AU9717301A - Dendritic cell receptor - Google Patents

Dendritic cell receptor Download PDF

Info

Publication number
AU9717301A
AU9717301A AU97173/01A AU9717301A AU9717301A AU 9717301 A AU9717301 A AU 9717301A AU 97173/01 A AU97173/01 A AU 97173/01A AU 9717301 A AU9717301 A AU 9717301A AU 9717301 A AU9717301 A AU 9717301A
Authority
AU
Australia
Prior art keywords
dec
ligand
polypeptide
human
seq
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.)
Abandoned
Application number
AU97173/01A
Inventor
Derek Nigel John Hart
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Individual
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Individual filed Critical Individual
Priority to AU97173/01A priority Critical patent/AU9717301A/en
Publication of AU9717301A publication Critical patent/AU9717301A/en
Abandoned legal-status Critical Current

Links

Landscapes

  • Peptides Or Proteins (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Description

Regulation 3.2
AUSTRALIA
Patents Act 1990 DIVISIONAL APPLICATION Name of Applicant: Actual Inventor(s): Address for Service: Invention Title: Derek Nigel John HART Derek Nigel John HART DAVIES COLLISON CAVE, Patent Attorneys, Level 3, 303 Coronation Drive, Milton, Queensland, 4064, Australia "Dendritic cell receptor" Details of Parent Application No: 29176/97 The following statement is a full description of this invention, including the best method of performing it known to me/us: Q:\Oper\Vpa\Deccmber\Dcc 2001\2478039 div har.345.doc 1/12101 DENDRITIC CELL RECEPTOR FIELD OF THE INVENTION This invention relates to dendritic cell receptors. In particular, it relates to human DEC- 205, to the production and use thereof, and to ligands which bind to it. Human DEC-205 and its ligands are useful in prophylaxis and therapy.
BACKGROUND OF THE INVENTION f* Dendritic cells perform important immunoregulatory functions by presenting antigens in the form of peptides bound to cell-surface major histocompatability complex (MHC) molecules to T cells. Identification of the mechanisms by which this antigen presentation function is achieved therefore has important implications in manipulating immune response in prophylaxis and therapy, particularly in humans.
Jiang et al, Nature 375: 151-155 (1995) disclose a murine dendritic cell receptor having a molecular weight of 205 kDa (murine DEC-205). However, they do not disclose a receptor on human dendritic cells.
The applicant has now identified a receptor on human dendritic cells. It is broadly to this receptor (likely to be the human homolog of murine DEC-205) that the present invention is directed.
SUMMARY OF THE INVENTION The present invention has a number of aspects. In a first aspect, the invention provides an isolated polypeptide corresponding to which has an approximate molecular weight of 198- 205 kDa and which includes the following amino acid sequences:
TVDCNDNQPGAICYYSGNETEKEVKPVDSVKCPSPVLNTPWIPFQN
-2-
CCYNFIITKNRHMATTQDEVHTKCQKLNPKSHILSIRDEKENNFVLEQLLYFNYM
ASWVMLGITYRNNSL; and (ii) SQHRLFHLHSQKCLGLDITKSVNELRMFSCDSSAML; or a functionally equivalent fragment thereof.
In a further aspect, the invention provides an isolated polypeptide, which comprises the amino acid sequence shown in Figure 11 (SEQ ID No:2) or a functionally equivalent 10 fragment thereof.
In a still further aspect, the invention provides an isolated polypeptide, which comprises amino acids 27 to 1722 shown in Figure 11 (SEQ ID No:2).
In yet a further aspect, the invention provides an extracellular domain of a polypeptide as broadly described above or a functionally-equivalent fragment thereof.
In a preferred embodiment, the extracellular domain includes at least a portion of carbohydrate recognition domain (CRD7), spacer, and a portion of carbohydrate recognition domain (CRD8) of human DEC-205 (amino acids 1208 to 1323 of the amino acid sequence of Figure 11 (SEQ ID No:2)).
o In a still further aspect, the invention provides a polynucleotide encoding a polypeptide as broadly described above or its extracellular domain as broadly described above. This polynucleotide is preferably DNA, more preferably cDNA, but can also be RNA.
In a specific embodiment, the polynucleotide coding for human DEC-205 includes the following nucleotide sequences: (iii) A ACA GTT GAT TGC AAT GAC AAT CAA CCA GGT GCT ATT TGC TAC TAT TCA GGA AAT GAG ACT GAA AAA GAG GTC AAA CCA GTT GAC AGT GTT AAA TGT CCA TCT CCT GTT CTA AAT ACT CCG TGG ATA CCA TTT CAG AAC TGT TGC TAC AAT TTC ATA ATA ACA AAG AAT AGG CAT ATG GCA ACA ACA CAG GAT GAA GTT CAT ACT AAA TGC CAG AAA CTG AAT CCA AAA TCA CAT ATT CTG AGT ATT CGA GAT GAA AAG GAG AAT AAC TTT GTT CTT GAG CAA CTG CTG TAC TTC AAT TAT ATG GCT TCA TGG GTC ATG TTA GGA ATA ACT TAT AGA AAT AAX TCT CCT; and (iv) GAG GAC AAG TTA TGG AAG TGG GTG TCC CAG CAT CGG CTC TTT CAT TTG CAC TCC CAA AAG TGC CTT GGC CTC GAT ATT ACC AAA TCG GTA AAT GAG CTG AGA ATG TTC AGC TGT GAC TCC AGT GCC ATG CTG TGG TGG AAA TGC GAG CAC CA wherein X is T or G.
In a further embodiment, the polynucleotide comprises part or all of the nucleotide sequence of Figure 10 (SEQ ID No:1).
In yet a further aspect, the invention provides a vector including a polynucleotide as 20 broadly described above.
In still a further aspect, the invention provides a method of producing a polypeptide as broadly described above, the extracellular domain thereof or a functional fragment comprising the steps of: culturing a host cell which has been transformed or transfected with a vector as broadly described above to express the encoded polypeptide, extracellular domain or fragment; and recovering the expressed polypeptide, extracellular domain or fragment.
As yet an additional aspect, the invention provides a ligand that binds specifically to a polypeptide as broadly described above or its extracellular domain as defined above.
C
-3A- Preferably, the ligand is an antibody or antibody binding fragment or carbohydrate bearing protein.
The antibody or antibody binding fragment can be used in methods for extracting or isolating activated dendritic cells.
In still a further aspect, the invention provides a construct for use in therapy or prophylaxis. The construct will usually be a ligand-antigen construct or a DEC-205antigen construct although ligand-toxin and DEC-205-toxin constructs are also contemplated. The ligand-antigen construct preferably consists of an antibody or antibody binding fragment which binds to a polypeptide as broadly described above and a hostprotective antigen. The DEC-205-antigen construct preferably consists of at least the extracellular domain of a polypeptide as broadly described above and a host-protective antigen.
In yet further aspects, the invention contemplates methods of therapy or prophylaxis which employ a polypeptide as broadly described above, ligands or constructs containing them.
-4- In yet a further aspect, the invention provides a molecule (hapten) which may be used to generate antibodies for identifying or purifying human dendrtic cells, which inciudes a peptide based upon part or all of the sequence of Figure 11 (SEQ ID No:l).
DESCRIPTION OF THE DRAWINGS While the invention is broadly as defined above, it will be appreciated by those persons skilled in this art that it is not limited thereto and that it includes embodiments more particularly described below. It will also be better understood by reference to the accompanying drawings, in which Figure 1 shows the structure of human DEC-205; Figure 2 shows the strategy for isolation of human DEC-205 cDNA.
15 A. A schematic presentation of human DEC-205 mRNA with the regions corresponding to DEC-205 domains. The positions of the primers used for the cDNA cloning and analysis are indicated with arrows. The positions of reverse transcriptase-polymerase chain reaction (RT-PCR) fragments 1 to 6 and the clone pBK14-1 are indicated with bars: B. RT-PCR amplification of fragment 1 and 2 from L428 and HEL cell line RNA.
20 L428 and HEL cells were subjected to RT-PCR with two pairs of degenerate primers (DEC-a/-b, and DEC-d/-e), fractionated by electrophoresis through 2% agarose gel, and stained with ethidium bromide. C. RT-PCR and 3'-RACE amplification of fragment 3 and 4 from L428 cells using the primers 028/023 and 029/019, respectively. A cDNA pool of L428 cells was subjected to 3'-RACE and RT-PCR, electrophoresed through 25 0.8% agarose gel, and stained with ethidium bromide. The numbers on the top correspond to the name of fragment in Fig. 2A. The positions of DNA molecular size standard are indicateds to the right. The estimated molecular size of the RT-PCR products are indicated to the left; Figure 3 shows protein similarity between human and mouse DEC-205.
A. The predicted amino acid sequence of human DEC-205 is aligned with the mouse homolog. The regions corresponding to DEC-205 domain structure are bracketed. The positions of amino acids are shaded where there are identical or conservatively replaced amino acids between the sequences, and the asterisks indicates conserved cysteines. The diamonds indicates potential N-glycosylation sites conserved between the sequences.
The arrow indicates one amino acid deletion in CRD-5 of human DEC-205. The circles indicate conserved potential serine-phosphorylation sites by protein kinase C (open circle) or casein kinase (closed circle). B. The identity between human and mouse DEC-205 is indicated above each domain (boxed, See Fig. 2A for key); Figure 4 shows that human DEC-205 is probably a one-copy gene. Genomic
DNA
isolated from the peripheral blood of four individuals was digested with the restriction enzymes BglII, BamHI, HindI or EcoRI and subjected to Southern blot analysis with the 32 P]cysteine-rich domain probe. The final wash was 0.3 x SSC at 65 C. The positions of the DNA molecular size standards are indicated to the right; Figure 5 shows that human DEC-205 gene localizes on chromosome 2.
A somatic cell hybrid panel blot (restriction-digested with PstI) was subjected to Southern blot analysis with the 3 P]cysteine-rich domain probe. The final wash was 0.3 x SSC at 65 0 C. The positions of the DNA molecular size standards are indicated to the 15 right. The estimated molecular size of the probe-specific bands are indicated to the left.
