-transformed cell line is preferably a MDCK cell line that is capable of expressing recombinant active Raf-1, as described in Example 2.
- the methods of the invention may be performed in vitro, either in intact cells or tissues, with broken cell or tissue preparations or at least partially purified components. Alternatively, they may be performed in vivo.
- the cells tissues or organisms in/on which the use or methods are performed may be transgenic. In particular they may be transgenic for the Smad interacting protein under consideration or for a further Smad interacting protein or Smad.
- a transgenic animal for example a transgenic rodent, for example mouse or rat, amphibian, for example Xenopus, or insect, for example Drosophila, transgenic for the Smad interacting protein under consideration or for a further Smad interacting protein or Smad may be useful, for example in the screening methods of the invention.
- screening assays which are capable of high throughput operation will be particularly preferred.
- Examples may include cell based assays, for example as described in Chen et al (1997) and protein-protein binding assays.
- An SPA-based (Scintillation Proximity Assay; Amersham International) system may be used.
- beads comprising scintillant and a Smad polypeptide, for example Smad2 or a fragment (for example the MH2 domain) may be prepared.
- the beads may be mixed with a sample comprising the interacting polypeptide into which a radioactive label has been incorporated and with the test compound. Conveniently this is done in a 96- well format.
- the plate is then counted using a suitable scintillation counter, using known parameters for the particular radioactive label in an SPA assay. Only the radioactive label that is in proximity to the scintillant, ie only that bound to the interacting polypeptide that is bound to the Smad polypeptide anchored on the beads, is detected. Variants of such an assay, for example in which the Smad polypeptide is immobilised on the scintillant beads via binding to an antibody or antibody fragment, may also be used.
- FRET Fluorescence Energy Resonance Transfer
- the compound may be a drug-like compound or lead compound for the development of a drug-like compound for each of the above methods of identifying a compound. It will be appreciated that the said methods may be useful as screening assays in the development of pharmaceutical compounds or drugs, as well known to those skilled in the art.
- drug-like compound is well known to those skilled in the art, and may include the meaning of a compound that has characteristics that may make it suitable for use in medicine, for example as the active ingredient in a medicament.
- a drug-like compound may be a molecule that may be synthesised by the techniques of organic chemistry, less preferably by techniques of molecular biology or biochemistry, and is preferably a small molecule, which may be of less than 5000 daltons molecular weight.
- a drug- like compound may additionally exhibit features of selective interaction with a particular protein or proteins and be bioavailable and/or able to penetrate cellular membranes, but it will be appreciated that these features are not essential.
- lead compound is similarly well known to those skilled in the art, and may include the meaning that the compound, whilst not itself suitable for use as a drug (for example because it is only weakly potent against its intended target, non-selective in its action, unstable, difficult to synthesise or has poor bioavailability) may provide a starting-point for the design of other compounds that may have more desirable characteristics.
- the compound may be a polypeptide that is capable of competing with the interacting polypeptide of the invention for binding to the Smad polypeptide, and may be (1) a transcription factor capable of interacting with the said Smad polypeptide and/or (2) a polypeptide capable of interacting with a Smad polypeptide, the interaction requiring ⁇ -helix2 of the said Smad polypeptide and/or (3) a polypeptide comprising a SIM, for example comprising the amino acid sequence PP(T/N)K.
- a screening method as described above may be useful in identifying polypeptides that may interact with the Smad polypeptide.
- Methods that may be useful in identifying polypeptides that may interact with the Smad polypeptide include yeast-2-hybrid, co-immunoprecipitation, ELISA, GST- pulldown, bandshift and transcription assays. Transcription assays may be performed in vivo or in vitro.
- tissue culture cells may be used which comprise a reporter construct in which expression of the reporter gene is controlled a promoter comprising a binding site(s) for the putative interacting polypeptide. The effect of treating the cells with TGF ⁇ on expression of the reporter gene may then be measured; TGF ⁇ -dependent expression of the reporter gene may indicate that the putative interacting polypeptide is capable of being regulated by TGF ⁇ and therefore may interact with the said Smad polypeptide.
- a transcription assay may be performed in a transgenic animal, for example a transgenic Drosophila or Xenopus.
- a further aspect of the invention is a kit of parts useful in carrying out a method, for example a screening method, of the invention.
- a kit may comprise a Smad polypeptide, for example Smad2 or Smad3 or a fragment either therof and an interacting polypeptide, for example a polypeptide corresponding to amino acids 283 to 307 of Mixer.
- a further aspect of the invention provides a compound identified by or identifiable by the screening method of the invention.
- such a compound may be an inhibitor of the formation or stability of a complex of the Smad polypeptide used in the screen, for example Smad2 or Smad3, with interacting polypeptide(s), for example Smad4 and a transcription factor, for example FASTI, FAST2, Mixer, Milk or Bix2/3, and therefore ultimately of the activity of that complex, for example in promoting the transcription from a promoter to which the complex binds.
- the intention of the screen may be to identify compounds that act as modulators, for example inhibitors or promoters, preferably inhibitors of the activity of the complex, even if the screen makes use of a binding assay rather than an activity (for example transcriptional activity or DNA binding) assay.
- the inhibitory action of a compound found to bind the Smad or Smad interacting polypeptide may be confirmed by performing an assay of, for example, transcriptional or DNA binding activity in the presence of the compound.
- a further aspect of the invention provides a compound identified by or identifiable by the screening method of the invention for use in medicine.
- a still further aspect of the invention provides an interacting polypeptide or polypeptide containing PP(T/N)K or molecule of the invention or nucleic acid of the invention or antibody of the invention for use in medicine.
- the compound, interacting polypeptide, polypeptide containing PP(T/N)K, molecule, nucleic acid or antibody of the invention is suitably packaged and presented for use in medicine.
- the aforementioned interacting polypeptide or molecule of the invention or nucleic acid of the invention or antibody of the invention or a formulation thereof may be administered by any conventional method including oral and parenteral (e.g. subcutaneous or intramuscular) injection.
- the treatment may consist of a single dose or a plurality of doses over a period of time.
- an interacting polypeptide or molecule of the invention or nucleic acid of the invention or antibody of the invention Whilst it is possible for an interacting polypeptide or molecule of the invention or nucleic acid of the invention or antibody of the invention to be admmistered alone, it is preferable to present it as a pharmaceutical formulation, together with one or more acceptable carriers.
- the carrier(s) must be "acceptable” in the sense of being compatible with the compound of the invention and not deleterious to the recipients thereof.
- the carriers will be water or saline which will be sterile and pyrogen free.
- the invention also provides pharmaceutical compositions comprising the interacting polypeptide or molecule of the invention or nucleic acid of the invention or antibody of the invention and a pharmaceutically acceptable carrier.
- the nucleic acid of the invention may be an antisense oligonucleotide, for example an antisense oligonucleotide directed against a nucleic acid encoding an interacting polypeptide of the invention, which may be a transcription factor comprising a SIM, for example comprising the amino acid sequence PP(N/T)K. It is preferred that the antisense oligonucleotide is directed against a nucleic acid encoding a human transcription factor.
- Antisense oligonucleotides are single-stranded nucleic acid, which can specifically bind to a complementary nucleic acid sequence. By binding to the appropriate target sequence, an RNA-RNA, a DNA-DNA, or RNA-DNA duplex is formed. These nucleic acids are often termed "antisense” because they are complementary to the sense or coding strand of the gene. Recently, formation of a triple helix has proven possible where the oligonucleotide is bound to a DNA duplex. It was found that oligonucleotides could recognise sequences in the major groove of the DNA double helix. A triple helix was formed thereby.
- the above oligonucleotides can inhibit the function of the target nucleic acid.
- This bould for example, be a result of blocking the transcription, processing, poly(A)addition, replication, translation, or promoting inhibitory mechanisms of the cells, such as promoting RNA degradations.
- Antisense oligonucleotides are prepared in the laboratory and then introduced into cells, for example by microinjection or uptake from the cell culture medium into the cells, or they are expressed in cells after transfection with plasmids or retroviruses or other vectors carrying an antisense gene.
- Antisense oligonucleotides were first discovered to inhibit viral replication or expression in cell culture for Rous sarcoma virus, vesicular stomatitis virus, herpes simplex virus type 1, simian virus and influenza virus. Since then, inhibition of mRNA translation by antisense oligonucleotides has been studied extensively in cell-free systems including rabbit reticulocyte ly sates and wheat germ extracts.
- the cap, 5' untranslated region, and poly(A) signal lie within the sequence repeated at the ends of retrovirus RNA (R region) and the oligonucleotides complementary to these may bind twice to the RNA.
- Oligonucleotides are subject to being degraded or inactivated by cellular endogenous nucleases. To counter this problem, it is possible to use modified oligonucleotides, eg having altered internucleotide linkages, in which the naturally occurring phosphodiester linkages have been replaced with another linkage. For example, Agrawal et al (1988) Proc. Natl Acad. Sci.
- Oligonucleotides having artificial linkages have been shown to be resistant to degradation in vivo.
- Shaw et al (1991) in Nucleic Acids Res. 19, 747-750 report that otherwise unmodified oligonucleotides become more resistant to nucleases in vivo when they are blocked at the 3' end by certain capping structures and that uncapped oligonucleotide phosphorothioates are not degraded in vivo.
- oligonucleotide is a deoxyribonucleic acid (DNA), although ribonucleic acid (RNA) sequences may also be synthesised and applied.
