Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
ISOLATED S. MANSONI NUCLEIC ACID MOLECULES AND USES THEREOF
Document Type and Number:
WIPO Patent Application WO/2005/023979
Kind Code:
A2
Abstract:
The invention relates to S. Mansoni proteins, and the nucleic acid molecules which encode them. Various uses are described, including immunoprophylactic, diagnostic and therapeutic methods.

Inventors:
VERJOVSKI-ALMEIDA SERGIO (BR)
LEITE LUCIANA C C (BR)
FARIAS LEONARDO P (BR)
MIYASATO PATRICIA A (BR)
KAWANO TOSHIE (BR)
DEMARCO RICARDO (BR)
GARCIA JULIO CESAR LEVANO (BR)
MARTINS ELIZABETH A L (BR)
HO PAULO L (BR)
NASCIMENTO ANA L T O (BR)
DIAS-NETO EMMANUEL (BR)
SETUBAL JOAO C (BR)
MENCK CARLOS F M (BR)
MADEIRA ALDA M B N (BR)
RODRIGUES VANDERLEI (BR)
GARGIONI CYBELE (BR)
Application Number:
PCT/BR2004/000170
Publication Date:
March 17, 2005
Filing Date:
September 10, 2004
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
FUNDACAO DE AMPARO A PESQUISA (BR)
VERJOVSKI-ALMEIDA SERGIO (BR)
LEITE LUCIANA C C (BR)
FARIAS LEONARDO P (BR)
MIYASATO PATRICIA A (BR)
KAWANO TOSHIE (BR)
DEMARCO RICARDO (BR)
GARCIA JULIO CESAR LEVANO (BR)
MARTINS ELIZABETH A L (BR)
HO PAULO L (BR)
NASCIMENTO ANA L T O (BR)
DIAS-NETO EMMANUEL (BR)
SETUBAL JOAO C (BR)
MENCK CARLOS F M (BR)
MADEIRA ALDA M B N (BR)
RODRIGUES VANDERLEI (BR)
GARGIONI CYBELE (BR)
International Classes:
C07H21/04; C07K14/435; C12N15/00; C12P21/06; C12N; (IPC1-7): C12N/
Other References:
VALADAO ET AL.: 'Y-box binding protein from Schistosoma mansoni: interaction with DNA and RNA' MOLECULAR AND BIOLOGICAL PARASITOLOGY vol. 125, no. 1-2, November 2002 - December 2002, pages 47 - 57
Attorney, Agent or Firm:
Advocacia, Pietro Ariboni (1909 - 7o Andar Brooklin Novo, -004 São Paulo - SP, BR)
Download PDF:
Claims:
WE CLAIM :
1. An isolated nucleic acid molecule which encodes an S. mansoni protein, or a portion thereof which is at least 20 amino acids in length.
2. The isolated nucleic acid molecule of claim 1, consisting of a nucleotide sequence set forth in the sequence listing.
3. Expression vector comprising the isolated nucleic acid molecule of claim 1, operably linked to a promoter.
4. Recombinant cell, transformed or transfected with the isolated nucleic acid molecule of claim 1.
5. Recombinant cell, transformed or transfected with the expression vector of claim 1.
6. Immunogenic composition comprising the isolated nucleic acid molecule of claim 1, in combination with a pharmaceutical acceptable adjuvant or carrier.
7. An isolated S. mansoni protein consisting of an amino acid sequence encoded by the isolated, nucleic acid molecule of claim 1.
8. Immunogenic composition comprising the isolated S. mansoni protein of claim 7, and a pharmaceutical acceptable adjuvant or carrier.
9. An isolated antibody which specifically binds to the isolated S. mansoni protein of claim 7.
10. The isolated antibody of claim 9, wherein said antibody is a monoclonal antibody.
11. Hybridoma cell line which produces the isolated antibody of claim 10.
12. A method for determining an S. mansoni infection in a subject comprising admixing a sample taken from said subject with the isolated protein of claim 7 and determining binding of antibodies to said protein as a determination of an S. mansoni infection.
13. The method of claim 12, wherein said protein is bound to a solid phase.
14. The method of claim 12, further comprising contacting said sample with a labeled antibody which binds to antibodies bound to said protein.
15. A method for determining an S. mansoni infection in a subject, comprising admixing a sample taken from said subject with the isolated antibody of claim 9, and determining binding of protein in said sample to said antibody as a determination of an S. mansoni infection.
16. The method of claim 15, wherein said antibody is bound to a solid phase.
17. The method of claim 15, further comprising contacting said sample with a labeled antibody which binds to protein bound to said isolated antibody.
18. Computer readable medium having recorded thereon a nucleic acid molecule from S. mansoni genome.
19. The computer readable medium of claim 1, consisting of a floppy disc, a hard disc, random access memory, read only memory, or CDROM.
Description:
ISOLATED S. MANSONI NUCLEIC ACID MOLECULES AND USES THEREOF RELATED APPLICATION [0001] This application is a continuation-in-part of application Serial Number 60/502,277 filed September 11,2003, incorporated by reference in its entirety.