The asterisk indicates weakly hybridized bands. M, male; F, female; Figure 6 shows that human DEC-205 gene maps to chromosome band 2q24. A. A metaphase spread of human chromosomes were subjected to fluorescent in situ 20 hybridization (FISH) with 6.6 kb human DEC-205 cDNA probe. The final wash was 0.1 x SSC at 60°C. The FISH image was overlaid with a DAPI-stained chromosome image.
The DEC-205 specific signals are indicated by the arrowheads. B. An inverted image of chromosome 2 containing DEC-205-specific signal (see Fig. 6A) is aligned with an ideogram of chromosome 2. The chromosome band corresponding to DEC-205 gene is 25 indicated to the right; Figure 7 shows that expression of DEC-205 transcripts within human hematopoetic cell lines. Total RNA prepared from the cell lines were subjected to Northern blot analysis with the 3 P]fragment 3 (A and or 3 2 P]-actin probes. The final wash was 0.1 x SSC at 65"C. The positions of the RNA molecular size standards are indicated to the right. The estimated molecular size of DEC-205 transcripts are indicated to the left. A, 24 h exposure; B, 72 h exposure; Figure 8 shows RT-PCR analysis of DEC-205 mRNA in human DC preparations.
Specific product is seen using lineage negative; fresh DC (lane 2) and a stronger signal -6with CMRF-44' low density cultured DC (lane CD8 T lymphocytes (lane 1) contain no DEC-205 mRNA Ethidium stain.
Figure 9 represents the result of an ELISA assay showing a monoclonal antibody binding specifically to DEC-205 peptide I and not peptide 3. Positive control binding of a hyperimmunized rabbit anti-DEC-205-peptide 1 serum and hyperimmunized rabbit anti- DEC-205-peptide 2 serum are shown; Figure 10 gives the DNA sequence for human DEC-205 (coding region only); Figure 11 gives the human DEC-205 amino acid sequence.
DETAILED DESCRIPTION OF THE INVENTION 15 A. Human DEC-205 The human DEC-205 of the invention is believed to be the human homolog of murine DEC-205 and has an approximate molecular weight of 198 to 205 kDa. It has the structure shown in Figures 1 and 2A. It also has the deduced amino acid sequence shown in Figure 11.
Human DEC-205 can usefully be provided in a number of different forms. These include human DEC-205 itself the "mature" form of human DEC-205, and the extracellular receptor domain of human DEC-205.
25 The "mature" form of human DEC-205 of the invention is human DEC-205 less its native amino-terminus leader or signal sequence, whereas the extracellular receptor domain is human DEC-205 lacking the signal sequence, the transmembrane region and cytoplasmic domain (where present).
The extracellular domain may be identified through commonly recognised criteria of extracellular amino acid sequences. The determination of appropriate criteria is known to those skilled in the art, and has been described, for example by Hopp et al., Proc. Natl.
Acad. Sci. USA 78, 3824-3828 (1991); Kyte et al., J. Mol. Biol. 157, 105-132 (1982); Emini, J. Virol 55, 836-839 (1985); Jameson et al. CA BIOS 4 181-186 (1988); and Karplus et al. Naturwissenschaften 72, 212-213 (1985). Amino acid domains predicted by these criteria to be surface exposed are characteristic of extracellular domains.
-7- The amino acid sequences of the predicted regions for human DEC-205 are shown in Figure 3A. These include the amino acid sequences for the signal peptide, cysteine-rich domain, fibronectin type II domain, Carbohydrate Recognition Domain-1, (CRD-1), CRD-2, CRD-3, CRD-4, CRD-5. CRD-6, CRD-7, CRD-8, CRD-9, transmembrane domain and cytoplasmic domain.
Human DEC-205 of the invention or its extracellular receptor domain (or parts thereof) may be prepared by methods known in the art. Such methods include protein synthesis from individual amino acids as described by Stuart and Young in "Solid Phase Peptide Synthesis", Second Edition, Pierce Chemical Company (1984). It is however preferred that human DEC-205 and/or its extracellular receptor domain or parts thereof be prepared by recombinant methods as will be detailed hereinafter.
1 15 Example 1 provides further details of human DEC-205.
Example 1
S
S
S
Langerhans cells were prepared from human skin. Epidermal cell suspensions were prepared from split thickness normal human breast skin by 30 min dispase (Boehringer- 20 Mannheim, Mannheim, Germany; 0.5% in PBS) treatment at 37*C, followed by 10 min disaggregation in the presence of trypsin (0.25% in PBS), DNase I (5U/ml in PBS) and EDTA at room temperature. Langerhans cells were then enriched by Ficoll/Metrizoate gradient separation (d=l.077g/cm 3 Final cell suspensions contained 3-15% Langerhans cells as determined by HLA-DR positivity. Total RNA was extracted 25 using Trizol reagent according to the manufacturer's instructions.
Degenerate primers were prepared on an Applied Biosystems DNA Synthesizer with the primer sequences and as set out below: 5'-GAX ACY GAX GGY TTX TGG AA-3' 3'-GCY GTX TTZ TCZ AAC CAC wherein X is C or T, Y is A, C, G or T, and Z is G or A.
Single stranded cDNA was prepared using total RNA and reverse transcribed by AMV reverse transcriptase using the 3' primer Subsequently, the cDNA was amplified -8using the and primer using PCT amplification according to techniques known in the art.
The amplified products were run on a 2% agarose gel and visualized with ethidium bromide staining.
The DNA was purified and ligated into the T tailed pGEM vector (available from Promega) using standard techniques. The ligation mixture was transformed into competent E. coli JM 109 bacteria (available from Promega) which were grown on agar plates with appropriate antibiotic selection. Two colonies were isolated. DNA was prepared and digested with restriction enzymes. Two inserts of the same size as the PCR product were sequenced by double-stranded DNA sequencing techniques using a Sequence Kit (Sequence 2.0 USB Lab Supply, Pierce). The two clones corresponded to human DEC- 205.
The amino acid sequence of human DEC-205 was determined to include the following amino acid sequences:
TVDCNDNQPGAICYYSGNETEKEVKPVDSVKCPSPVLNTPWIPFQNC
CYNFIITKNRHMATTQDEVHTKCQKLNPKSHILSIRDEKENNFVLEQLLYFNYMA
SWVMLGITYRNNSL; and (ii) SQHRLFHLHSQKCLGLDITKSVNELRMFSCDSSAML.
Determination of these sequences was fundamental to isolating the cDNA for human DEC- 205 detailed below.
-9- In the partial sequences given above, individual amino acids are represented by the single letter code as follows: Three-letter One-letter Amino Acid abbreviation symbol Alanine Ala
A
Arginine Arg
R
Asparagine Asn
N
Aspartic acid Asp
D
Asparagine or aspartic acid Asx
B
Cysteine Cys
C
Glutamine Gin
Q
Glutamic Acid Glu
E
Glutamine or glutamic acid Glx
Z
Glycine Gly
G
Histidine His
H
Isoleucine Ile
I
20 Leucine Leu
L
Lysine Lys
K
Methionine Met
M
Phenylalanine Phe
F
S: Proline Pro p Serine Ser
S
Threonine Thr
T
Tryptophan Trp
W
Tyrosine Tyr y Valine Val
V
30 Unidentified
X
This code also applies to the predicted full sequence of Figure 11, deduced from the DNA encoding human DEC-205 isolated as described below.
B. Polvnucleotides Encoding Human DEC-205 In another aspect of this invention, the applicants provide polynucleotides encoding human DEC-205 or its extracellular domain. These polynucleotides may be DNA (isolated from nature, synthesised or cDNA) or RNA. Most often, the polynucleotides will be DNA.
The polynucleotides of the invention specifically include those which include the nucleotides A ACA GTT GAT TGC AAT GAC TAC TAT TCA GGA AAT GAG ACT GAC AGT GTT AAA TGT CCA TCT ATA CCA TTT CAG AAC TGT TGC AAT AGG CAT ATG GCA ACA ACA TGC CAG AAA CTG AAT CCA AAA GAT GAA AAG GAG AAT AAC TTT TTC AAT TAT ATG GCT TCA TGG AGA AAT AAX TCT CTT; and AAT CAA CCA GGT GCT ATT TGC GAA AAA GAG GTC AAA CCA GTT CCT GTT CTA AAT ACT CCG TGG TAC AAT TTC ATA ATA ACA AAG CAG GAT GAA GTT CAT ACT AAA TCA CAT ATT CTG AGT ATT CGA GTT CTT GAG CAA CTG CTG TAC GTC ATG TTA GGA ATA ACT TAT 0* a
S.
0@ 0 a @0 (iv) GAG GAC AAG TTA TGG AAG TGG GTG TCC CAG CAT CGG CTC TTT CAT TTG CAC TCC CAA AAG TGC CTT GGC CTC GAT ATT ACC AAA TCG GTA AAT GAG CTG AGA ATG TTC AGC TGT GAC TCC AGT GCC ATG CTG TGG TGG AAA TGC GAG CAC CA wherein X is T or G, as well as the full nucleotide sequence shown in Figure but are not limited thereto.
The invention also includes within its scope functional equivalents of these polynucleotides.
This aspect of the invention will now be illustrated by the following Examples.
EXAMPLE 2 EXPERIMENTAL
PROCEDURES
Cell culture The cell lines, HEL, K562, KG-1, THP-1, U937, Mann and Jurkat were obtained from the American Type Culture Collection (Rockville, MD). L28 cells were provided by V. Diehl (Klinik for Innere Medizin, Cologne, Germany). HDLM2 and KMH2 cells were obtained from the German Collection of Micro-organisms and Cell
IOA-
Culture (Braun scfwei g, Germany). Mono Mac 6 cells (Bufler et al (1995) Eur. j. ImmunoL 604-610) were provided by H. Engelmann (Institute of Immunology, Munchen, Germany). All cell lines were maintained in RPVI 1640, 10% fetal calf 0:* -11 serum, 100 U/ml penicillin, 100 ug/ml streptomycin except that HDLM2 cells were with fetal calf serum.
Isolation of leukocytes-Leukocyte populations were isolated using standard laboratory procedures.
Isolation of cDNA encoding for human DEC-205-A set of degenerate oligonucleotide primers were designed based on the published amino acid sequence of mouse DEC-205 (Jiang et al (1995), above) and synthesized in house or by Life Technologies (Auckland, New Zealand) (see Fig. 2A). These primers were: DEC-a (5'-AAYATGCTNTGGAARTGGGT-3'), DEC-b (5'-TGRTGYTCRCAYTTCCACCA-3'), DEC-d (5'-GAYACNGAYGGNTTYTGGAA-3') and DEC-e (5'-GCNGTYTTRTCRAACCACAT-3'), 15 where Y=C or T, R= A or G, N=A or C or G or T. Total RNA isolated from L428 or HEL cells was reverse transcribed with avian myeloblastosis virus reverse transcriptase (Promega, Madison, WI) at 55C for 1 h using the primers DEC-b or DEC-e. PCR was performed using the resultant cDNA and Taq polymerase (Boehringer Mannheim, Auckland, New Zealand) with the primers DEC-a/-b for DEC-b-primed or DEC-d/-e for DEC-e-primed cDNAs. The PCR conditions used were the initial denaturation at 94 0
C
for 5 min, 35 cycles of denaturation at 94°C for 1 min, annealing at 54*C for 1 min, e*e:e extension at 72 0 C for 1 min, and the final extension at 72*C for 5 min. The PCR reactions were fractionated with 2% agarose gel in 40 mM Tris-acetate, pH 8.3, 1 mM EDTA (TAE) buffer, and stained with 0.5 ug/ml ethidium bromide. The PCR fragments 25 (fragment 1 and 2, see Fig. 2A and 2B) were cloned into pGEM-T vector (Promega), and sequenced manually using Sequenase DNA sequencing kit (Amersham Life Science, Auckland, New Zealand).