- DNA deoxyribonucleic acid
- RNA ribonucleic acid
- the oligonucleotides useful in the invention preferably are designed to resist degradation by endogenous nucleolytic enzymes. In vivo degradation of oligonucleotides produces oligonucleotide breakdown products of reduced length. Such breakdown products are more likely to engage in non-specific hybridization and are less likely to be effective, relative to their full-length counterparts. Thus, it is desirable to use oligonucleotides that are resistant to degradation in the body and which are able to reach the targeted cells.
- the present oligonucleotides can be rendered more resistant to degradation in vivo by substituting one or more internal artificial internucleotide linkages for the native phosphodiester linkages, for example, by replacing phosphate with sulphur in the linkage.
- linkages examples include phosphorothioates, methylphosphonates, sulphone, sulphate, ketyl, phosphorodithioates, various phosphoramidates, phosphate esters, bridged phosphorothioates and bridged phosphoramidates.
- Such examples are illustrative, rather than limiting, since other internucleotide linkages are known in the art. See, for example, Cohen, (1990) Trends in Biotechnology.
- the synthesis of oligonucleotides having one or more of these linkages substituted for the phosphodiester internucleotide linkages is well known in the art, including synthetic pathways for producing oligonucleotides having mixed internucleotide linkages.
- Oligonucleotides can be made resistant to extension by endogenous enzymes by "capping" or incorporating similar groups on the 5' or 3' terminal nucleotides.
- a reagent for capping is commercially available as Amino-Link IITM from Applied BioSy stems Inc, Foster City, CA. Methods for capping are described, for example, by Shaw et al (1991) Nucleic Acids Res. 19, 747-750 and Agrawal et al (1991) Proc. Natl Acad. Sci. USA 88(17), 7595-7599, the teachings of which are hereby incorporated herein by reference.
- oligonucleotides resistant to nuclease attack are for them to be "self-stabilised” as described by Tang et al (1993) Nucl Acids Res. 21, 2729-2735 incorporated herein by reference.
- Self-stabilised oligonucleotides have hairpin loop structures at their 3' ends, and show increased resistance to degradation by snake venom phosphodiesterase, DNA polymerase I and fetal bovine serum.
- the self-stabilised region of the oligonucleotide does not interfere in hybridization with complementary nucleic acids, and pharmacokinetic and stability studies in mice have shown increased in vivo persistence of self-stabilised oligonucleotides with respect to their linear counterparts.
- antisense agents also include larger molecules which bind to said interacting polypeptide mRNA or genes and substantially prevent expression of said interacting polypeptide mRNA or genes and substantially prevent expression of said interacting polypeptide.
- expression of an antisense molecule which is substantially complementary to said interacting polypeptide is envisaged as part of the invention.
- the said larger molecules may be expressed from any suitable genetic construct as is described below and delivered to the patient.
- the genetic construct which expresses the antisense molecule comprises at least a portion of the said interacting polypeptide coding sequence operatively linked to a promoter which can express the antisense molecule in the cell.
- Suitable promoters will be known to those skilled in the art, and may include promoters for ubiquitously expressed, for example housekeeping genes or for tissue-specific genes, depending upon where it is desired to express the antisense molecule.
- the genetic construct can be DNA or RNA it is preferred if it is DNA.
- the genetic construct is adapted for delivery to a human cell.
- constructs of the invention may be introduced into the cells by any convenient method, for example methods involving retroviruses, so that the construct is inserted into the genome of the (dividing) cell.
- Other methods involve simple delivery of the construct into the cell for expression therein either for a limited time or, following integration into the genome, for a longer time.
- An example of the latter approach includes liposomes (Nassander et al (1992) Cancer Res. 52, 646-653).
- Other methods of delivery include adenoviruses carrying external DNA via an antibody-polylysine bridge (see Curiel Prog. Med. Virol 40, 1-18) and transferrin-polycation conjugates as carriers (Wagner et al (1990) Proc. Natl Acad. Sci. USA 87, 3410-3414).
- the DNA may also be delivered by adenovirus wherein it is present within the adenovirus particle.
- naked DNA and DNA complexed with cationic and neutral lipids may also be useful in introducing the DNA of the invention into cells of the patient to be treated.
- Non-viral approaches to gene therapy are described in Ledley (1995) Human Gene Therapy 6, 1129-1144.
- Alternative targeted delivery systems are also known such as the modified adenovirus system described in WO 94/10323 wherein, typically, the DNA is carried within the adenovirus, or adenovirus-like, particle.
- Michael et al (1995) Gene Therapy 2, 660-668 describes modification of adenovirus to add a cell-selective moiety into a fibre protein.
- a further aspect of the invention provides a virus or virus-like particle comprising a genetic construct of the invention.
- suitable viruses or virus-like particles include HSV, AAV, vaccinia and parvovirus.
- a ribozyme capable of cleaving the interacting polypeptide RNA or DNA may be administered in substantially the same and using substantially the same vehicles as for the antisense molecules.
- Ribozymes which may be encoded in the genomes of the viruses or virus-like particles herein disclosed are described in Cech and Herschlag "Site-specific cleavage of single stranded DNA” US 5,180,818; Altman et al "Cleavage of targeted RNA by RNAse P" US 5,168,053, Cantin et al "Ribozyme cleavage of HIV-1 RNA” US 5,149,796; Cech et al "RNA ribozyme restriction endoribonucleases and methods", US 5,116,742; Been et al "RNA ribozyme polymerases, dephosphorylases, restriction endonucleases and methods", US 5,093,246; and Been et al "RNA ribozyme
- the genetic constructs of the invention can be prepared using methods well known in the art.
- a further aspect of the invention provides a method of modulating, for example enhancing or inhibiting, preferably inhibiting, activin or TGF ⁇ signalling in a cell in vitro or in vivo wherein the cell is exposed to a polypeptide, molecule, nucleic acid, antibody or compound of the invention. It is preferred that the said polypeptide, molecule, nucleic acid, antibody or compound of the invention is able to enter the cell. Methods of optimising delivery to and uptake of such molecules by a cell are known to those skilled in the art and their use is envisaged here.
- the cell may be a tumour cell, for example a late stage tumour cell.
- a further aspect of the invention provides the use of a polypeptide, molecule, polynucleotide, compound or antibody of the invention in the manufacture of a medicament for treatment of a patient in need of modulation, preferably inhibition, of activin or TGF ⁇ signalling.
- a further aspect of the invention provides a method of treatment of a patient in need of modulation, preferably inhibition, of activin or TGF ⁇ signalling wherein an effective amount of a polypeptide, polynucleotide, compound or antibody of the invention is admmistered to the patient.
- a further aspect of the invention provides the use of a polypeptide, molecule, polynucleotide, compound or antibody of the invention in the manufacture of a medicament for treatment of a patient with cancer.
- a further aspect of the invention provides a method of treatment of a patient with cancer wherein an effective amount of a polypeptide, polynucleotide, compound or antibody of the invention is admmistered to the patient.
- the patient is mammalian. It is further preferred that the patient is human.
- TGF ⁇ is believed to be involved, for example, in scarring, tissue regeneration and kidney response to diabetes and therefore inhibition of TGF ⁇ signalling via the type-I and type-II receptors may be useful in medicine.
- Activin type-I and type-II receptors may be mediate activins' roles in regulating endocrine cells from the reproductive system, promoters of erythroid differentiation and in inducing axial mesoderm and anterior structures in vertebrates. Inhibins may have effects antagonistic to those of activins.
- BMP receptors may be involved in similar processes to TGF ⁇ and activins, and particularly in bone growth and maintenance. TGF ⁇ s may be expressed in a wider range of tissues than other members of the superfamily, which may have more specialised roles.
- TGF ⁇ is also believed to be involved in carcinogenesis (see, for example Lawrence (1996), cited above) and therefore compounds that inhibit TGF ⁇ and related receptor signalling may be useful in the treatment of cancer.
- Losses of Smad4 may be particularly associated with pancreatic and colon cancers; these cancers may not require TGF ⁇ for progression.
- Breast cancer tumours are mentioned by Reiss (1997) Oncol Res 9, 447-457 as having high levels of TGF ⁇ associated with them and promoting tumour progression (see also Oft et al (1998) TGF ⁇ signalling is necessary for carcinoma cell invasiveness and metastasis Curr Biol 8, 1243-1252).
- a further aspect of the invention is the use of a polypeptide, molecule, polynucleotide, compound or antibody of the invention in the manufacture of a medicament for treatment of a patient in need of reducing extracellular matrix deposition, encouraging tissue repair and/or regeneration, tissue remodelling or healing of a wound, for example burn, injury or surgery, or reducing scar tissue formation arising from injury to the brain.
- a further aspect of the invention is a method of treatment of a patient in need of reducing extracellular matrix deposition, encouraging tissue repair and/or regeneration, tissue remodelling or healing of a wound, for example burn, injury or surgery, or reducing scar tissue formation arising from injury to the brain wherein an effective amount of a polypeptide, molecule, polynucleotide, compound or antibody of the invention is administered to the patient.
- Extracellular matrix deposition is a term well known to those skilled in the art, and is described for example in Grande (1997) and Lawrence (1996), cited above.
- Extracellular matrix components include collagens, fibronectin, tenascin, glycosaminoglycans and proteoglycans. Deposition of such components may lead to rapid wound healing but may also lead to scarring, particularly in the brain.