FIELD OF THE INVENTION [0002] The invention relates to the isolation and sequencing of genes associated with Schistosoma, Schistosoma mansoni in particular, as well as proteins antibodies and uses thereof.

BACKGROUND AND PRIOR ART [0003] Schistosoma mansoni is the major causative agent of schistosomiasis, which affects approximately 200 million people in at least 74 countries. It is the most widespread species of trematode parasite in the Middle East, Africa, the Caribbean, and South America. See WHO: TDR Strategic Direction For Research: Schistosomiasis (World Health Organization, Geneva, 2002).

[0004] The parasite lays eggs in the hepatic portal vasculature of hosts, and this is the principal cause of morbidity caused thereby. Ensuing pathology can be fatal. See King, Initiation and Regulation of Disease in Schistosimiasis, 213-264 (Imperial College Press, London, 2001). It is estimated that 600 million people live in at risk areas.

Berquist, et al., Actu. Trop. , 82: 183-192 (2002).

[0005] While the disease has been treated chemotherapeutically, such as with Praziquantel, this can lead to the emergence of drug resistant parasites. Also, Praziquantel has little effect on re-infected individuals. See Doenhoff, et al., Trans. R.

Soc. Trop. Med. Hyg., 96: 465-469 (2002). This highlights the need to identify new drug targets, and to develop additional chemotherapeutic agents. See Berquist, et al., Actu.

Trop. , 82: 183-192 (2002).

[0006] The parasite has very effective immune evasion strategies, which has made it difficult to characterize protective immune mechanisms in hosts, such as humans.

See Berquist, et al., supra ; Dunne, et al., Resistance to Infection in Humans and Animal Models (imperial College Press, London, 2001). There has been some success in vaccination of rodents and primates using attenuated larvae (Coulson, Adv. Parasital, 39: 271-336 (1997) ), demonstrating the feasibility of effective vaccination. Nonetheless, there is little sequence database information available, as a source for vaccine candidates.

[0007] Schistosomes are platyhelminths, which are the lowest group of bilateria to diverge from metazoan lineage. See Haurdosf, Syst Biol., 49: 130-142 (2000). These organisms have a blind ending gut, and no body cavity. Notwithstanding this apparently simple body plan, the organisms have tissues, which correspond to the major organ systems of higher animals. They have a complex life cycle, exhibit sexual dimorphism, and heteromorphic sex chromosomes. They have an intimate association with gastropod mollusc intermediates, such as snails, as well as the final mammalian host. It has been suggested that they may rely on host signals for development. Active transmission between hosts and internal migrations demonstrate that they have capacity for sophisticated neuromuscular coordination.

[0008] The limited availability of sequence information regarding the S. mansoni genome has been referred to, supra. The large size of the genome (270M bases, as per Simpson, et al., Mol. Biochem. Parasital, 6: 125-137 (1982) ) as well as its complexity, has deterred full scale sequencing. Prior to the present invention, knowledge was limited to 163 full-length cDNAs, and about 16,000 ESTs, of which about 75% derive from adult worms. See Santos, et al., Mol. Biochem. Parasitol, 103: 79-97 (1999); and Williams, et al., Parasitology, 118 Suppl : S19-38 (1999).