A set of oligonucleotide primers nested within the DNA sequence of fragment 1 and 2 were synthesized (see Fig. 2A). These primers were: 0 2 3 (5'-GCTCTAGAAACATGACCCATGAAGCC-3' containing a XbaI site), 028 (5'-GCTCTAGACATCGGCTCTTTCATTTGT-3' containing a Xbal site) and 0 2 9 (5'-CGGGATTCACAGTTGATTGCAATGACA-3' containing a EcoRI site) where incorporated restriction sites are underlined. Two ug of poly(A) RNA from L428 cells was reverse transcribed with 200 U of SuperScriptlI (LifeTechnoloies) at 45"C for 1 h using an oligo d(T) adaptor primer 12- 018 (5'-GACTAGTCTGCAGAATTCTTTTTTT TTTTTTTTT3' containing a Spel, Pstl, and EcoRI sites). After heat-inactivation at 70"C for 15 min, the reaction was incubated with 1 U RNaseH (Life Technologies) at 37"C for 30 min, heatinactivated at 70"C for 15 min, and diluted to 1 ml with 10 mM Tris-HCI, pH 8.0,1 mM EDTA (L428 cDNA pool). In order to isolate the fragment 3 (connecting the fragment 1 and 2) (see Fig. 2A), PCR was performed with 5 ul of L428 cDNA pool, the primers 028 and 023, and 2.5 U of Expand enzyme mix (Boehringer Mannheim). The PCR conditions were the initial denaturation at 94*C for 2 min, 10 cycles of 10 cycles of denaturation at 94C for 15 sec, annealing at 53 *C for 30 sec, and extension at 68°C for 4 min, followed by 20 cycles of denaturation at 94*C for 15 sec, annealing at 53 0 C for sec, and extension at 68°C for 4 min plus additional 20 sec for each cycle, and the final extension at 68 0 C for 15 min. 3'-rapid amplification of cDNA ends (3'-RACE) (Frohman et al (1988) Proc. Natl. Acad Sci. USA 85, 8998-9002) was performed in order to isolate the fragment 4 (connecting the fragment 1 and the 3'-untranslated region of DEC-205) (see Fig. 2A). PCR was performed with 5 ul of L428 cDNA pool and the primer 029 and an adaptor primer 019 (5'-GACTAGTCTGCAGAATTC, containing a Spel, PstI and EcoRI site), in the same conditions for the fragment 3. The PCR reactions were fractionated with 0.8% agarose gel in TAE buffer, and stained with ethidium bromide. Both the fragment 3 and 4 were restriction digested with XbaI and EcoRI, respectively, and cloned into pBluescript II (Stratagene, La Jolla, CA). The representative clones from the fragment 3 (pB38fl) and 4 (pb30-3) were sequenced with a LI-COR automated sequencer (LI-COR, Lincoln, Nebraska) using SequiTherm cycle sequecing kit (Epicentre Technologies, Madison, WI). If required, these plasmids were subjected to exonucleasem-nested deletion using Erase-A-Base system (Promega), and 25 used for sequencing.
An oligo dT-primed L428 cDNA library was prepared using ZAP Express cDNA Gigapack Cloning kit (Stratagene) according to manufacturer's instruction. The fragment 3 was labeled with [a-32P]dCTP (NEN) using Multiprime system (Amersham Life Science). The library was screened by plaque hybridization with the 2 P]fragment 3 using standard techniques (Sambrook, Fritsch, and Maniatis, T. (1989) Molecular Cloning: A Laboratory Manual, 2Ed., Cold Spring Harbour Laboratory, New York, USA). The specific activity of the probe was 0.8 xl0 9 cpm/ug DNA and used at 1 x 106 cpm/ml. The final wash was in 0.1 x SSC, 0.5% SDS at 65C (1 x SSC is 0.15 M NaC1, 15 mMM Na-citrate, pH7.0). Positive clones were converted to phagemid pBK-CMV (Stratagene) and sequenced using an automated sequencer.
-13- In order to verify the DNA sequence obtained from the PCR clones, pB3 8f for fragment 3 and pB30-3 for fragment 4, the fragment 5 was PCR-amplified from a L428 cDNA pool using primers 058 (5'-CGGGATCCCTCTGGCCGCGCACTAATGA-3' containing a BamHI site) and 050 (5'-CCGCTCGAGCTGTGGATACCAGCACATGCCT-3' containing a XhoI site) (see Fig. 2A). The PCR conditions were identical to that for the fragment 3 except using longer extension period (6 min) for cycling. The fragment 5 was sequenced directly using the IRD, 4 -labeled custom primers (MWG-Biotech, Ebersberg, Germany) and a LI-COR automated sequencer without cloning. These primers were: IRD001 (5'-GATGGGAACTCTTATGGGAGACCT-3' at nucleotide 523-555), IRD002 (5'-TGATGCAGGCTGGCTGCCAAATAA-3' at nucleotide 1134-1157), IRD003 (5'-AACTGGGCAACTGTTGGTGGAAGA-3' at nucleotide 1759-1782), IRD004 (5'-ATGGCGAAGAGGCTGGCATTTCTA-3' at nucleotide 2334-2357), IRD005 (5'-CTCAAGCAAGCGATACCTGTCACT-3' at nucleotide 2972-2995), IRD006 (5'-TGGGCAACTCGAAGACTGTGTAGT-3' at nucleotide 3624-3647), IRD007 (5'-CACCAGCACAGCATTCTTGCTTGT-3' at nucleotide 4168-4191) and IRD008 (5'-ATTTGTGAGCAGACTGATGAGGGA-3' at nucleotide 4797-4820).
The sequences of these primers were based on those of pb38fl and pb30-3, and they were positioned as 540-650 bp apart, ensuring the generation of contigs overlapping by at least 100 bp after automated sequencing.
Southern blot analysis-Genomic DNA was prepared from peripheral blood of patients with hematological disorders (each patient was karyotyped at Canterbury Health Laboratories, Christchurch, New Zealand). Approximately 8 ug of genomic DNA was digested with BglII, BamHI, EcoRI, or HindI, fractionated in 0.8% agarose gel in 89 25 mM Tris-borate, pH 8.3, 2 mM EDTA, and transfered to Hybond N+ by capillary reaction. A PCR-fragment corresponding to the cyteine-rich domain was PCR-amplified from pBK14-1 using the primers 058 and 059 CGGAATTCGATCTCATGATAAGGCTGGTCACA-3' containing a EcoRI site) (see Fig. 2A). Briefly, PCR was performed with 2 ng of pBK14-1, the primer 058 and 059, and Taq polymerase. The PCR conditions used were the initial denaturation at 94C for 2 min, 30 cycles of denaturation at 94*C for 15 sec, annealing at 55°C for 15 sec, extension at 72 0 C for 30 sec, and the final extension at 72*C for 5 min. The 450 bp PCR product was labeled with [a- 3 2 P]dCTP using Multiprime labeling system (Amersham Life Science). The blot was hybridized with the probe using standard technique (Sambrook et al, (1989), above). The specific activity of the probe was 0.8 x 109 cpm/ug -14- DNA and used at 1 x 106 cpm/ml. The final wash was in 0.3 x SSC, 0.5% SDS at and exposed to X-OMAT AR film (Kodak) with an intensifying screen at -70 0
C.
A blot containing PstI-digested genomic DNA from a human-rodent somatic hybrid cell panel was obtained from Oncor (Gaithersburg, MD), and probed with the 32 P]cysteinerich domain fragment as described above.
Fluorescent in situ hybridization-Metaphase spreads were prepared from phytohaemagluttinin-stimulated peripheral blood lymphocytes of a 46,XY male donor using standard cytogenetic procedures. The fragment 6 was amplified by recombinant PCR with the fragment 3 and 4 (see Fig. 2A). PCR was performed with each of the fragment 3 and 4 and the primers 028 and 019 in the same conditions for the fragment 3 except using longer extension period (7 min) for cycling. The fragment 6 was labelled with biotin-14-dCTP using a BioPrime random prime labelling kit (Bethesda Research Laboratories, Gathersberg, Maryland), and hybridized to metaphase cells on slides S:.Conditions for hybridization and immunofluorescent detection were essentially as described (Morris et al, (1993) Human Genetics, 91, 31-36), except that Cot 1 suppression was not required, slides were washed to a stringency of 0. IxSSC, 60*C after S. hybridization, and an additional amplification step was needed because of the small size of the probe. For precise chromosome band localization, DAPI and FITC images were captured separately for each metaphase from the fluorescent microscope to computer using a Photometrics KAF1400 CCD camera and IPLAB Spectrum software (Signal Analytics, VA), and colour-joined using Multiprobe extension software.
25 Northern blot analysis--Approximately 10 ug of total RNA from cultured cells were fractionated in formaldehyde-denatured 1% agarose gel and transferred to Hybond N+ (Amersham) using 3 M NaC1, 8 mM NaOH, 2 mM sarkosyl with Turboblotter (Schleicher Schuell, Keene, NH) for 3 h. The membrane was UV-crosslinked (Stratalinker, Stratagene), and hybridized with ["P]fragment 3 or 2 P]human §-actin probe using standard techniques (Sambrook et al (1989), above). The specific activity of the probes were 0.9-1.1 x 10 9 cpm/ug DNA and used at 0.7-1.1 x 10 cpm/ml. The final wash was in 0.1 x SSC, 0.5% SDS at 68*C, and exposed to X-OMAT AR film (Kodak) with intensifying screen at Reverse transcription-PCR analysis--Total RNA from isolated leukocytes was incubated with RNase-free DNaseI (Life Technologies), and was reverse transcribed using SuperscriptlI with the oligo dT adaptor primer 018. PCR was performed using a pair of DEC-205 specific primers 06 0 (GTGGATCCAGTACAAGGGTCA at nucleotide 4655-4686) and 056 (ACCAAATCAGTCCGCCCATGA at nucleotide 5116-5096) with Taq polymerase in the presence of a PCR additive, Q buffer (Qiagen) by touch down PCR (Don, Cox, Wainwright, Baker, and Mattick, (1991) NucleicAcidRes. 19, 4008). PCR conditions used were the initial denaturation at 92°C for 2 min, 21 cycles of denaturation at 92*C for 15 sec, annealing at 60*C minus for 15 sec, extension at 68 0 C for 30 sec, 15 cycles of denaturation at 92 0
C,
annealing at 50°C, extention at 68 0 C for 1 min and the final extension at 68 0 C for 5 min.
Human glycelaldehyde-3-phosphate dehydrogenase (GAPDH) (Tokunaga,
K.,
Nakamura, Sakata, Fujimori, Ohkubo, Sawada, and Sakiyama,
S.
(1987) Cancer Res. 47, 5616-5619) was used for normalization. The primers for GAPDH were 053 (ATGGGGAAGGTGAAGGTCGGA-3' at nucleotide 61-81), and 055 (AGGGGCCATCCACAGTCTTCT-3' at nucleotide 634-614). The PCR reactions were S 15 fractionated with 1.5 agarose gel in TAE buffer, and stained with 0.5 ug/ml ethidium bromide.
Sequence data analysis--The National Center of Biotechnology Information (NCBI) Center electronic mail server BLAST was used to search for homologous sequences.
Sequence alignments and motif search were done using Bestfit and Motifs programs, SC'. respectively, of GCG computer package (Madison, WI).
RESULTS
25 Isolation of cDNA for human DEC-205. Based on the amino acid sequence of mouse DEC-205, a set of degenerate primers were synthesized and used to perform RT-PCR using the Hodgkin's disease-derived L428 cell line and the myeloid HEL cell lines (Fig.