- TGF ⁇ may inhibit degradation of the extracellular matrix (for example by inhibiting production of proteases and stimulating the production of specific protease inhibitors. It will be appreciated that the medicament may be applied before surgery. It will be appreciated that the injury may be mechanical injury. It is preferred that it is not reperfusion injury.
- a still further aspect of the invention is the use of a polypeptide, molecule, polynucleotide, compound or antibody of the invention in the manufacture of a medicament for treatment of a patient with or at risk of end-stage organ failure, pathologic extracellular matrix accumulation, a fibrotic condition, disease states associated with immunosuppression (such as different forms of malignancy, chronic degenerative diseases, and AIDS), diabetic nephropathy, tumour growth, kidney damage (for example obstructive neuropathy, IgA nephropathy or noninflammatory renal disease) or renal fibrosis.
- a further aspect of the invention provides a method of treating a patient with or at risk of end-stage organ failure, pathologic extracellular matrix accumulation, a fibrotic condition, disease states associated with immunosuppression (such as different forms of malignancy, chronic degenerative diseases, and AIDS), diabetic nephropathy, tumour growth, kidney damage (for example obstructive neuropathy, IgA nephropathy or noninflammatory renal disease) or renal fibrosis wherein an effective amount of a polypeptide, molecule, polynucleotide, compound or antibody of the invention is administered to the patient.
- immunosuppression such as different forms of malignancy, chronic degenerative diseases, and AIDS
- diabetic nephropathy tumour growth
- kidney damage for example obstructive neuropathy, IgA nephropathy or noninflammatory renal disease
- renal fibrosis wherein an effective amount of a polypeptide, molecule, polynucleotide, compound or antibody of the invention is administered to the patient.
- the patient may alternatively have, or be at risk of, a form of a disorder of bone growth or homeostasis (such as osteoporosis), arthritis or atherosclerosis in which TGF ⁇ or a related protein (for example an activin, inhibin or BMP) has been implicated, in causing or exacerbating the condition.
- a form of a disorder of bone growth or homeostasis such as osteoporosis
- arthritis or atherosclerosis in which TGF ⁇ or a related protein (for example an activin, inhibin or BMP) has been implicated, in causing or exacerbating the condition.
- TGF ⁇ or a related protein for example an activin, inhibin or BMP
- the patient may be suffering from a TGF ⁇ -related condition as reviewed in Roberts & Sport (1993) Physiological actions and clinical applications of transforming growth factor ⁇ (TGF ⁇ ) Growth Factors 8, 1-9, for example hepatic cirrhosis, idiopathic pulmonary fibrosis, scleroderma, glomerulonephritis, certain forms of rheumatoid arthritis, schistosomiasis or proliferative vitreoretinopathy.
- TGF ⁇ transforming growth factor ⁇
- the polypeptide, molecule, polynucleotide, compound, antibody, composition or medicament of the invention may be administered in any suitable way, usually parenterally, for example intravenously, intraperitoneally or intravesically, in standard sterile, non-pyrogenic formulations of diluents and carriers.
- the polypeptide, molecule, polynucleotide, compound, antibody, composition or medicament of the invention of the invention may also be administered topically, which may be of particular benefit for treatment of surface wounds.
- the polypeptide, molecule, polynucleotide, compound, antibody, composition or medicament of the invention may also be administered in a localised manner, for example by injection.
- a further aspect of the invention provides a substantially pure complex comprising (1) a Smad2 or Smad3 polypeptide, (2) a Smad4 polypeptide and (3) a Mixer and/or Milk and/or Bix2/3 and/or FAST3 polypeptide.
- a substantially pure complex comprising (1) a Smad2 or Smad3 polypeptide, (2) a Smad4 polypeptide and (3) a Mixer and/or Milk and/or Bix2/3 and/or FAST3 polypeptide.
- a further aspect of the invention provides a preparation comprising (1) Smad2 or Smad3 polypeptide, (2) a Smad4 polypeptide and (3) a Mixer and/or Milk and/or Bix2/3 and/or FAST3 polypeptide (in the form of a complex or otherwise) when combined with other components ex vivo, said other components not being all of the components found in the cell in which said (1) Smad2 or Smad3 polypeptide, (2) a Smad4 polypeptide and (3) a Mixer and/or Milk and/or Bix2/3 and/or FAST3 polypeptide (in the form of a complex or otherwise) are naturally found.
- the preparation may comprise a polypeptide that stabilises the preparation, for example bovine serum albumin or gelatin.
- substantially pure we mean that the complex is substantially free of other proteins.
- any composition that includes at least 30% of the protein content by weight as the said complex or its components, preferably at least 50%, more preferably at least 70%, still more preferably at least 90% and most preferably at least 95 % of the protein content is the said complex or its components.
- the substantially pure complex may include a contaminant wherein the contaminant comprises less than 70% of the composition by weight, preferably less than 50% of the composition, more preferably less than 30% of the composition, still more preferably less than 10% of the composition and most preferably less than 5% of the composition by weight.
- the substantially pure said complex may be combined with other components ex vivo, said other components not being all of the components found in the cell in which said complex is naturally found.
- FIG. 1 Activin-responsive transcription via the goosecoid DE.
- A Activin-responsive transcription via the DE is partially dependent on new protein synthe sis.
- One-cell embryos were injected with REF-globin internal control together with globin reporters driven by the minimal ⁇ -actin promoter ( ⁇ A), or by multiple copies of the DE or ARE upstream of the mimimal promoter.
- Animal caps, cut at St 8 were cultured for 6 h ⁇ activin in the absence or presence of 5 ⁇ g/ml cycloheximide.
- Globin transcripts from reporter genes (Test-globin) or the internal control (REF-globin) were detected by RNase protection (Howell and Hill, 1997).
- Transcriptional activation was calculated as a ratio of the levels of Test-globin to REF-globin. Activin-induced transcription is expressed as fold inductions. In close agreement with the data shown in this experiment, a similar independent experiment measuring the activin inducibility of the DE gave a 22.1- fold induction in the absence of cycloheximide and 6.1 -fold in the presence of cycloheximide.
- An activin-inducible factor (DEBP) binds the goosecoid DE. Whole cell extracts prepared from St 8 or St 11 embryos or St 11 embryos overexpressing activin, were analysed by bandshift assay using the single DE probe. Activin- inducible factor, DEBP is indicated.
- Competitor oligonucleotides were used at a 50-fold molar excess over probe where indicated.
- the paired-like homeodomain binding site comprising 2 inverted TAAT motifs (Wilson et al 1993) is denoted by arrows; a third homeodomain binding site at the 3' end is also indicated by an arrow.
- GSTSmad2C (lanes 2,3, 6,7, 10,11) prior to addition of probe. Open arrow, DEBP; black arrow, GSTSmad2C associated with DEBP.
- FIG. 4 Characterization of the Smad Interaction Motif (SIM) (A) Schematics of Mix.1, Mixer and Milk, with the conserved homeodomains and a C-terminal acidic domain indicated. Black box; a region conserved in Milk and Mixer, also present in Xenopus Fast-1 and mouse Fast-2 (expanded below where the black line denotes the boundaries of the conserved sequences). Black shading, identical amino acids; gray shading, similar amino acids. The numbers indicate the positions of these amino acids in the full length sequences of the individual proteins.
- SIM Smad Interaction Motif
- GST (lanes 2,5,8,11,14) or GSTSmad2C (lanes 3,6,9,12,15) and bound protein was visualized by SDS-PAGE and autoradiography. A fraction of input protein was analysed for comparison (lanes 1,4,7,10,13).
- the endogenous ARF complex is indicated.
- FIG. 7 Mixer and Milk mediate TGF- ⁇ -dependent transcriptional activation via the DE (A).
- NIH3T3 cells were transfected with the CAT reporters, and plasmids expressing transcription factors, Smad2 and Smad4 as indicated.
- Cells were cultured ⁇ TGF- ⁇ 1 (2 ng/ml) for 8 hr.
- Cells were harvested and CAT activity measured relative to lacZ activity from the internal control. The data are from a representative experiment, and similar results were obtained in at least three further independent experiments.
- NIH3T3 cells were transfected with the (DE)4-globin reporter and REF-globin internal control with or without Mixer expression plasmid.
- Cells were cultured ⁇ TGF- ⁇ l (2 ng/ml) for 4 hr in the absence or presence of 50 ⁇ g/ml cycloheximide.
- Globin transcripts from the reporter genes (test-globin) or the internal control (REF-globin) were detected by RNase protection and quantitated as in Figure 1.
- FIG. 8 The temporal and spatial expression patterns of Mixer and Milk in Xenopus embryos makes them good candidates for mediating transcription of goosecoid in response to an endogenous activin-like signal.
- A Co-expression of goosecoid with Mixer and Milk at early gastrula stages.Xenopus embryos were fixed at St 10.25 and processed for in situ hybridization with probes against goosecoid (Gsc), Mixer or Milk either singly (left panels) or sequentially (right panels). Arrowhead, dorsal lip. Gsc mRNA is visualized with deep purple stain. Mixer and Milk mRNA are visualized with a turquoise stain.
- Figure 9 Mixer and Milk mediate TGF- ⁇ -dependent transcriptional activation via a single DE.
- NIH3T3 cells were transfected with a CAT reporter gene driven by a single copy of the goosecoid DE, and plasmids expressing transcription factors Mixer, Mixer (PP mut), Milk and Fast-1 as shown.