[0009] There has been interest in developing prophylactic and therapeutic, anti- schistosomiasis vaccines, because it has been shown, by Coulson, Adv. Parasitol., 39: 271-336 (1997), that animals that have been immunized with irradiated cercaria have shown up to 80% protection, when subsequently challenged with an elevated parasite dose.

[0010] In the case of schistosomiasis, over 100 antigens have been identified, and tested. (See Berquist, et al., Acta. Trop. 82 (2): 183-192 (2002) ). Of these, about a dozen showed levels of protection in between were 35-50%. Clearly, this is insufficient.

Current thinking is that a combination of antigens may be necessary in a truly effective vaccine.

[0011] An ongoing project, the Schistosoma mansoni EST genome project, head quartered in Sao Paulo, Brazil, has generated 163,000 open reading frame expressed sequence tags ("ORESTES") from six stages of the parasite, which resulted in 31,000 assembled genes and gene fragments, which it is estimated, corresponds to 92% of the assumed 14,000 parasite genes. This has increased the available number of ESTs by 10 fold, and the number of complete genes from 156 to 522. See Verjovski-Almeida, et al., Nat. Genet, 35 (2): 148-157 (2003), incorporated by reference in its entirety.

[0012] Following the generation of this information automatic annotation within the Gene Oncology system led to an ability to attribute biological functions to various genes within the parasite, and the characterization of function and vaccine potential. In general, genes and/or proteins were selected which corresponded to potential roles in the survival of the parasite, based upon attributed function. Of particular interest were genes and proteins which interact with the host during the various parasite life cycle stages.

Exemplary were proteins recognized as toxins, as surface receptors for cellular adhesion, surface proteins, enzymes, and host factor receptors. A complete listing of these nucleic acid molecules is presented in the accompanying sequence listing.

[0013] The disclosure which follows presents details on six nucleic acid molecules which, when expressed in vivo, produced immunogenic molecules which generated a protective response.

[0014] Details are set forth in the disclosure which follows.

DETAILED DESCRIPTION OF PREFERRED EMBODIMENTS EXAMPLE 1 [0015] BH and PR isolates of S. mansoni were maintained in the laboratory by routine passage through mice and snails. Parasite life cycle stages were recovered via various ways. Specifically, via portal perfusion of mice, 7-9 weeks post infection. Eggs were extracted from mouse liver, in accordance with Dalton, et al., Parasitology, 115 (PE1) : 29-32 (1997). In order to secure miracidia, some eggs were allowed to hatch.

Germ balls from daughter sporocysts were obtained via careful dissection of the hepatopancreas 21 days after snails were exposed to miracidia. Cercaria were obtained by exposing snails to light. Following in vitro transformation of cercaria by removal of trails, to form schistosomula, these were cultured, for 7 days, and then recovered, in accordance with Basch, J. Parasitol, 67: 179-185 (1981). Cercaria, schistosomula, and adults were concentrated via centrifugation, and stored at-20°C in buffer, before extracting mRNA. Freshly isolated parasites from eggs, miracidia, and germ balls were not processed prior to mRNA extraction.

[0016] In order to extract mRNA, MACs mRNA isolation kits, which are commercially available, were used, to obtain DNA-ase treated mRNA.

[0017] This mRNA was then used to construct both cDNA and SAGE libraries.