The two pair of primers (DEC-d/-e, and DEC-a/-b) gave rise to the specific RT-PCR products, fragment 1 (390 bp) and 2 (150 bp), respectively (Fig. 2A and 2B). These specific fragments were cloned and sequenced (data not shown). The deduced amino acid sequences of fragment 1 and 2 were -80% identical to that of mouse DEC-205, indicating that these fragments were derived from the cDNA of human DEC-205.
Primers nested within these fragments were synthesized and further RT-PCR and 3'- RACE performed using a L428 cDNA pool reverse transcribed with an oligo dT adapter primer 018. A 3.8 kb RT-PCR product (fragment 3) was obtained using primer 028 and -16- 023 (Fig. 2A and 2C). A 3.2 kb 3'-RACE product (fragment 4) was obtained using primer 029 and an adaptor primer 019 (Fig. 2A and 2C). The fragment 3 was cloned and several identical clones were identified by restriction enzyme map analysis (data not shown), and one of which, pb38fl, was fully sequenced: The DNA sequence of the fragment 3 (pB38fl) extending from the middle of cysteine-rich domain to the middle of CRD-8 (Fig. 2A), was 82% identical to the published mouse DEC-205 cDNA sequence. The fragment 4 was cloned and two distinct clones identified by restriction enzyme map analysis. Both clones were partially sequenced and the 3' end DNA sequence of one clone (eg. pb30-3) was found to contain a poly A tail, and with 72% identical to 3'-untranslated region of mouse DEC-205 (data not shown). Therefore, the pb30-3 was sequenced to obtain the DNA sequence of the coding region of DEC-205 plus partial 3 '-untranslated region. The resulting DNA sequence for the coding region was -80% identical to that of mouse DEC-205 spanning from the middle of CRD-8 to the end ofcytoplasmic domain (Fig. 2A). The DNA sequences obtained from pb38fl :i 15 and pb30-3 overlapped by 320 bp, covering 95% of human DEC-205 coding region.
In order to complete the 5' end of the DEC-205 cDNA sequences a L428 cDNA library was screened by plaque hybridization using 32 P-labeled fragment 3 as a probe. A clone •(pBK 4-1) was isolated, and the 1.5 kb insert of this clone was sequenced (Fig. 2A).
20 The sequence was -80% identical to the mouse sequence and corresponded to the signal peptide, cysteine-rich domain, fibronectin type II domain, CRD-1 and part of the CRD-2.
The pBK14-1 contained 51 bp 5'-untranslated region, and overlapped with fragment 3 by -1.2 kb.
25 To validate the DNA sequence obtained from the PCR clones, a further RT-PCR fragment (fragment 5) amplified with primers 058 (nested in the cysteine-rich domain) and 050 (located -130 bp downstream of the stop codon) was prepared (Fig. 2A). The fragment 5 PCR product was sequenced directly using IRD,,-labeled custom primers without cloning. A total of 10 point mutations, presumably generated because of the low fidelity of thermostable polymerases were found and corrected in the PCR clone-derived DNA sequence. The complete cDNA sequence for human DEC-205 is 5166 bp in size, and encodes for a predicted 198 kDa type I transmembrane protein with 1722 amino acids before post translational modification.
The deduced amino acid sequence of human DEC-205 showed 77% overall identity with the homologous mouse protein (Fig. 3A). All the cysteines, and putative N-glycosylation -17sites in the extracellular domain of mouse DEC-205, were conserved in the human sequence. In the cytoplasmic domain the putative serine phosphorylation sites by protein kinase C or casein kinase, and a tyrosine, which appears to be important for coated pit-mediated internalization (Ezekowitz, Sastry, Bailly, and Warner, A. (1990) J. Exp. Med 172, 1785-1794; and Zvaritch, Lambeau, and Lazdunski, M. (1996) J. Biol. Chem. 271, 250-257), were also conserved. There was one amino acid deletion within the CRD-5 in human DEC-205. All the extracelluar domains, including the cysteine-rich domain, fibronectin type II domain, and were 74-87% identical between human and mouse sequences (Fig. 3B), suggesting the importance of these domains for the function of DEC-205. In contrast, the two hydrophobic domains, including the signal peptide and transmembrane domain, showed much lower identity (57% and 52%, respectively (Fig. 3B)) with the mouse protein, confirming the observation that these hydrohobic domains are more variable, and rapidly evolved structures (Von Heijne, G. (1990) J Membrane Biol. 115, 195-201).
DEC-205 is a single copy gene with polymorphism-Peripheral blood-derived genomic SDNA from 4 individuals was restriction enzyme-digested with BglII, BamHI, HindIII or EcoRI, and subjected to Southern blot analysis. The cysteine-rich domain of the macrophage mannose receptor (Kim, Ruiz, Bezouska, and Drickamer,
K.
(1992) Genomics 14, 721-727; and Harris, Peters, Eicher, Rits, M., Raspberry, Eichbaum, Super, and Ezekowitz, R.A.B. (1994) Biochem.
Biophys. Res. Con. 198, 682-692) and phospholipase A2 receptor (Ancian, Lambeau, Mattei, and Lazdunski, M. (1995) 270, 8963-8970) is encoded by one exon.
Therefore, we amplified the cysteine-rich domain of human DEC-205 using primers 058 25 and 059 as a potential single exon probe (450 bp), and used this to probe the Southern blot in high stringency. A single band appeared in BglI-, BamHI- or Hindm-digested genomic DNA from all individuals, indicating that DEC-205 is a single copy gene (Fig.
The EcoRI digests, however, produced a single band in two individuals and double bands in another, indicating that the DEC-205 gene is polymorphic. Further Southern blot analysis with larger panel of individuals showed identical results (data not shown).
Therefore, DEC-205 is a single copy gene with at least one polymorphic site.
DEC-205 gene maps to chromosome band 2q24--In order to map the human DEC-205 gene, a somatic cell hybrid panel Southern blot (PstI-digested) was probed with the 3 P]cysteine-rich domain as described above (Fig.5). A 3.0 kb band in human genomic DNA was found to hybridize strongly, and the identical band appeared in chromosome -18- 2-containing somatic human-mouse hybrid cells, indicating that DEC-205 gene localizes on chromosome 2. The probe also hybridized weakly with hamster DNA, suggesting the presence of DEC-205 homolog in hamster as well as in the mouse (which also hybridized strongly). The origin of the weakly hybridized bands with apparent polymorphism in the human DNA-containing lanes is not known. The identical band appeared in chromosome 2, and may either be related to an alternative exon structure for this region of DEC-205 or result from weak cross hybridization to another gene on chromosome 2.
Fluorescent in situ hybridization then was used to map the DEC-205 gene in detail (Fig.
6A and 6B). The 6.4 kb recombinant PCR fragment (fragment 6) (Fig. 2A) was prepared from fragment 3 and 4, labeled with biotinylated nucleotides, and used as a probe in a high stringency (Fig. 6A). Ninety-one of a combined total 114 metaphase cells analysed from three experiments showed fluorescent signals on one (27) or both (64) chromosomes 2 in the middle of the long arm, specifically in band q24 (Fig. 6B). High 15 resolution banding analysis provided a more precise location of signals (not shown). No additional site-specific signals were detected on any other chromosome.
DEC-205 exhibits multiple transcripts in cell lines--A panel of human cell lines, including myeloid, B lymphoid, T lymphoid and Hodgkin's desease-derived cell lines, 20 were analyzed for the expression of DEC-205 transcripts by Northern blot analysis with the 2 P]fragment 3 as a probe (Fig. 7A and 7B). Two DEC-205 transcripts, 7.8 and kb in size, were detected, and the 7.8 kb transcript was the most abundant. The expression level varied between cell lines, however the myeloid cell line THP-1, the B lymphoid cell line Mann and the Hodgkin's desease cell line KMH2 showed the highest 25 level of expression. Even with longer exposure, DEC-205 transcripts were not detectable in K562, KG-I, Monomac and Jurkat cells, suggesting these cells are DEC- 205 negative (Fig. 7B). Interestingly all Hodgkin's disease-derived cell lines tested express the transcripts. Semiquantitative RT-PCR studies also support these results (data not shown).
C. Recombinant Expression of Human DEC-205 In yet another aspect, the present invention relates to the recombinant expression of human DEC-205 or of its extracellular domain.
The Polynucleotides that encode human DEC-205 or the extracellular domain of the invention may be inserted into known vectors for use in standard recombinant
DNA
-19techniques. Standard recombinant DNA techniques are those such as are described in Sambrook et al.; "Molecular Cloning" 2nd Edition Cold Spring Harbour Laboratory Press (1987) and by Ausubel et al., Eds, "Current Protocols in Molecular Biology" Greene Publishing Associates and Wiley-Interscience, New York (1987).
Vectors for expressing proteins in bacteria, especially E. coli are known. Such vectors include the PATH vectors described by Dieckmann and Tzagoloff in J. Biol. Chem 260.
1513-1520 (1985). These vectors contain DNA sequences that encode anthranilate synthetase (TrpE) followed by a polylinker at the carboxy terminus. Other expression vector systems are based on beta-galactosidase (pGEX); lambda P maltose binding protein (pMAL); and gluthathione S-transferase (pGST) see Gene 67, 31 (1988) and Peptide Research 3, 167 (1990).
Vectors useful in yeast and insect cells are available and well known. A suitable 15 example of a yeast vector is the 21. plasmid.
Suitable vectors for use in mammalian cells are also known. Such vectors include wellknown derivatives of SV-40, adenovirus, retrovirus-derived DNA sequences and vectors S derived from combination of plasmids and phage DNA.
S Further eucaryotic expression vectors are known in the art P.J. Southern and P.
Berg, J. Mol. Appl. Genet. 1, 327-341 (1982); S. Subramani et al, Mol. Cell. Biol. 854-864 (1981); RJ. Kaufmann and P.A. Sharp, "Amplification And Expression of Sequences Cotransfected with a Modular Dihydrofolate Reductase Complementary
DNA
Gene," J. Mol. Biol. 159, 601-621 (1982); RJ. Kaufmann and P.A. Sharp, Mol. Cell.
Biol. 159, 601-664 (1982); S.I. Scahill et al, "Expression And Characterization Of The Product Of A Human Immune Interferon DNA Gene In Chinese Hamster Ovary Cells," Proc. Natl. Acad. Sci. USA 80, 4654-4659 (1983); G. Urlaub and L.A. Chasin, Proc.
Natl. Acad. Sci. USA 27, 4216-4220, (1980).