- Cells were cultured ⁇ TGF- ⁇ 1 (2 ng/ml) for 8 hr. Cells were harvested and CAT activity measured. The data are from a representative experiment, and similar results were obtained in two further independent experiments.
- An activin-inducible factor binds the paired-like homeodomain binding site of the goosecoid DE.
- Whole cell extracts prepared from St 8 or St 11 embryos or St 11 embryos overexpressing activin were analysed by bandshift assay either on the wild type DE probe, or mutant DE probes as indicated.
- Activin-inducible factor, DEBP is indicated.
- sequences of wild-type DE and mutant oligonucleotides, where only the altered nucleotides are indicated.
- the paired-like homeodomain binding site comprising 2 inverted TAAT motifs (Wilson et al 1993) is denoted by arrows; a third homeodomain binding site at the 3' end is also indicated by an arrow.
- FIG. 11 Mapping the Mixer interaction domain in Smad2.
- In-vitro translated Mixer was tested in a bandshift assay, using radiolabelled DE as probe, for its ability to interact with different Smad2 effector domain mutants, produced bacterially as GST fusion proteins. Mixer complexed with probe is indicated; black arrow, ternary complex with GSTSmad2C derivatives.
- Helix 2 of the Smad2 effector domain is required for the interaction with Mixer. The assay was as above. The effector domain of Smadl does not interact with Mixer.
- GSTSmad2C H2 swap
- Helix 2 from Smadl replaces Helix 2 of Smad2. This mutant contains only 4 amino acid changes relative to GSTSmad2C (Shi et al 1997) and no longer interacts with Mixer.
- FIG. 12 Amino acid sequence of human and Xenopus Smad2 and Smad3.
- Xenopus Smad3 is a novel Smad polypeptide.
- FIG. 13 A. Alignment of the SIM in proteins of the Mix and Fast families that are known to interact with Smad2.
- the regions of the proteins are as follows: Mixer, 273-317; ZF Mixer, 228-272; Milk, 309-350; Bix3, 291-332; XFastl , 459-503; HFastl, 316-360; MFast2, 352-396; XFast3, 288-334.
- residues that are either completely conserved in all the SIMs or exhibit highly conserved substitutions.
- Underlined is a pair of amino acids (an acidic residue followed by a hydrophobic residue) that is present in all the SIMs.
- Zebrafish Fast-1 has recently been cloned (accession number AF263000) and contains the conserved residues that define the SIM.
- XFast-3 forms an ARF complex in extracts made from Xenopus embryos injected with mRNA expressing activin and Flag-tagged XFast-3.
- Flag- tagged XFast-3 forms a complex in Xenopus embryos, ARF2 (Howell et al., 1999) that also contains Smad2 as demonstrated by the observation that the complex supershifts with antibodies against the flag tag on XFast-3 and against Smad2 (lanes 4-6).
- the complex also contains Smad4 (data not shown). This behaviour is similar to the behaviour of XFast-1, which forms an equivalent complex containing Smad2 and Smad4 (ARF1; lanes 1-3).
- the high salt extracts containing Flag-tagged XFast-1 were made at 80 min post Stage 8 and those containing Flag-tagged XFast-3 were made 240 min post Stage 8 (Howell et al., 1999).
- FIG. 15 A mutational analysis of the SIM indicates that the affinity of a Mixer derivative for Smad2 in vitro correlates well with the TGF- ⁇ inducible transcriptional activity of the Mixer derivative in vivo.
- A. The sequence of the Mixer SIM indicating the residues that have been mutated to alanine in the single mutations. Underlined is the core of the SIM that is conserved in all functional SIMs (see Figure 13). All mutations have been made in the context of full length Mixer.
- B Bandshift analyses to demonstrate the interaction of the Mixer mutants with GSTSmad2C (Example 1 and Germain et al. , 2000).
- the radiolabelled DNA probe is the goosecoid DE.
- the Mixer derivatives were expressed in reticulocyte lysate.
- the complex labelled Mixer is a Mixer derivative bound to its binding site in the DE.
- the black arrow denotes the ternary complex of Mixer/Smad2C/DNA.
- the titration of GSTSmad2C was in two-fold dilutions. The highest amount added was 20 ng as estimated by Bradford assay. The amounts added were therefore : 20, 10, 5, 2.5, 1.25 and 0.625 ng.
- the cells were NIH3T3s.
- the reporter was the DE-luciferase reporter which is equivalent to the DE-CAT reporter used previously (Example 1 and Germain et al., 2000) but based on pGL3-basic vector (Promega).
- TGF- ⁇ 1 inductions were with 2 ng/ml TGF- ⁇ and for 8 h.
- 50 ng expression plasmid for each mutant derivative and 350 ng reporter plasmid were transfected together with 100 ng EF-lacZ (Example 1 and Germain et al., 2000) as an internal control.
- Luciferase was quantitated relative to ⁇ -Gal activity, and the value for TGF- ⁇ induced transcription using wild type Mixer was set at 100.
- the data are means and standard deviations of 3 independent experiments.
- the Mixer mutants were all expressed at approximately equal levels in the NIH3T3 cells as determined by bandshift analysis (data not shown).
- FIG. 16 A. The SIM peptide specifically disrupts the formation of the Smad3/Smad4 complex in vitro.
- the radiolabelled probe was the Smad binding sites from the c-jun promoter (Lehmann et al., 2000).
- HaCaT cells were treated with 2ng/ml TGF- ⁇ 1 for 1 h, and nuclear extracts were prepared. 10 ⁇ g of nuclear extract was preincubated with antibodies (l ⁇ l anti-Smad 3 (Nakao et al 1997) ⁇ 10 g competing peptide ); 0.2 ⁇ g anti-Smad4 (B8, Santa Cruz) or Mixer SIM peptide or mutant Mixer SIM peptide (5, 10, 25, 50, 100 pmoles) for 5 minutes at room temperature prior to probe mix addition.
- the ⁇ -Smad3 competing peptide was pre-incubated with nuclear extract for 5 minutes before antibody addition.
- the Smad3/4 complex is indicated.
- the black and white arrows indicate antibody-supershifted complexes.
- B The SIM peptide specifically disrupts the formation of the XFast- 1/Smad2/Smad4 complex in vitro, but not the XFast-3/Smad2/Smad4 complex.
- Whole cell extracts were made from NIH3T3 cells transiently transfected with Flag-XFast-1 and Flag-XFast-3 expression plasmids, that were either untreated or induced with 2ng/ml TGF- ⁇ 1 for 1 h.
- FIG 17. The SIM peptide specifically disrupts the formation of the Smad3/Smad4 complex in vivo.
- Nuclear extracts were prepared from HaCaT cells treated with 5, 25 or 50 ⁇ M Mixer SIM peptide or mutant Mixer SIM peptide for 30 min prior to treatment with 2 ng/ml TGF- ⁇ 1 for 1 h.
- the bandshift assay was as in Figure 16A; 10 ⁇ g nuclear extract was used in each lane.
- the antibody supershifts were as in Figure 16B.
- the black and white arrows indicate antibody-supershifted complexes.
- SIM peptide specifically disrupts the formation of the XFast- 3/Smad2//Smad4 complex in vivo.
- NIH3T3 cells were transfected with XFast-3. 48 h after transfection cells were treated for 30 min with 50 ⁇ M Mixer SIM peptide or mutant Mixer SIM peptide and then treated for 1 h with 2 ng/ml TGF- ⁇ l. Nuclear extracts were prepared and 3 ⁇ g was used for each lane in the bandshift assay with the ARE probe.
- the SIM peptide specifically inhibits transcription of the JunB gene in vivo.
- HaCaT cells were treated with 50 ⁇ M Mixer SIM peptide or mutant Mixer SIM peptide and then treated for 1 h with 2 ng/ml TGF- ⁇ 1.
- Total RNA was extracted and the RNase protection was performed as described (Howell et al., 1999).
- the ⁇ -actin probe was as described (Enoch et al., 1986).
- the JunB probe protects amino acids 42-109 of human JunB.
- Figure 18 Nucleotide sequence of the XFAST-3 coding region.
- DEBP activin-activated protein
- Smad4 This allows recruitment of Smad4 to form an activin/TGF ⁇ -inducible complex that mediates transcriptional activation via the goosecoid DE, the activin-responsive element of the Xenopus goosecoid promoter.
- This Smad interaction motif (SIM) is also conserved in the C-terminal regions of the unrelated Smad2-interacting forkhead transcription factors, Fast-1 and Fast-2.
- the distal element (DE) in the Xenopus goosecoid promoter is a cis-acting element necessary and sufficient to activate transcription in response to activin (Watabe et al 1995).
- globin reporter genes with four copies of the DE or three copies of the ARE linked to a minimal promoter and measured transcription by RNase protection assay, quantitating it relative to the activity of a co-injected constitutively active reference globin gene (Howell and Hill, 1997).
- the amount of transcript from TEST-Globin has to be divided by the amount of transcript from the REF-globin.
- REF-globin acts as an internal control for injection efficiency, RNA extraction efficiency and as a loading control.
- the minimal promoter was unresponsive to activin ( Figure IA, left panel).