The cDNA synthesis and amplification were performed in accordance with Dias-Neto, et al., Proc. Nafl. Acad. Sci. USA, 97: 3491-3496 (2000), and Fietto, et al., Biotechniques, 32: 1404-1408,1410-1411 (2002), incorporated by reference, with modifications. In brief, about 4: g of mRNA obtained with the MACs kit from each of the six stages of the parasite's life were eluted, in 200: 1 of DEPC treated water, and then treated with RNAse free DNAse (1 U/10 : 1), for 30 minutes, at 37°C. The DNAse was inactivated by heating at 65°C, for 10 minutes. The purity and integrity of the mRNA were checked via RT-PCR, using known methods. Then, 3: 1 samples were combined with from 15-40 ng of polyA (+) RNA, 60 units of reverse transcriptase, and 22.5 pmol of an arbitrary, non- degenerate, 18 to 22 mer primer. The reaction was allowed to proceed for from 60 to 90 minutes at 37°C, after which double stranded cDNA synthesis and amplification was carried out, in the same tube, using 22: 1 of diluted, PCR beads (lyophilized, Taq polymerase in salt buffer. These were reconstituted in water, and then prepared as described, with no other additions). The cycling protocol was: 75°C for 5 minutes, to denature mRNA, followed by two amplification cycles, where denaturation was carried out at 94°C for 30 seconds, followed by primer annealing at 52°C for 1 minute, and primer extension at 72°C, for 1 minute. Cycling was repeated, with a 1°C reduction every two amplification cycles, until the 52°C became 45°C, followed by 26 cycles of denaturation at 94°C for 30 seconds, 72°C for 1 minute, 48°C for 30 seconds, and primer extension at 72°C for 1 minute. Finally, an extension step of 7 minutes, at 72°C was performed.

[0018] Three microliter samples of amplification products were evaluated, in ethidium bromide stained gels, before cloning and sequencing.

[0019] The profiles generally exhibited a smear, indicating a diverse number of amplification products, which were cloned into mini-libraries, in accordance with Dias- Neto, et al., supra.

[0020] Any profiles which exhibited few prominent bands were excluded from further processing, except as discussed infra.

[0021] At least two, 96 well plates of clones were sequences for each mini- library. A total of 867 different ORESTES mini-libraries were generated, using 312 arbitrary primers and mRNA from all six different stages of the parasite library..

[0022] Normalized, poly-dT-primed cDNA libraries were prepared, in accordance with Soares, et al., Proc. Natl. Acad. Sci. USA, 91: 9228-9232 (1994), using abundantly available mRNA from adult worms.

[0023] Once the library was obtained, sequencing was carried out using standard, fluorescence labeling dye terminator protocols. As sequences were generated, redundancy was monitored on line, in accordance with Paquola, et al., Bioinformatics, 19: 1587-1588 (2003). Sequencing continued, until from 45-50% of new sequences were not redundant.

[0024] A total of 163,586 EST"reads"were obtained, with 151,684 coming from the ORESTES library, and the remainder from the normalized adult worm library.

[0025] For the SAGE library ("Serial Analysis of Gene Expression"), mRNA from adult worms was used to construct the library, using a commercial kit. Poly-A mRNA was treated with DNA prior to extraction with oligo-dT. Concatamers were cloned and sequenced, and tags were derived high quality sequence segments only.

[0026] Relative abundance of transcript was determined by comparing the SAGE tag list with the complete SmAE data set, and with all full-length cDNA sequences from S. mansoni.

EXAMPLE 2 [0027] EST sequence chromatograms, obtained as described, supra, were stored, processed and trimmed via Paquola, et al., supra. Sequences with at least 100 phred-15 base pairs, as defined by Ewing, et al., Genome Res., 8 : 175-185 (1998), incorporated by reference, were accepted, and evaluated. They were filtered using BLASTN analysis, as well as the GenBank NT database, and BlastMachine, in order to eliminate sequences that matched non-S. mansoni sequences with e-values # 10-15, and had at least 98% identity along at least 75 nucleotides. Further reads were excluded that matched S. mansoni ribosomal or mitochondria sequences, as well as transposon sequences which had e-values # 10-15, and at least 85% identity along at least 75 nucleotides. Further reads which were excluded were those which matched bacterial sequences, with e-values # 10-2°, with at least 95% identity along at least 75 nucleotides.