The expression vectors useful in the present invention contain at least one expression control sequence that is operatively linked to the DNA sequence or fragment to be expressed. The control sequence is inserted in the vector in order to control and to regulate the expression of the cloned DNA sequence. Examples of useful expression control sequences are the lac system, the tm system, the tac system, the trc system, major operator and promoter regions of phage lambda, the control region of fd coat protein, the glycolytic promoters of yeast, e.g. the promoter for 3 -phosphoglycerate kinase, the promoters of yeast acid phosphatase, e.g. Pho5, the promoters of the yeast alpha-mating factors, and promoters derived from polyoma, adenovirus, retrovirus, and simian virus, e.g. the early and late promoters or SV40, and other sequences known to control the expression of genes ofprokaryotic and eucaryotic cells and their viruses or combinations thereof.
Vectors containing the receptor-encoding DNA and control signals are inserted into a host cell for expression of the receptor. Some useful expression host cells include wellknown prokaryotic and eucaryotic cells. Some suitable prokaryotic hosts include, for example, E. coi such as E. coli SG-936, E. col HB 101, E. coli W3110, E. coli X1776, E. coli X2282, E. coli DHT, and E. coli MR01, Pseudomonas, Bacillus, such as Bacillus subtilis, and Streptomyces. Suitable eucaryotic cells include yeast and other fungi, i. nsect, animal cells, such as COS cells and CHO cells, human cells and plant cells in tissue culture.
D. Ligands The invention also includes ligands that bind to human DEC-205 of the invention.
20 The ligand will usually be an antibody or an antibody binding fragment raised against human DEC-205 or its extracellular domain, or against fragments thereof.
Such antibodies may be polyclonal but are preferably monoclonal. Monoclonal antibodies may be produced by methods known in the art. These methods include the 25 immunological method described by Kohler and Milstein in Nature 256 4497 (1975) •and Campbell in "Monoclonal Antibody Technology, the Production and Characterization of Rodent and Human Hybridomas" in Burdon et al. Eds, Laboratory Techniques in Biochemistry and Molecular Biology, Volume 13, Elsevier Science Publishers, Amsterdam (1985); as well as by the recombinant DNA method described by Huse et al. in Science 246. 1275-1281 (1989).
In yet another form, the ligand may also be a non-protein, probably carbohydrate containing, molecule that acts as a ligand when it binds to, or otherwise comes into contact with, human DEC-205.
-21 In addition, ligands may be of two functional types. The first functional type of ligand is a molecule which binds to human DEC-205 and stimulates it in performing its normal function (a "stimulant ligand"). The second functional type of ligand is a molecule which binds to human DEC-205 and inhibits or prevents it performing its normal function (an "antagonistic ligand").
Both types of ligand will find application in either therapeutic or prophylactic treatments as described below.
Example 3 describes the production of anti-DEC-205 antibodies.
EXAMPLE 3 Production of Anti-DEC-205 Antibodies A BALB/c mouse was immunized ip/sc with L428 cells and boosted SC with two 15 peptides derived from the DEC-205 cDNA sequence. DEC-205 peptide 1 ATTQDEVHTKC (aa1267-aa1277) and DEC-205-peptide 2 TEKEVKPVDSVKC (aa1227-aa1239) were synthesized by Chiron Mimotopes Pty Ltd (Clayton, Victoria, Australia). After a third immunization with the two DEC-205 peptides sc/ip/IV the mouse was sacrificed and a spleen cell suspension prepared. The spleen cells were fused 20 with the NS-1 myeloma cell line using standard techniques (Hock et al, Immunology 1994;83:573). A hybridoma was subsequently isolated, 2F5, which produced monoclonal antibody binding to the DEC-205-peptide 1 but not the DEC-205-peptide 2 or a third control DEC-205-peptide 3 (KCLGLDITKSVNELR) (aa82-aa96). This is shown by Figure 9.
E. Constructs The invention also provides constructs. The constructs will generally include antigens against which an immune response is desired but can also include other products to be delivered specifically to dendritic cells. Toxins, such as the ricin A chain are not excluded. The other component of the construct will vary, being either a ligand as described above or at least the extracellular domain of human DEC-205. Both constructs will have the potential to manipulate the immune system of the host.
In the ligand-antigen constructs, ligands which bind to human DEC-205 (usually antibodies, antibody-binding fragments or carbohydrates expressing proteins) can be coupled or otherwise associated with the antigen against which an immune response is -22desired. An example of such antigens are sugar-coated antigens such as tumourassociated antigens In use, the ligand component binds to human DEC-205 and the dendritic cell is 'primed' with the associated antigen. This 'priming' action will assist in the induction of an immediate immune response against the antigen.
The ligand-antigen construct can take any appropriate form for administration to the dendritic cells. Such forms may differ depending upon whether the therapeutic protocol involves isolation of the patients dendritic cells (so that the priming action can take place in vitro) or whether the construct is to be administered to a patient in vivo.
The construct can be directly administered to a patient for in vivo treatment. It can also be administered in a form which allows the construct to be expressed within the patient.
One example of such a form for administration to a patient in vivo is a live recombinant 15 viral vaccine. Such a vaccine includes a polynucleotide encoding the DEC-205 ligand (or a portion thereof) and the antigen. The vaccine is administered to the patient and, once within the patient, expresses the encoded ligand and antigen to bind to the patients dendritic cells (via human DEC-205).
20 A number of such live recombinant viral vaccine systems are known. An example of such a system is the Vaccinia virus system (US Patent 4603112; Brochier et al., Nature 354:520 (1991)).
Administration can be via intravenous, intramuscular, subcutaneous, topical, oral, intra 25 nasal, rectal or intracerebroventricular routes, as appropriate.
F. Applications Human DEC-205, its ligands and the constructs discussed above can be employed therapeutically or prophylactically in accordance with this invention to promote or inhibit any of the known actions of dendritic cells and/or to manipulate the immune system.
Thus, the antagonistic ligands per se have potential application inter alia blocking or inhibiting the immune response during transplantation procedures.
-23- Ligands also have application in delivering other products with which they are associated directly to dendritic cells. This can be for therapeutic purposes (where the delivered product is an immunogenic antigen) as discussed above. It can also be to target a toxin (such as the ricin A-chain specifically to dendritic cells to selectively destroy them as part of an immunosuppressive process.
G. The Use of Human DEC-205 to Detect Dendritic Cells in Cell Suspensions on Tissues and to Purify Dendritic Cells Monoclonal antibodies or other ligands binding to DEC-205 may be used to identify or isolate DC for scientific study or therapeutic application. For this application, the antibodies or ligands can be used in conjunction with conventional identification/separation systems. An example of such a system is the avidin-biotin immunoaffinity system available from Cell-Pro Inc, Washington, USA (see 15 US 5,215,927, US 5,225,353, US 5,262,334 and US 5,240,856).
This system employs directly or indirectly a biotinylated monoclonal antibody directed against a target cell and a column containing immunobilized avidin and can be readily adapted to extract activated human dendritic cells, in this case from human peripheral 20 blood, using the anti-DEC-205 antibody as follows: 1. A sample of human peripheral blood containing the human dendritic cells is mixed with biotinylated anti-DEC-205 antibody and incubated to allow formation of antibody/human DC complexes.
2. Following incubation, the mixture is introduced into a CeUPro continuous- 25 flow immunoadsorption column filled with avidin-coated beads, the strong affinity between biotin and avidin causing the biotin-coated antibodies (together with the human DC to which they have bound) to adhere to the avidin-coated beads.
3. After unwanted cells present in the mixture are washed away, captured activated human DC are removed from the column by gentle agitation and are available for use.
Variations on this theme using the anti-DEC-205 antibody as primary antibody (to bind to activated DC) and a biotinylated secondary antibody (to bind to the anti-DEC-205 antibody) can also be employed.
-24- It will be appreciated that before admixture with the anti-DEC-205 antibody in accordance with the above protocol, the human peripheral blood sample should be treated to ensure that the DC the sample contains are activated. This can easily be achieved by, for example, overnight incubation of the sample.
H. Functional Equivalents The invention includes functional equivalents of human DEC-205, extracellular domains and nucleic acid molecules described above.
Human DEC-205 and its extracellular domain are or include proteins. A protein is considered a functional equivalent of another protein for a specific function if the equivalent protein is immunologically cross-reactive with, and has the same function as, the original protein. The equivalent may, for example, be a fragment of the protein, or a substitution, addition or deletion mutant of the protein.
SFor example, it is possible to substitute amino acids in a sequence with equivalent amino acids using conventional techniques. Groups of amino acids known normally to be equivalent are: Ala(A) Ser(S) Thr(T) Pro(P) Gly(G); 20 Asn(N) Asp(D) Glu(E) Gln(Q); His(H) Arg(R) Lys(K); Met(M) Leu(L) Ile(I) Val(V); and Phe(F) Tyr(Y) Trp(W).
25 Substitutions, additions and/or deletions in human DEC-205 may be made as long as the resulting equivalent protein is immunologically cross-reactive with, and have the same function as, the native human DEC-205.