- the goosecoid DE binds an activin-inducible factor, DEBP
- DEBP DE-binding protein
- Figure IB lanes 1-3, open arrow
- the DE contains binding sites for several different DNA-binding proteins: a consensus for a paired-like homeodomain protein at its 5' end, consisting of two inverted TAAT motifs separated by 3 nucleotides (Wilson et al 1993; McKendry et al 1998; arrows, Figure IB); an additional homeodomain core bindmg site at the 3' end (arrow); a sequence reminiscent of a half site for the T-box protein, brachyury (dotted line; Kispert et al 1995), and overlapping this, a binding site for the ZFH-1 family of zinc finger homeodomain proteins (underlined; Funahashi et al 1993).
- DE ml which is mutated in the paired-like homeodomain binding site (Watabe et al 1995), competed very poorly for binding (lanes 7-9), indicating that this site is required.
- This mutant was also completely inactive in activin-responsive transcription assays (data not shown; Watabe et al 1995; McKendry et al 1998), indicating that this paired-like homeodomain binding site is absolutely required for activin-responsive transcription.
- DE m2 which is mutated in all three homeodomain binding sites, did not compete for DEBP binding at all (lanes 10- 12).
- the activin-inducible DEBP binds to the paired-like homeodomain binding site of the DE.
- the fact that the integrity of this binding site is absolutely required for all the activin-responsive transcription of the DE strongly suggests that DEBP is involved in this (McKendry et al 1998).
- low levels of maternal DEBP might mediate the component of activin- responsive transcription of the DE that does not require new protein synthesis (McKendry et al 1998; see Discussion).
- the activin-inducible DEBP can interact with Smad2 Activin signals are transduced from activated receptors to the nucleus via a complex of activated Smad2 and Smad4 (Massague, 1998). We therefore asked whether DEBP might correspond to a Smad/transcription factor complex, by analogy with the Fast-1/Smad complex, ARF.
- An antibody specific for Smad2 did not supershift DEBP, indicating that DEBP did not contain endogenous Smad2 and was thus unlikely to be a Smad transcription factor complex (data not shown). The same Smad2 antibody however could efficiently supershift the ARF complex (data not shown; see Figure 6B).
- the activin-inducible DEBP therefore appears to act as a platform for recruiting Smad2.
- UV-cross-linking experiments indicated that DEBP corresponded to a monomer of approximately 45-50 kDa (data not shown).
- DEBP binds the paired-like homeodomain binding site of the DE and is synthesized in response to activin.
- a group of transcription factors with precisely these properties are the paired-like homeodomain proteins of the Mix family. There are seven family members: Mix.l and the highly related Mix.2 (Rosa, 1989; Vize, 1996), Mixer
- Smad2C H2 Swap The region of Smad2 thought to contact Fast-1 has previously been elucidated and is the ⁇ -helix-2 (Chen et al 1998). We therefore generated a mutant in which this helix in Smad2 was replaced with the equivalent region of Smadl (Smad2C H2 Swap; Chen et al 1998). This mutant was inactive, indicating that ⁇ -helix2 of Smad2 is also required for binding to Mixer, Milk and endogenous DEBP (Table
- SIM Smad interaction motif
- TGF- ⁇ was used to stimulate the NIH3T3s as it activates Smad2 and Smad4 in the same way as activin (Liu et al 1997) and NIH3T3s respond strongly to TGF- ⁇ , and not to activin.
- Figure 6A shows a co-immunoprecipitation assay in which Flag-tagged, Mixer,
- TGF- ⁇ activation of transcription mediated by Mixer results from Mixer's interaction with the Smads, we would expect it to be independent of new protein synthesis.
- TGF- ⁇ - induced transcription via the DE is absolutely dependent on Mixer ( Figure 7B, lanes 1 ,2,5,6) and crucially is not decreased when cycloheximide was added at the same time as TGF- ⁇ (lanes 5-8).
- TGF- ⁇ induced transcription does not require on-going protein synthesis.
- the Smads appear to require other transcription factors to recruit them to DNA because they interact with DNA themselves either very weakly (Smad3 and Smad4) or not at all (Smad2; (Shi et al 1998; Hill, 1999).
- Smad3 and Smad4 are the functionally cooperative interactions such as that seen between the Drosophila homeodomain protein, tinman and MAD and MEDEA (Xu et al 1998) where no physical contact between the transcription factor and Smads has been reported.
- the direct transcription factor-Smad complexes such as that described here and for Fast-1 and also AP-1 family members (Derynck et al 1998).
- activated Smads may also be able to release repressors from DNA. Recent work suggests that the homeodomain protein Hoxc-8 functions as a repressor, and interaction with activated Smadl releases Hoxc-8 from DNA (Shi et al 1999).
- Smads The interaction of Smads with distinct transcription factors must contribute to cell- type specificity of TGF- ⁇ responses, allowing specific genes to be up-regulated only in cells where the essential co-operating transcription factor is also expressed. This is likely to be of particular significance in the patterning of the early Xenopus embryo. The same signalling pathway will activate different genes in distinct regions of the embryo depending on the particular Smad-recruiting transcription factors expressed by the cells in that region. In addition, differential affinities of specific transcription factors for Smads, coupled with the presence or absence of Smad binding sites on adjacent DNA could allow distinct genes to be activated by different levels of active Smad complexes.
- TGF- ⁇ family members This sort of mechanism might underlie the morphogenetic properties of TGF- ⁇ family members, whereby different doses of TGF- ⁇ ligands elicit different transcriptional responses (Green and Smith, 1990). Determination of the relative affinities of Mixer, Milk, Bix proteins and Fast-1 for Smad complexes will be important to test these ideas.
- CTAGCCATTAATCAGATTAACGGTGAGCAATTAGA DE-top
- CCGACTAGTATCTGCTGCCCTAAAATGTGTATTCCATGGAAATG ARE top
- CTAGCCAGTCAGCAGCTGACCGGTGAGCAATTAGA (DE ml top)
- CTAGCCAGTCATCAGAGTCACGGTGAGCAAGTCGA (DE m2 top)
- GST fusion protein purification, GST "pull-downs” and in vitro transcription/ translation Expression of GST-fusion proteins, SDS-PAGE and in vitro coupled transcription/translation in reticulocyte lysate (Promega) were performed using standard methods. Mixer, Milk and Fast-1 C-terminal deletion mutants were synthesized in vitro using linear templates generated by restriction enzyme digestion.
- [35s]-labelled transcription factors were mixed with GST- or GST-Smad2C-Se ⁇ harose beads for 2h at 4°C in 20 mM Tris pH7.5, 20% glycerol, 1 mM EDTA, 5 mM MgCl2, 0.1% NP40 and 220 mM NaCl. The beads were washed three times with five bead volumes of binding buffer, and the protein remaining bound to the beads were analysed by SDS-PAGE followed by autoradiography.
- RNA isolation and RNase protection assays were performed as described (Howell and Hill, 1997).
- the antisense probes were as follows: human ⁇ -globin (Howell and Hill, 1997); goosecoid (Blumberg et al 1991); Xenopus FGF receptor, protecting amino acids 539-580; Mixer, amino acids 173-237; Milk, amino acids 143-226.
- the Milk probe also detects a smaller product, whose size and expression characteristics are consistent with it being the protected fragment of the highly Milk-related gene Bix3 (Tada et al 1998).
- Whole mount in situ hybridizations were carried out essentially as described (Harland, 1991), using probes against goosecoid, Mixer and Milk which were identical to those used in the RNase protections, either singly or in combination.
- NIH3T3 cells were transfected using lipofectamine (Gibco BRL).
- the following amounts of plasmids were used per 6-cm dish for transcriptional assays, 0.5 ⁇ g CAT reporters, 0.2 ⁇ g of transcription factors, 0.3 ⁇ g of EF-Smad2 and EF-Smad4 and 0.5 ⁇ g EF-LacZ as an internal control for transfection efficiency, as indicated in the Figure legends.
- globin transcriptional assays 1 ⁇ g globin reporter, 0.45 ⁇ g REF-globin and 0.2 ⁇ g Mixer were transfected.
- two 6 cm plates were transfected with 0.6 ⁇ g of each plasmid.
- one 6-cm plate was transfected with 1.2 ⁇ g transcription factor.
- the amounts of DNA transfected was kept constant by adding control plasmid EF-plink as appropriate.
- cells were maintained 18 hr in DMEM containing 10% FBS, before induction by TGF- ⁇ 1 (2 ng/ml, Calbiochem) for times indicated in the Figure legends.
- oligonucleotide 1 and its complement were labelled with [ ⁇ 2p]dATP and [ ⁇ 32p]dCTP by PCR. Competitions were performed with double-stranded oligonucleotides: DE ml, DE m2, DE m3 or ARE (produced by annealing and filling in oligonucleotides 2 and 3).
- Whole cell Xenopus embryo extracts were prepared by homogenizing embryos in buffer (10 ⁇ l per embryo) containing 200 mM KCl, 50 mM Tris-HCl pH 7.4, 10% glycerol, 25 mM ⁇ -glycerophosphate, 1 mM EGTA, 1 mM EDTA, 2 mM DTT, and protease inhibitors as above. Lysates were cleared by repeated centrifugation. Binding reactions were performed with 30 ⁇ g of protein extract incubated with 0.2 ng DE probe in 20 ⁇ l of buffer containing 140 mM KCl, 8 mM MgCl2, 12.5 mM ⁇ -glycerophosphate, 1 mM EGTA, 1 mM
- the final buffer concentrations were 8 mM Hepes pH 7.6, 90 mM KCl, 5 mM MgCl2, 4 mM ⁇ -glycerophosphate, 40 ⁇ M EDTA, 40 ⁇ M Spermidine, 2 ⁇ g poly(dl-dC), 5% glycerol.