[0028] Additional transposon and bacterial sequences were filtered by comparing the data set, with BLASTX, against the set of transposon and bacterial sequences from GenBank NR, and eliminating the sequences with a matching e-value &num 10-4, and at least 30% identity along at least 75 amino acids with transposons, or a matching e-value of # 10-6, and at least 95% identity along at least 75 amino acids with bacteria. The remaining ESTs were clustered and assembled using CAP3, in accordance with Huang, et al., Genome Res. , 9: 868-877 (1999), incorporated by reference.

[0029] Of a total of 163,586 EST reads (151,684 from ORESTES, 11,902 from a normalized worm library), a filtered dataset of 124,681 analyzed, EST reads were assembled into 30,988 assembled EST sequences. These, as summarized in Table 1, which follows, will be referred to as"SmAEs"for Schistosoma mansoni Assembled EST sequences.

BRIEF DESCRIPTION OF THE FIGURES [0030] Figure 1 shows the results of in vivo experiments using SEQ ID NO: 2141.

[0031] Figure 2 shows the results of in vivo experiments using SEQ ID NO: 2143.

[0032] Figure 3 shows the results of in vivo experiments using SEQ ID NO: 2145.

[0033] Figure 4 shows the results of in vivo experiments using SEQ ID NO: 2147.

[0034] Figure 5 shows the results of in vivo experiments using SEQ ID NO: 2149.

[0035] Figure 6 shows the results of in vivo experiments using SEQ ID NO: 2151.

[0036] Figure 7 shows the results of in vivo experiments using a portion of SEQ ID NO: 2143.

TABLE 1 Number of reads Total sequenced reads 163, 586 Total analyzed reads 124,640 Adults 33, 180 Eggs 19,077 Miracidia 18,638 Germ balls 16,715 Cercariae 10,014 Cultured day-7 schistosomula 27,016 Average EST size (bp after trimming) 385.4 Total number of SmAE sequences 30,988 Number of cotings 12, 322 Number of orphan sequences (singlets) 18,666 Average contig size (bp) 505 Total SmAEs matching known S. mansoni sequences 7,086 (23%) match to S. mansoni known genes from Genbank 639 (2%) match to S. mansoni known ESTs from dbEST 6, 447 (21 %) Total SmAEs with putative new S. mansoni gene fragments 23,902 (77%) match to S. mansoni known proteins (new paralogs) 449 (1%) match to genes of other organisms (new orthologs) 6,274 (20%) no-match in GenBank (Fragments with known function) 17,179 (55%) Estimated gene complement 13, 960-14,205 [0037] The data set is estimated to have sampled 92% of the S. mansoni transcriptome. A total of 77% of the sequences are new, S. mansoni sequences, as summarized in Table 1. Of these, 1% are believed to be novel paralogs, 20% novel orthologs, and the remaining 55%, new sequences with unknown functions. As compared to GenBank, on the average an SmAE sequence had 32% coverage in a matching gene in GenBank, and 359 of the novel orthologs had their entire coding region fully sequenced. A listing of these novel orthologs, which have been sequenced completely is set forth in Table 2.

[0038] The estimated number of genes in the parasite was predicted to be around 14,000, which is in accordance with other sequenced invertebrates, such as D. melanogaster (Adams, et al., Science, 298 : 2157-2167 (2002) ) ; C. intestinalis (Dehal, et al., Science, 298: 2157-2167 (2002), and C. elegans (C. Elegans, Sequencing Consortium, Science, 282: 2012-2018) (2002) ). Based upon extrapolation from non-redundant bases acquired from adult worms ESTs, indicates that about 7200 genes are expressed at this stage. A total of 58,846 SAGE tags was obtained, and the unique tags numbered 6,263.

These data suggest almost all adult transcripts were sampled. The accompanying sequence listing provides data for this material.