The equivalent human DEC-205 will normally have substantially the same amino acid sequence as the native human DEC-205. An amino acid sequence that is substantially the same as another sequence, but that differs from the other sequence by means of one or more substitutions, additions and/or deletions is considered to be an equivalent sequence. Preferably, less than 25%, more preferably less than 10%, and most preferably less than 5% of the number of amino acid residues in the amino acid sequence of the native human DEC-205 are substituted for, added to, or deleted from.
Equivalent nucleic acid molecules include nucleic acid sequences that encode proteins equivalent to human DEC-205 as defined above. Equivalent nucleic acid molecules also include nucleic acid sequences that, due to the degeneracy of the nucleic acid code, differ from native nucleic acid sequences in ways that do not affect the corresponding amino acid sequences.
Those persons skilled in the art will of course appreciate that the above description is provided by way of example only and that the invention is limited only by the lawful scope of the appended claims.
S Throughout this specification, unless the context requires otherwise, the word "comprise", or variations such as "comprises" or "comprising", will be understood to imply the ~inclusion of a stated element or integer or group of elements or integers but not the exclusion of any other element or integer or group of elements or integers.
*The reference to any prior art in this specification is not, and should not be taken as, an acknowledgement or any form of suggestion that that prior art forms part of the common general knowledge in Australia.
.:o.oi oooo• -26- SEQUENCE
LISTING
GENERAL
INFORMATION:
APPLICANT: DEREK NIGEL JOHN.
HART
TITLE: DENDRJTIC CELL
RECEPTOR
NUMBER OF SEQUENCES: COMPUTER READABLE
FORM:-
MEDIUM TYPE: 3.5"HD FLOPPY
DISC
COMPUTER: IBM PC COMPATIBLE OPERATING SYSTEM:
MS-DOS
SOFTWARE: WORDPEIRFECT 6.1 FOR
WINDOWS
INFORMATION FOR SEQUENCE ID NO. 1: SEQUENCE
CHARACTERISTICS:
LENGTH: 5166
NUCLEOTIDES
TYPE: NUCLEIC
ACID
TOPOLOGY:
LINEAR
MOLECULE TYPE: cDNA SEQUENCE DESCRIPTION: SEQ ID NO. 1: 1 ATGAGGACAG GCTGGGCGCA CCCCTCGCCG CCCGGCGGGG
CTCCTCATGC
51 rGCTC-rCTG GTTC1TCGAT CTCGCGGAGC CCTCTGGCCG
CGCACTAATG
101 ACCCCTTCAC CATCGTCCAT GGA4ATACGG
GCAAGTGCATCAAGCCAGTG
151 TATGGCTGGA TAGTAGCAGA CGACTGTGAT GAAACT~G
ACAAGUATG
201 GAAGTGGGTG TCCCAGCATC GGCTCTTTCATTTGCACTCC
CAAAAGTGCC
251 TTGGCCTCGA TATTACCAAA TCGGTAAArG AGCTGAGAAT
GTTCAGCTGT
301 GACTCCAGTG CCATGCTGTG GTGGAAATGT GAGCACCACT
CTCTGTACGG
351 AGCTGCCCGG TACTGGCTGG
CTCTGAAGGATGGACATGGCACAGCATCT
401 CAAATGCATC TGATGTCTGG AAGAAAGGAG GCTCAGAGGA AAGCC1rTGT 451 GACCAGCCTT ATCATGAGAT CTATACCAGA GATGGGAACT CTtATGGGAG 501 ACCTTGTGAA TTCCATTCT TAATTGATGG GACCTGGCAT
CATGAJTGCA
551 rTCTTGATGA AGATCATAGT GGGCCATGGT GTGCCACCAC CTTAAArAT 27 601 .GAATATGACC GAAAGTGGGG CATCTGCTrA AAGCCTGAAA ACGGTrTGTGA 651 AGATAATTGG GAAAAGAACG AGCAGTTTGG AAGTTGCTAC CAATrrAA-A 701 CTCAGACGGC TCTTC1TGG AAAGAAGCTT ATGTTTCATG
TCAGAATCMA
751 GGAGCTGAUr TACTGAGCAT CAACAGTGCT GCTGAATAA
CTTACCTMA
801 AGAAAAAGAA~ GGCATTGCTA AGATTTTCTG GATTGGTTrA
AATCAGCTAT
851 ACTCTGCTAG AGGCTGGGAA TGGTCAGACC ACAAACCAUT
AAACTTTCTC
901 AACTGGGATC CAGACAGGCC CAGTGCACCT ACTATAGGTG
GCTCCAGCTG
951 TGCAAGAATG GATGCTGAGT CTGGTCTGTG GCAGAGC-J-I
TCCTGTGAAG
1001 CTCAACTGCC CTATGTCTGC AGGAAACCAT TAAATAATAc AGTGGAGTrA 1051 ACAGATGTCT GGACATACTC AGATACCCGC TGTGATGCAG
GCTGGCTGCC
1101 AAATAATGGA 1TTGCTATC TGCTGGTAMA TGAAAGTAAT
TCCTGGGATA
1151 AGGCACATGC GAAATGCAAA GCCTTCAGTA GTGACCTAAT
CAGCATTCAT
1201 TCTCTAGCAG ATGTGGAGGT GGTTGTCACA I'AACTCCATA
ATGAGGATAT
1251 CAAAGAAGAA GTGTGGATAG GCCTrAAGAA CATAAACATA
CCAACTTTAT
.15 1301 T-rCAGTGGTC AGATGGrACr GAAGTTACTC TAACATATTrG
GGATGAGAA-
1351 GAGCCMATG 1TCCCTACAA TAAGACGCCC A.ACTGTGrTr
CCTACTTAGG
.1401 AGAGCTAGGT CAGTGGAAAG TCCAATCATG rGAGGAGAAA
CTAAAATATG
1451 TATGCAAGAG AAAGGGAGAA AAACTGAATG ACGCAAGTTC
TGATAAGATG
**.1501 TGTCCTCCAG ATGAGGGCTG GAAGAGACAT GGAGAAACCT
GTTACAAGAT
20 1551 TTATGAGGAT GAGGTCCCTr TTGGAACAAA CTGCIAATCTG
ACTATCACTA
*.1601 GCAGATTTGA GCAAGAATAC CTAAATGA1T TGATGAAAAA
GTATGATAAA
1651 TCTCTAAGAA ATACTTCTG GACTGGCCTG AGAGATGTAG
ATTCTTGTGG
1701 AGAGTATAAC TGGGCAACTG TTGGTGGAAG AAGGCGGGCT GTAACCrrrr 1751 CCAACTGGAA TTTCTTGAG CCAGCTTCCC CGGGCGGCTG
CGTGGCTATG
1825 TCTACTGGAA AGTCTGTG:AAGTGGGAG GTGAGGCT
GCAGAGT
1901 AAGCATCCCC TAAGCCTGAT GACCCCTGTC CTGAAGGCTG
GCAGAGTC
1951 CCCGCAAGTC MTTTTA TAAGGTATrC CATGCAGAA
GAATTGTAAG
2001 AAAGAGGAAC TGGGAAGAAG CTGAACGAUT CTGCCAAGCC CTrGGAGCAC 2051 ACCrT1-CTAG CTTCAGCCAT GTGGATGAAA TAAAGGAA1-r TCT-TCACTr 2101 TTAACGGACC AGTrCAGTGG CCAGCATTGG CTGTGGATTG GTT7GAATAA 2151 AAGGAGCCCA GA1TACMAG GATCCTGGCA ATGGAGTGAT
CGTACACCAG
2201 TGTCTACTAT TATCATGCCA AArGAG1-rrC AGCAGGATTA
TGACATCAGA
2251 GACTGTGCTG CTGTCAAGGT ATCATAGG CCATGGCGAA
GAGGCTGGCA
2301 T1TCTATGAT GATAGAGMAT TTA11TATT-r GAGGCC1--I--
GCTTGTGATA
2351 CAAAAC1-rGA ATGGGTGTGC CAAATTCCAA, AAGGCCGTAC
TCCAAAAACA
-28- 2401 CCAGACTGGT ACAATCCAGA CCGTGCTGGA A1TCATGGAC
CTCCACTTAT
2451 AATrGAAGGA AGTGAATATT GG1TTGTTGC TGATCTTCAC
CTAAACTATG
2501 AAGAAGCCGT CCTGTACTGT GCCAGCMATC ACAGCTTCT
TGCGACTATA
2551 ACATCITTTG TGGGACTAAA AGCCATCAAA AACAAAATAG
CAAATATATC
2601 TGGTGATGGA CAGAAGTGGT GGATAAGAAT TAGCGAGTGG
CCMATAGATG
2651 ATCATTTAC ATACTCACGA TATCCATGGC ACCGCT-1-CC
TGTGACATTT
2701 GGAGAGGAAT GCTTGTACAT GTCTGCCAAG ACTTGGC1TA
TCGACTTAGG
2751 TAAACCAACA GACTGTAGTA CCAAG1TGCC CTTCATCTGT
GAAAAATATA
2801 ATGTTTCTTC GTTAGAGAAA TACAGCCCAG ATTcTGCAGC
TAAAGTGCAA
2851 TGTTCTGAGC AATGGATTCC TrCAGAAT .AAGTGTrC
TAAAGATCAA
2901 ACCCGTGTCT CTCACATT CTCAAGCAAG CGATACCTGT CACrCCTATG 2951 GTGGCACCCT TCCTTCAGTG 1TGOAGcCAGA TTGAACAAGA CiTATTACA 3001 TCCTrGCTTC CGGATATGGA AGCTAC~MA TGGArGG1-1
TGCGCTGGAC
3051 TGCCTATGAA AAGATAAACA AATGGACAGA TAACAGAGAG
CTGACGTACA
3101 GTAACTrTCA CCCATrATTG GTrAGrGGGA GGCTGAGAAT ACCAGAAAAT 3151 1TflTGAGG AAGAGTCTCG CTACCACTGT GCCCTAATAC
TCAACCTCCA
3201 AAAATCACCG TTACTGGGA CGTGGMATrT TACATCCTGC AGTGAACGCC 3251 ACT17GTGTC TCTCTGTCAG AAATArrCAG AAGTTAAAAG
CAGACAGACG
3301 FTGCAGAATG CTTCAGAAAC TGTAAAGTAT CTAAATAATC
TGTACAAAAT
3351 AATCCCAAAG ACTCTGACTT GGCACAGTGC TAAAAGGGAG TGTCTGAAAA *3401 GTAACATGCA GCTGGTGAGC ACCGC TACGAGCTCT 3451 AGTGTGCAGG CGCTCC1TCA CAACTCTTCC TTATGGATCG
GACTCTTCAG
3501 TCAAGATGAT GAACTCAACT TTGG77GGTC AGATGGGAAA
CGTCTTCATT
S 25 3551 TTAGTCGCTG GGCTGAAACT AATGGGCAAC TCGAAGACTG TGTAGTA1TA 3601GACCTGTG ATTCTGAA AACAG-TTGAT ITLCAA1ACA ATCAACCAGG 3651 TGCTATGC TACTATTCAG GAAATGAGAC TGAAAAAGAG
GTCAAACCAG
3701 TTGACAGTGT TAAATGTCCA TCTCCTGTrC TAAATACTCC
GTGGATACCA
3751 TTTCAGAACT GTTGCTACAA 1TrCATAATA ACAAAGAATA
GGCATATGGC
3801 AACAACACAG GATGAAG17C ATACTAAATG CCAGAAACTG
AATCCAAAAT
3851 CACATATTCT GAGTA1TCGA GATGAAAAGG AGAATAACTT
TGTTCTTGAG
3901 CAACTGCTGT ACTTCAATTA TATGGCTTCA TGGGTCATG1-
TAGGAATAAC
i951 TTATAGAAAT AATCTCTTA TGTGGTTTGA TAAGACCCCA
CTGTCATATA
4001 CACATTGGAG AGCAGGAAGA CCAACTATAA AAAATGAGAA GTTT GGCT 4051 GGTTTAAGTA CTGACGGCTT CTGGGATATT CAAACCTrrA AAGTrATTGA 4101 AGAAGCAGUT TATT~CACC AGCACAGCAr TCTTGC1-TGT
AAAATTGAAA
29 4151 TGGTTGACTA CAAAGAAGAA CATMATACTA CACTGCCACA GTT7ATGCCA 4201 TATGAAGATG GTATTACAG TGTTA1-TCAA AAAAAGGTMA
CATGGTATGA
4251 AGCATTAAAC ATGTGTTCTC AAAGTGGAGG TCAC1TGGCA
AGCGTTCACA
4301 ACCAAAATGG CCAGCTCTTT CTGGMAGATA TTGTAAAACG
TGATGGAMT
4351 CCACTATGGG TTGGGCTCTC AAGTCATGAT GGAAGTGAAT CMAG1TrTGA 4401 ATGGTCTGAT GGTAGTACAT 1TGACTATAT CCCATGGAAA
GGCCAAACAT
445i CTCCTGGAAA TTGTGTTCTC TTGGATCCAA AAGGAACTTG
GAAACATGMA
4501 AAATGCAACr CTG77AAGGA TGGTGCTATT TGTTATAAAC
CTACAAAATC
4551 TMAAAAGCTG TCCCGTCTTA CATATCATC .AAGATGTCCA
GCAGCAAAAG
4601 AGAATGGGTC ACGGTGGATC CAGTACAAGG GTCACTGUTA CAAGTCTGAT 4651 CAGGCATTGC ACAGT1TTTC AGAGGCCAAA AAATTGTGTT
CAAAACATGA;
4701 TCACTCT1GCA ACTATCGTFI- CCAAAAAGA TGAAGATGAG AATAAA1TrG 4751 TGAGCAGACT GATGAGGGA AATAATAACA TrACCATGAG AGTTTGGCTr 4801 GGATTATCTC AACATTrCTGT TGACCAGTCT TGGAGTTGGT
TAGATGGATC
4851 AGAAGTGACA TTTGTCAAAT GGGAAAATAA AAGTAAGAGT ddtGtrffdA *4901 GATGTAGCAT GTTGATAGCT TCAAATGAAA CTTGGAAAAA
AGTTGAATGT
4951 GAACATGGTT 1TGGAAGAGT TGTCTGCAAA GTGCCTCTGG
GCCCTGATTA
5001 CACAGCAATA GCTATCATAG TTGCCACACT AAGTATCTTA G1TCTCATGG 5051 GCGGACTGAT TrGGTTCCTC TTCCAAAGGC ACCG11-rGCA
CCTGGCGGGT
5101 TTCTCATCAG TTCGATATGC ACAAGGAGTG AATGAAGATG
AGATTATGCT
:5151 TCCTrTTTc
CATGACTAA.
30 INFORMATION FOR SEQUENCE.1ID N0.2: SEQUENCE
CHARACTERISTICS:
LENGTH: 1722 AMINO
ACIDS
TYPE: AMI1NO
ACID
STRANDEDNESS:
SINGLE
TOPOLOGY:
LINEAR~
MOLECULE TYPE:
PROTEI
SEQUENCE DESCRIPTION: SEQ ID NO. '2: 1 MRTGWAHPSP PGGAPHAALL VLRSRGALWp RTNDPFTIVH GNTGKCn'Kpv 51 YGWIVADDCD ETEDKLWKWV SQHRLFHLHS QKCLGLDITK
SVNELRMFSC
101 DSSAMLWWKC EHHSLYG;AAR YWLALKDGHG TAJSNASDVW
KKGGSEESLC
151 DQPYHEIYTR DGNSYGRPCE FPFLIDGTWH HDCILDEDHS
GPWCATTLNY
201 EYDRKWGICL KPENGCEDNW EKNEQFGSCY QFNTQTALSW
KEAYVSCQNO
20 251 GADLLSINSA AELlYLKEKE GLAKIFWIGL NQLYSARGWE
WSDHKPLNFL
301 NWDPDRPSAP 11GGSSCARM DAESGLWQSF~ SCEAQLPYVC RKPLNN17VEL 351 TDVWVTYSDTR CDAGWLPNNG FCYLLVNESN SWDKAH-AKCK
AFSSDUSIH
401 SLADVEVVVT KLHNEDIKEE VWIGLKNINI PTLFQWSOGT
EVTLTYVVDEN
451 EPNVPYNKTP NCVSYLGELG OWKVQSCEEJ( LKYVCKRKGE
KLNDASSDKM
25 501 CPPDEGWKRH GETCYKIYED EVPFGTNCNL TITSRFEQEY
LNDLMKKYDK
551 SLRKYFWVTGL RDVDSCGEYN WA1VGGRRRA VTFSNWNFLE PAI AGCVAM 601 STGKSVGKWE VKDCRSFKAL SICKKMSGPL GPEEASPKPO
DPCPEGWQSF'
651 PASLSCYKVF HAERIVRKRN WEEAERFCQA LGAHLSSFSH
VDEIKE-FLHF
701 LTDOFSGQHW LWIGLNKRSP DLQGSWQWSD RTPVSTIIMP
NEFQOYDIR
751 DCAAVKVFHR PWRRGWHFYD DREFJYLRP ACDTKLEWVVC
QIPKGRTPKT
801 PDWYNPDRAG IHGPPLIIEG SEYWPVADLH LNYEEAVLYC
ASNHSFLATI
851 TSPVGLKAIK NKIANISGDG QKWWIRISEW PIDDHFTYSR YPWHRPp~j 901 GEECLYMSAK TWLIDLGKPT DCSTKLPFIC EKYNVSSLEK YSPDSAAXvQ 951 CSEQWIPFQN KCFLKIKPVS LTFSQASDTC HSYGGTLPSV LSQIEQ DFIT 1001 SLLPDMEATL WIGLRWTAYE KINKWTDNRE LTYSNFHPLL
VSGRLRIPEN
-31 1051 FFEEESRYHC ALILNLQKSP FTGTWNFTSC SERHFVSLCQ
KYSEVKSRQT
1101 LQNASETVKY LNNLYKIIPK TLTWHSAKRE CLKSNMQLVS
JTOPYQQAFL
1151 SVQALLHNSS LWIGLFSQDD ELNFJGWSDGK RLHFSRWAT
NGQLEDCWL
1201 DTDGFWKTVD CNDNQPGAIC YYSGNETEKE VKPVDSVKCP
SPVLNTPWIP
1251 FQNCCYNFII TKNRHMATTO DEVHTKCQKL NPKSHILSIR
DEKENNFVLE
1301 QLLYFNYMAS WVMLG~IYRN NSLMWFDKTP LSYTHWRAGR
PTIKNEKFLA
1351 GLSTDGFWDI QTFKVJEEAV YFHQHSILAC KIEMVDYKEE
HNTTLPQFMP.
1401 YEDGIYSVIQ KKV1WY'EALN MCSQSGGHLA SVHNONGQLF
LEDIVKRDGF
1451 PLWVVGLSSHD GSESSFEWSD GSTFDYIPWK GQTSPGNCVL
LDPKGTWKHE
1601 KCNSVKDGAI CYKPT KSKKL SRLTYSSRCP AAKENGSRWI
OYKGHCYKSD
1551 QALH-SFSEAK KLCSKHDHSA TIVSIKDEDE NKFVSRLMRE
NN-NITMRVWL
1601 GLSQHSVDQS WSWLDGSEVT FVKWENKSKS GVGRCSMUA
SNETWKKVEC
1651 EHGFGRVVCK VPLGPDYTAI AJIVATLSIL VLMGGLIWFL
FQRHRLHLAG
1701 FSSVRYAQGV NEDEIMLPSF
HD'
a a a. a a a