- Extracts from NIH3T3 cells transfected with Flag- tagged transcription factors were prepared as described (Marais et al 1993) and final bandshift conditions were: 14 ⁇ g total protein in 10 mM Hepes pH 7.5, 15% glycerol, 210 mM KCl, 5.5 mM MgCi2, 0.2% Triton, 5 mM EGTA, 2.5 mM
- EDTA 2 ⁇ g poly(dl-dC), 0.25 mM DTT and protease inhibitors with 0.2 ng labelled probe.
- Specific antibodies (1 ⁇ l) anti-Smad2 (Nakao et al 1997), anti- Smad4 (B8; Santa Cruz) or anti-Flag were added to the binding reactions. In all cases electrophoresis was in 5% polyacrylamide gels/0.5X TBE containing 2.5% glycerol.
- the wild type SIM-containing peptide used in Figure 5 was: Biotin.Aminohexanoicacid-
- RQIKIWFQNRRMKWKKLLMDFNNFPPNKTITPDMNVRIPPI The first 16 amino acids are from the helix 3 of Antennapedia which allows internalization of these peptides into live cells (Derossi et al 1998); the last 25 amino acids are codons 283-307 of Mixer. The mutant was the same except that the 2 prolines at positions 26 and 27 were alanines. Peptides were included in the binding reactions at the concentrations given in the Figure legend.
- ⁇ - crystallin enhancer binding protein ⁇ EFl is a zinc f ⁇ nger-homeodomain protein implicated in postgastrulation embryogenesis. Development 119, 433-446.
- RhoA RhoA
- XSmad2 directly activates the activin- inducible, dorsal mesoderm gene XFKH1 in Xenopus embryos. EMBO J. 16, 7411-7421.
- Xenopus Smad4 ⁇ is the co-Smad component of developmentally-regulated transcription factor complexes responsible for induction of early mesodermal genes. Dev. Biol. in press.
- TGF- ⁇ superfamily new members, new receptors, and new genetic tests of function in different organisms. Genes Dev. 8, 133-146.
- Brachyury is a tissue-specific transcription factor. EMBO J. 14, 4763-4772.
- FAST-2 is a mammalian winged-helix protein which mediates transforming growth factor ⁇ signals. Mol.
- the SRF accessory protein Elk-1 contains a growth factor-regulated transcriptional activation domain.
- the cdc2-related protein p40MO15 is the catalytic subunit of a protein kinase that can activate p33cdk2 and p34cdc2.
- Smad proteins act in combination with synergistic and antagonistic regulators to target
- PPTK does not appear to form part of a functional SIM in the contect of Bix 1 , but there are other differences between the SIM region of Bixl and the Mixer SIM which may be responsible for the apparent inability of Bixl to interact with GSTSmad2C. However, it remains possible that in other contexts PPTK would be functional.
- Figure 13 shows a line up of functional SIMs, including the new Zebrafish Mixer. It also shows the line up of the SIM region from all the known Mix family members. It is clear that the two Mixers, Milk and Bix3 contain a SIM, but Bixl, Bix4, Mix.l and Mix.2 do not. Experiments discussed below indicate that those that contain recognizable SIMs bind GSTSmad2C and those that do not, do not bind GSTSmad2C. This confirms that the SIM is responsible for the interaction with GSTSmad2C. Zebrafish Fast-1 also has a SIM (not shown). XFast-3 has a SIM that appears to be functional in this protein.
- the SIM peptides work as inhibitors of Smad2/3-transcription factor interactions in vitro and in vivo.
- the formation of transcription factor/Smad complexes that contain Smad2 or Smad3 can be inhibited both in vitro and in vivo.
- the TGF- ⁇ - induced transcriptional activation of the junB gene can be inhibited. This indicates that interfering with Smad/transcription factor interactions in vivo inhibits a biological response to TGF- ⁇ .
- the SIM peptide fused to Antennapedia third helix is transported into cells and reaches the nucleus.
- TGF- ⁇ -responsive reporter cell lines useful for testing inhibitors of TGF- ⁇ signalling.
- TGF- ⁇ responsive reporter cell lines can be used to test potential inhibitors of TGF- ⁇ signalling, for example peptides or small molecules.
- SIM Smad interaction motif
- FIG. 13 shows SIMs in different members of the FAST and Mix families from different species.
- Figure 14 relates to characterisation of Xenopus FAST3 (XFAST3) and complexes which comprise XFAST3.
- XFast-3 can also form complexes with Smad2 and Smad4 in tissue culture cells (see Figure 16B).
- This is a highly cooperative complex that can not be disrupted by the Mixer SIM peptide in vitro. However it is destroyed when the peptide is added to the cells in vivo prior to TGF- ⁇ stimulation, indicating that the peptide can prevent XFast- 3/Smad2/Smad4 complexes forming in vivo and that the major Smad2/XFast-3 interaction is through the SIM ( Figure 17B).
- this peptide is important for the applications of this peptide as an in vivo inhibitor of Smad2 activity and therefore TGF- ⁇ /activin inducible transcription. It can clearly prevent active complexes forming in vivo. In addition, this data suggests that the XFast-3 SIM may be stronger than the Mixer SIM.
- XFast-3 The SIM in XFast-3 is functional. XFast-3 made in reticulocyte lysate bound to the ARE will interact with GSTSmad2C to give a supershift using the methods described in Example 1 and Germain et al. , 2000. XFast-3 that is C-terminally truncated so that the SIM is no longer present, does not bind GSTSmad2C efficiently in this assay (data not shown).
- a fluorescently-labelled SIM peptide derived from XFast-3 fused to the Antennapedia third helix can prevent the formation of a XFast-1 /Smad2/Smad4 complex in vitro and formation of a DNA-bound Smad3/Smad4-containing complex in vitro.
- An equivalent mutant peptide cannot. This indicates that the XFast-3 SIM is capable of specifically interacting with both Smad2 and Smad3.
- the family members that interact with GSTSmad2C are Mixer, Milk and Bix3 and Zebrafish Mixer. Bixl, Bix4 and Mix. l do not interact with GSTSmad2C. This correlates precisely with the presence of a recognizable SIM in Xenopus and Zebrafish
- the SIM alone is sufficient to confer TGF- ⁇ inducibility in vivo
- a Gal4 DNA-binding domain (Gal4(l-95)) (Sadowski and Ptashne, 1989) fusion of the SIM (residues 283-307 of Mixer) and a mutant version with the two prolines of the PPNK mutated to alanine.
- the Gal4(l-95)-SIM can confer approximately 8-fold TGF- ⁇ - inducible transcription onto this reporter.
- the Gal4(l-95)-mutant SIM is completely inactive.
- the TGF- ⁇ inducible transcription mediated by Gal4(l-95)- SIM is competed by overexpression of Mixer or Fast-1, but not by Mixer mutated in the two prolines in the PPNK sequence.
- the Mixer SIM peptide can disrupt formation of Smad2/3-transcription factor DNA complexes in vitro.
- the data indicate that the Mixer SIM peptide can efficiently disrupt formation of the Smad3/Smad4-containing complex and the XFast-1 /Smad2/Smad4 complex in vitro, but not the XFast-3/Smad2/Smad4 complex, perhaps because the interaction of XFast-3 with Smad2 is stronger than the Mixer SIM peptide interaction with Smad2 (see above). Thus the SIM peptide therefore specifically interacts with Smad2 and Smad3 in vitro.
- the SIM peptide is efficiently taken up by HaCaT and NIH3T3 cells.
- the Mixer SIM peptide and the mutant Mixer SIM peptide are taken up by the both NIH3T3 cells and by HaCaT cells when incubated in the normal growth media (10% FCS/DMEM). They are fused to the protein transduction domain, the Antennapedia helix 3, which is why they translocate the plasma membrane. Concentrations that have been tested are between 5 ⁇ M and 40 ⁇ M.
- the peptide is found throughout the cells in the cytoplasm and nucleus. The SIM peptide, but not the mutant SIM peptide further accumulates in the nucleus when the cells are treated with TGF- ⁇ .
- SIM peptide is associated with Smad2 and Smad3 which are cytoplasmic in untreated cells. Some peptide will also be uncomplexed. Upon TGF- ⁇ stimulation, the Smad2 and Smad3 translocate to the nucleus, and take the associated peptide with them. The mutant SIM peptide does not show this behaviour probably because it cannot bind the Smads.
- SIM peptide binds Smad2 and Smad3 in vivo and inhibits these Smads forming active DNA-binding complexes with different trancription factors in a TGF- ⁇ -inducible manner.
- SIM peptide but not the mutant peptide, inhibits TGF- ⁇ induction of the junB gene by approximately 30%. This is very significant, as it demonstrates that the peptide can inhibit physiological TGF- ⁇ responses in vivo.
- TGF- ⁇ signalling contributes to diseases such as cancer and fibrosis is through its ability to regulate the transcription of target genes
- our ability to inhibit TGF- ⁇ induction of transcription of target genes indicates that the SIM peptide (or a small molecule with the same activity) may be an efficient method by which to inhibit TGF- ⁇ responses.