Claims (16)

1. NO:2. An isolated polypeptide comprising the amino acid sequence set forth in SEQ ID
2. An isolated polypeptide consisting essentially of the amino acid sequence set forth in SEQ ID NO:2. a a 0 a a *eoo *ee
3. 10 NO:2.
4. NO:2. An isolated polypeptide comprising amino acids 27 to 1722 as set forth in SEQ ID An isolated polypeptide comprising amino acids 27 to 1661 as set forth in SEQ ID 15 5. An isolated polypeptide comprising amino acids 1208 to 1323 as set forth in SEQ ID NO:2.
6. An isolated polynucleotide encoding a polypeptide according to any one of claims 1 to
7. An isolated polynucleotide according to claim 6 which comprises the nucleotide sequence set forth in SEQ ID NO:1.
8. An isolated polynucleotide according to claim 6 which comprises nucleotides 64 to 5166 as set forth in SEQ ID NO:1.
9. An isolated polynucleotide according to any one of claims 6 to 8 which is DNA. A vector which includes a polynucleotide according to claim 9. Q:XOp.\Vp\D.bobrDv 2001\2478039 -mdd W-lins (o div H0.345.doc-I I Dnb 2001 -33-
11. A method of producing a polypeptide according to any one of claims 1 to comprising the steps of: culturing a host cell which has been transformed or transfected with a vector according to claim 10; and recovering the expressed polypeptide.
12. A ligand that binds specifically to a polypeptide according to any one of claims 1 to 3. 10 13. A ligand that binds specifically to a polypeptide according to claim 4 or claim
14. A ligand as claimed in claim 12 or claim 13 which is an antibody, or antibody binding fragment. 15 15. A construct for use in prophylaxis or therapy comprising a ligand as claimed in any C one of claims 12 to 14 coupled to an antigen capable of inducing protective immune response in a patient. *0*Cee
16. A construct for use in prophylaxis or therapy comprising a ligand as claimed in any *oooo• 20 one of claims 12 to 14 coupled to a toxin.
17. A construct for use in prophylaxis or therapy comprising a polypeptide according to any one of claims 1 to 5 coupled to an antigen capable of inducing a protective immune response in a patient.
18. A construct for use in prophylaxis or therapy comprising a polypeptide according to any one of claims 1 to 5 coupled to a toxin.
19. A method of prophylaxis or therapy which comprises administering to a patient in need of the same a polypeptide according to any one of claims 1 to 5, a ligand according to any one of claims 12 to 14, or a construct according to any one of claims 15 to 18. QAOp\Vpa\D bcr\Dcc 200I\2478039 mded claims f div Hw.345.do-lI Dec-h 0 2001 -34- A process for isolating activated dendritic cells expressing a polypeptide according to any one of claims 1 to 5 on the surface thereof, comprising the step of contacting a sample containing said cells with a ligand as claimed in claim 14, and isolating those cells to which the ligand has bound.
21. An isolated polypeptide according to any one of claims 1 to 5, or an isolated polynucleotide according to any one of claims 6 to 9, or a vector according to claim 10, or a method according to claim 11, or a ligand according to any one of claims 12 to 14, or a 10 construct according to any one of claims 15 to 18, or a method according to claim 19, or a process according to claim 20 substantially as hereinbefore described with reference to the S Figures and/or Examples. 15 DATED this 11th day of December, 2001 DEREK NIGEL JOHN HART By his Patent Attorneys SDAVIES COLLISON CAVE DAVIES COLLISON CAVE
AU97173/01A 1996-05-29 2001-12-11 Dendritic cell receptor Abandoned AU9717301A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU97173/01A AU9717301A (en) 1996-05-29 2001-12-11 Dendritic cell receptor