- TGF- ⁇ Inducible Stable Reporter gene Cell Lines We have shown that the distal element (DE) of the goosecoid promoter is TGF- ⁇ inducible and that this inducibility is dependent on the presence of Mixer and active Smad2/Smad4 complexes in transiently transfected NIH3T3 cells (Example 1 and Germain et al., 2000). Similarly we have shown that the activin response element (ARE) confers XFast-1 dependent TGF- ⁇ inducibility on a
- Stable cell lines may be used to assay potential TGF- ⁇ signal transduction pathway inhibitors.
- NIH3T3 and HaCaT cell lines may be employed for this purpose.
- the DE and ARE elements may be cloned into, for example, the destabilised enhanced green fluorescent protein promoter cloning vector (pdEGFP-1, Clontech) to generate pDEdEGFP-1 and ⁇ AREdEGFP-1.
- pdEGFP-1 the destabilised enhanced green fluorescent protein promoter cloning vector
- pdEGFP-1 the destabilised enhanced green fluorescent protein promoter cloning vector
- These plasmids carry the neomycin drug resistance gene and hence cells transfected stably with these plasmids can be selected for by growth in media containing G418 (Geneticin, Gibco BRL).
- the DE and ARE may also be cloned into the secreted alkaline phosphatase reporter gene plasmid pSEAP-2 (Clontech), and into luciferase reporter gene plasmid pGL3-basic (Promega). Stable selection of cell lines carrying these reporter genes may be performed by co-transfecting the plasmid TKNeo (Cruzalegui et al, 1999). Following isolation of stable clones carrying these reporter genes, these clones may be transfected with expression plasmids for Mixer (for DE reporters) and XFast-1 (for ARE reporters).
- XFast-1 may be cloned into the episomal eukaryotic expression vector pCEP4 (Invitrogen).
- This plasmid contains the hygromycin resistance gene, the Epstein-Barr Virus plasmid origin of replication, the EBNA-1 gene and drives expression of the gene of interest from the CMV immediate early promoter.
- Stable cell lines which carry both the reporter and the transcription factor may be selected for by growth in media containing both G418 and Hygromycin (Boehringer Mannheim). Similar cell lines may also be generated using the TGF ⁇ -inducible reporter driven by 12 Smad-binding sites from the PAI-1 gene (Dennler et al., 1998). In this case, no transcription factors have to be co- expressed.
- Effects of the peptide on reporter gene expression may be analysed by preicubating the cell lines with peptide for 30 minutes prior to TGF- ⁇ induction for 8 h or longer.
- GFP production may be assessed using a confocal microscope and SEAP production can be assayed by sampling the media of the cells and measuring SEAP production using the Great escAPETM SEAP fluorescent detection kit (Clontech) and a microtitre plate benchtop fluorimeter (Perspective Biosystems). Luciferase can be measured in cell extracts using a luminometer.
- These cell lines may also be used as the basis of screens for cell soluble small compounds that can interfere with the TGF- ⁇ signalling pathway.
- Testing in animals may also be useful in identifying or characterising compounds, or in assessing their effects. Because of the high level of conservation of the TGF ⁇ signalling system and components thereof, as discussed above, for example between Xenopus and human, it may be appropriate to carry out tests on animals that are not transgenic for components of the TGF ⁇ signalling pathway. However, it may be convenient to use transgenic animals, for example a transgenic animal modified to facilitate detection of modulation of the TGF ⁇ signalling pathway, for example in a manner analogous to the reporter cell lines discussed above.
- the Antennapedia third helix is underlined.
- the peptides were purified by reverse phase HPLC using an Aquapore ODS 20 micron column (Anachem) in 0.08% trifluoroacetic acid in a gradient of acetonitrile.
- bandshift assays and transfections were as described in Germain et al., 2000.
- the luciferase reporter assays were performed as described by Jonk et al., 1998, and the ⁇ -gal assays were performed using CDGP (Calbiochem) as a substrate and were quantitated spectrophotometrically.
- CDGP Calbiochem
- Peptides were dissolved in water, and added to the growth media of the cells for the stated times.
- TGF- ⁇ signalling pathway in cancer may be useful.
- the following is a selection of references showing that tumours secrete TGF- ⁇ and that it promotes tumour formation and invasiveness in vitro and in vivo.
- TGF- ⁇ Overexpression of TGF- ⁇ in human tumours: Derynck et al., 1987 demonstrates that a variety of human tumours overexpress TGF- ⁇ . Gomella et al., 1989 concerns enhanced expression of TGF- ⁇ in renal cell carcimoma. Steiner and Barrack, 1992 indicates that TGF- ⁇ is overproduced in prostate cancer.
- TGF- ⁇ causes increased tumorigeniciry and inhibiting TGF- ⁇ signalling by various means inhibits tumour invasiveness and metastasis: Welch et al. , 1990 indicates that TGF- ⁇ stimulates mammary adenocarcinoma cell invasion and metastatic potential. Arteaga et al., 1993a indicates that TGF- ⁇ can induce estrogen-independent tumorigenicity of human breast cancer cells in athymic mice. Chang et al., 1993 suggests that increased TGF- ⁇ expression inhibits cell proliferation in vitro, yet increases tumorigenicity and tumor growth in Meth A sarcoma cells.
- TGF- ⁇ l and Ha-Ras collaborate in modulating the phenotypic plasticity and invasiveness of epithelial tumor cells.
- the invasive phenotypes of the cells is entirely dependent on TGF- ⁇ signalling and can be inhibited by neutralizing TGF- ⁇ antibodies.
- TGF- ⁇ signaling is necessary for carcinoma cell invasiveness and metastasis.
- Several human carcinoma lines lost invasiveness when treated with neutralizing TGF- ⁇ antibodies or soluble receptor variants.
- Portella et al., 1998 indicates that TGF- ⁇ is sufficient to significantly enhance tumorigenicity and the maligant and invasive characteristics of the tumor in vivo.
- TGF- ⁇ signalling pathway was inhibited by overexpresison of a dominant negative TGF- ⁇ type II receptor.
- Yin et al., 1999 indicates that TGF- ⁇ signaling blockade inhibits parathyroid hormone related protein secretion by breast cancer cells and bone metastases development.
- Lehmann et al., 2000 suggests that the ERK MAP kinase pathway synergizes with TGF- ⁇ in promoting malignacy. This is reversible by treating cells with neutralizing TGF- ⁇ antibodies.
- TGF- ⁇ antibodies inhibit breast cancer cell tumorigenicity and increase mouse spleen natural killer cell activity. Implications for a possible role of tumor cell/host TGF- ⁇ interactions in human breast cancer progression. J Clin Invest, 92, 2569- 2576. (1993c)
- TGF ⁇ l inhibits the formation of benign skin tumors, but enhances progression to invasive spindle carcinomas in transgenic mice. Cell, 86, 531-542. (1996)
- TGF beta 1 Transforming growth factor beta 1 (TGF beta 1) is an autocrine positive regulator of colon carcinoma U9 cells in vivo as shown by transfection of a
- TGF beta 1 antisense expression plasmid Cell Growth Differ, 6, 1635-
- TGF ⁇ production while blocking its apoptotic but not invasive responses a mechanism leading to increased malignancy in epithelial cells.
- Genes Dev. in press. (2000) Nakao, A. , Imamura, T. , Souchelnytskyi, S., Kawabata, M. , Ishisaki, A. , Oeda, E., Tamaki, K., Hanai, J., Heldin, C.H. , Miyazono, K. and ten Dijke, P. TGF- ⁇ receptor-mediated signalling through Smad2, Smad3 and Smad4. EMBO J. 16, 5353-5362. (1997)
- Vize P.D. DNA sequences mediating the transcriptional response of the Mix.2 homeobox gene to mesoderm induction. Dev. Biol. Ill, 226-231.(1996)
- Any method which requires the binding of one of the partners to a solid phase and then measurement of th ebinding of the second partner to it may be used.
- direct (including SPA) or indirect (including ELISA, displacement) radiochemical, enzymatic or fluorescent methods may be used.
- ELISA type assays where either Smad or SIM is chemically or electrostatically bound to a microwell plate and the binding of the partner molecule detected by enzyme linked antibodie(s) to the partner molecule.
- SPA Scintillation proximity assays
- Displacement assays where the binding of Smad to SIM is detected by immobilising one partner on a surface and measuring the displacement or binding of labelled (radiochemical or fluorescent) partner.
- Fluorescence resonance energy transfer measuring the Smad-SIM interaction by labelling each with a different fluor, the fluorescent wavelength of one giving rise to fluorescence in the other only when the partners are in close proximity.
- FRET Fluorescence resonance energy transfer
- Fluorescence correlation microscopy measuring the diffusion time of either labelled Smad2 or SIM in the presence and absence of the other partner by confocal FCS.
- tumour cells have an intact TGF- ⁇ signalling pathway
- late stage and metastatic tumours actively overexpress TGF- ⁇ 1 which acts as a potent tumour promoter. It has direct effects on the tumour cells and also indirect effects through its ability to induce angiogenesis, immunosuppression and alterations in stromal tissue plasticity (Akhurst and Balmain, 1999).
- the direct tumour promoting effects of TGF- ⁇ on the epithelial cells have been well characterized and involve an EMT (epithelial-to- mesenchymal transition) in which the cells loose their polarized phenotype, down-regulate epithelial markers such as E-cadherin and become fibroblastoid in character (Akhurst and Balmain, 1999).