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
NZ286692 1996-05-19
AU97173/01A AU9717301A (en) 1996-05-29 2001-12-11 Dendritic cell receptor

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
AU29176/97A Division AU738557C (en) 1996-05-29 1997-05-29 Dendritic cell receptor

Publications (1)

Publication Number Publication Date
AU9717301A true AU9717301A (en) 2002-02-07

Family

ID=3764407

Family Applications (1)

Application Number Title Priority Date Filing Date
AU97173/01A Abandoned AU9717301A (en) 1996-05-29 2001-12-11 Dendritic cell receptor

Country Status (1)

Country Link
AU (1) AU9717301A (en)

Similar Documents

Publication Publication Date Title
US8618262B2 (en) Monoclonal antibodies against dendritic cell receptor
JP3703834B2 (en) Activated CD4 (upper +) ligand for receptor on the surface of T cell (ACT-4-L)
ES2281072T3 (en) SURFACE RECEIVER OF ACTIVATED T CELLS: ACTS-4.
US20070065875A1 (en) NTB-A, a surface molecule involved in natural killer cells activity
US6171803B1 (en) Isolation, characterization, and use of the human β subunit of the high affinity receptor for immunoglobulin E
US6280973B1 (en) Mammalian methadone-specific opioid receptor gene and uses
US20020123617A1 (en) Novel immunoglobulin superfamily members of APEX-1, APEX-2 and APEX-3 and uses thereof
AU2002325333A1 (en) NTB-A, a surface molecule involved in natural killer cells activity
AU9717301A (en) Dendritic cell receptor
JP2004000193A (en) Pigr-2, member of immunoglobulin gene super-family
CA2288307C (en) Novel triggering receptor involved in natural cytotoxicity mediated by human natural killer cells and antibodies that identify the same
JPH07505051A (en) AAMP-1, a protein with local homology to HIV-1 ENV and NEF proteins
JPH11514848A (en) Melanoma-related protein
JP2004529638A (en) Novel polypeptide
JPH11514968A (en) Nucleotide and peptide sequences for the treatment of myasthenia

Legal Events

Date Code Title Description
MK5 Application lapsed section 142(2)(e) - patent request and compl. specification not accepted