- tumours in vivo have been shown to be reversed by inhibiting TGF- ⁇ signalling either with neutralizing antibodies or by overexpressing dominant negative TGF- ⁇ type II receptors (Oft et al 1998; Oft et al 1996; Portella et al 1998). This indicates that the maintenance of the tumour phenotype requires TGF- ⁇ signalling.
- EMT epithelial-to-mesenchymal transition
- Normal MDCK cells which have approximately equal levels of Smad2, Smad3 and Smad4, respond to TGF- ⁇ by growth arresting and dying by apoptosis.
- the cells secrete substantial amounts of active TGF- ⁇ and become completely fibroblastoid, are invasive in collagen gels, and no longer growth arrest or apoptose in response to TGF- ⁇ . They require autocrine TGF- ⁇ signalling to maintain this phenotype.
- the SIM peptides' or test compounds' ability to reverse EMT in Raf-1 - expressing MDCK cells may be measured.
- Neutralizing TGF- ⁇ antibodies can inhibit EMT, as can expression of dominant negative TGF- ⁇ type II receptor (Lehmann et al 2000; Oft et al 1998; Oft et al 1996). Inhibition of specific Smad interactions may also reverse EMT in these cells.
- the ability of SIM peptides or test compounds to reverse invasiveness of metastatic carcinoma cells such as the mouse colon carcinoma cells (CT26) which can be reverted to an epithelial phenotype by TGF- ⁇ neutralizing antibodies (Oft et al 1998) may also be measured. The results from these experiments may indicate whether a peptide or other test compound has potential as an inhibitor of TGF- ⁇ -mediated tumour progression, and may indicate how specific the peptide or compound may be.
Landscapes
- Chemical & Material Sciences (AREA)
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Organic Chemistry (AREA)
- General Health & Medical Sciences (AREA)
- Molecular Biology (AREA)
- Biochemistry (AREA)
- Biophysics (AREA)
- Zoology (AREA)
- Genetics & Genomics (AREA)
- Medicinal Chemistry (AREA)
- Gastroenterology & Hepatology (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Toxicology (AREA)
- Peptides Or Proteins (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
- Investigating Or Analysing Biological Materials (AREA)
- Micro-Organisms Or Cultivation Processes Thereof (AREA)
Abstract
Description
Claims
Applications Claiming Priority (3)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
GB9920000 | 1999-08-25 | ||
GBGB9920000.8A GB9920000D0 (en) | 1999-08-25 | 1999-08-25 | Polypeptides |
PCT/GB2000/003265 WO2001014413A2 (en) | 1999-08-25 | 2000-08-25 | Polypeptides capable of interacting with the smad peptide and comprising the sequence pp (t/n)k |
Publications (1)
Publication Number | Publication Date |
---|---|
EP1212414A2 true EP1212414A2 (en) | 2002-06-12 |
Family
ID=10859702
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
EP00954771A Withdrawn EP1212414A2 (en) | 1999-08-25 | 2000-08-25 | Polypeptides capable of interacting with the smad peptide and comprising the sequence pp(t/n)k |
Country Status (5)
Country | Link |
---|---|
EP (1) | EP1212414A2 (en) |
AU (1) | AU6712600A (en) |
CA (1) | CA2382852A1 (en) |
GB (1) | GB9920000D0 (en) |
WO (1) | WO2001014413A2 (en) |
Families Citing this family (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
GB0105063D0 (en) * | 2001-03-01 | 2001-04-18 | Imp Cancer Res Tech | Polypeptides |
CN103558395B (en) * | 2013-10-28 | 2015-08-26 | 深圳市第二人民医院 | The application of SMAD3 gene on detecting in bladder transitional cell carcinoma |
CN115960167B (en) * | 2022-08-18 | 2023-11-07 | 齐齐哈尔大学 | Corn anti-adhesion peptide and preparation method and application thereof |
Family Cites Families (6)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US6184205B1 (en) * | 1994-07-22 | 2001-02-06 | University Of North Carolina At Chapel Hill | GRB2 SH3 binding peptides and methods of isolating and using same |
FR2726471B1 (en) * | 1994-11-07 | 1997-01-31 | Pf Medicament | PROCESS FOR IMPROVING THE IMMUNOGENICITY OF AN IMMUNOGENIC COMPOUND OR A HAPTENA AND APPLICATION TO THE PREPARATION OF VACCINES |
FR2766192B1 (en) * | 1997-07-17 | 2001-07-13 | Pf Medicament | EPITOPES OF RSV AND ANTIBODIES COMPRISING SAME, USEFUL IN DIAGNOSIS AND THERAPY |
AU774694B2 (en) * | 1998-12-16 | 2004-07-01 | Temple University - Of The Commonwealth System Of Higher Education | Inhibition of angiogenesis by high molecular weight kininogen domain 3 peptide analogs |
AU2736600A (en) * | 1999-01-26 | 2000-08-07 | Scripps Research Institute, The | Control of virus infection using replication associated proteins, compositions and methods of use |
GB9909133D0 (en) * | 1999-04-22 | 1999-06-16 | Peptide Therapeutics Ltd | Inhibition of endopeptidase by cystatins |
-
1999
- 1999-08-25 GB GBGB9920000.8A patent/GB9920000D0/en not_active Ceased
-
2000
- 2000-08-25 EP EP00954771A patent/EP1212414A2/en not_active Withdrawn
- 2000-08-25 WO PCT/GB2000/003265 patent/WO2001014413A2/en not_active Application Discontinuation
- 2000-08-25 CA CA002382852A patent/CA2382852A1/en not_active Abandoned
- 2000-08-25 AU AU67126/00A patent/AU6712600A/en not_active Abandoned
Non-Patent Citations (1)
Title |
---|
See references of WO0114413A3 * |
Also Published As
Publication number | Publication date |
---|---|
AU6712600A (en) | 2001-03-19 |
GB9920000D0 (en) | 1999-10-27 |
WO2001014413A3 (en) | 2001-09-07 |
WO2001014413A2 (en) | 2001-03-01 |
CA2382852A1 (en) | 2001-03-01 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
US20020144298A1 (en) | Novel human genes and gene expression products | |
JP2010143936A (en) | Zinc finger binding domain for gnn | |
JP3633616B2 (en) | Transcription factor DP-1 | |
Tkachenko et al. | Misexpression of disrupted HMGI architectural factors activates alternative pathways of tumorigenesis | |
KR20060054196A (en) | Transducible dna-binding protein | |
WO2000012702A2 (en) | Human genes differentially expressed in colorectal cancer | |
KR100766952B1 (en) | Regulatory zinc finger proteins | |
AU732766B2 (en) | CD95 regulatory gene sequences and transcription factors | |
US5905146A (en) | DNA binding protein S1-3 | |
US7258973B2 (en) | Method for detecting a differentially expressed sequence | |
Hollington et al. | Expression and localization of homeodomain proteins DLX4, HB9 and HB24 in malignant and benign human colorectal tissues | |
AU726356B2 (en) | Novel methods for the characterization of compounds which stimulate STF-1 expression in pancreatic islet cells | |
WO2001014413A2 (en) | Polypeptides capable of interacting with the smad peptide and comprising the sequence pp (t/n)k | |
WO2002070702A2 (en) | Polypeptides capable of interacting with smad polypeptide | |
WO1998022585A1 (en) | HUMAN POLYHOMEOTIC 2(hph2) ACTS AS AN ONCOGENE | |
WO2002018545A1 (en) | Genetic demonstration of requirement for nkx6.1, nkx2.2 and nkx6.2 in ventral neuron generation | |
US6673911B1 (en) | Human polyhomeotic 2 (hph2) acts as an oncogene | |
US20040248139A1 (en) | Eit-6, a polypeptide encoded by an estrogen regulated gene | |
Huang | Identification and characterization of Krk-1, a novel kidney-specific KRAB-domain-containing zinc finger protein | |
US20040053210A1 (en) | Genetic demonstration of requirement for nkx6.1, nkx2.2 and nkx6.2 in ventral neuron generation | |
JP2003061672A (en) | New human bmcc1 gene | |
EP1593687A2 (en) | Human genes differentially expressed in colon cancer | |
CA2385234A1 (en) | Nrage nucleic acids and polypeptides and uses thereof | |
AU5406501A (en) | CD95 regulatory gene sequences and transcription factors |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
PUAI | Public reference made under article 153(3) epc to a published international application that has entered the european phase |
Free format text: ORIGINAL CODE: 0009012 |
|
17P | Request for examination filed |
Effective date: 20020321 |
|
AK | Designated contracting states |
Kind code of ref document: A2 Designated state(s): AT BE CH CY DE DK ES FI FR GB GR IE IT LI LU MC NL PT SE |
|
AX | Request for extension of the european patent |
Free format text: AL;LT;LV;MK;RO;SI |
|
17Q | First examination report despatched |
Effective date: 20020719 |
|
RAP1 | Party data changed (applicant data changed or rights of an application transferred) |
Owner name: CANCER RESEARCH TECHNOLOGY LIMITED |
|
RAP1 | Party data changed (applicant data changed or rights of an application transferred) |
Owner name: CANCER RESEARCH TECHNOLOGY LIMITED |
|
STAA | Information on the status of an ep patent application or granted ep patent |
Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN |
|
18D | Application deemed to be withdrawn |
Effective date: 20050322 |