TABLE 2 &num SmAE Orthologgene Accession e-value 1 C603809. 1 Elongation factor 2b [Drosophila melanogaster] NP_525105 0.0 2 C604453.1 actin [Crassostrea gigas] 017320 0.0 3 C605243.1 CG1345 gene product [Drosophila NP651617 0.0 melanogaster] 4 C607639.1 RIKEN cDNA 0610042A05 gene [Mus AAH08646 0.0 musculus] 5 C605838.1 ebiP3616 [Anophelesgambiae str. PEST] EAA09206 0.0 6 C600652.1 translation initiationfactor 4A-like protein CAC18543 0.0 [Echinococcus multilocularis] 7 C607326.1 tubulin, beta, 2 [Homo sapiens] NP006079 0.0 8 C603077.1 adenine homocysteine hydrolase [Xenopus P51893 0.0 laevis] 9 C611641.1 Vha55-P1 ; VacuolarH [+] -ATPase 55kD B Nu-476908 0.0 subunit [Drosophila melanogaster] 10 C601069.1 CG9539 gene product [Drosophila Nu 609034 0.0 melanogaster] 11 C606462.1 transketolase [Mus musculus] NP_033414 0.0 12 C610252.1 beta-tubulin [Xenopus laevis] P30883 0.0 13 C609566.1 chaperonin-containing T-complex protein 1 eta AAM34673 0.0 subunit [Danio rerio] 14 C605279.1 pyruvate kinase [Takifugu rubripes] BAB92968 0.0 15 C610735.1 F-1-ATPase beta-subunit precursor [Bos P00829 0.0 taurus] 16 C604097.1 glutamate dehydrogenase 3 [Oncorhynchus AAM73777 0.0 mykiss] 17 C607185.1 proteasome (prosome, macropain 26S subunit, XP123843 0.0 ATPase 3 [Mus musculus] 18 C600603.1 karyopherin alpha 3 (importin alpha 4 [Homo 000505 0.0 sapiens] 19 C609435.1 expressed sequenceAA617263 [Mus NP_598890 1. 0E-180 musculus] 20 C606116.1 GTP-binding protein beta subunit [Loligo P23232 1. 0E-176 forbesi] 21 C604221.1 agCP5574 [Anophelesgambiae str. PEST] EAA11760 1. 0E-174 22 C607065.1 protein phosphatase 2A: SUBUNIT=alpha 1803244A 1. 0E-171 [Xenopus laevis] 23 C600740.1 ribosomal protein L3 [Spodoptera frugiperda] AAL62468 1. 0E-166 24 C611330.1 ornithineaminotransferase [Rattusnorvegicus] NP071966 1. 0E-164 25 C601851.1 methionineadenosyltransferase II, alpha NP_005902 1. 0E-162 [Homo sapiens] 26 C607426.1 splicing factor U2AF [Mus musculus] P26369 1. 0E-158 27 C604236.1 RE21802p [Drosophila melanogaster] AAM11402 1. OE-157 28 C608337.1 aspartate aminotransferase [Caenorhabditis NP509047 1. 0E-157 elegans] 29 C606569.1 prolidase [Suberitesdomuncula] CAA75230 1. 0E-155 30 C612084.1 septin 6 [Mus musculus] AAH10489 1. OE-154 31 C608354.1 poly (A bindingprotein, cytoplasmic 1 [Rattus NP_599180 1. 0E-154 norvegicus] 32 C604668.1 40-kDa V-ATPase subunit [Manduca sexta] Q25531 1. 0E-152 33 C607154.1 rab GDP-dissociationinhibitor [Branchiostoma CAB46230 1. 0E-151 floridae] 34 C611775.1 phosphoribosylpyrophosphate synthetase 1 NP002755 1. 0E-150 [Homo sapiens] 35 C605174.1 histidyl tRNAsynthetase [Mus musculus] NP_032240 1. 0E-148 36 C606537.1 MAP kinase [Aplysiacalifornica] AAA83210 1. 0E-144 37 C600176.1 similar to GLUTAMINESYNTHETASE (GLUTAMATE-- aMMONIA LIGASE [Mus musculus] XP_123533 1.0E-142 38 C600019.1 SWI/SNF related, matrixassociated, actin AAH26783 1. 0E-140 dependent regulator of chromatin, subfamily d, member 1 [Mus musculus] 39 C605781.1 eukaryotic initiationfactor 4a NP_594854 1. 0E-137 [Schizosaccharomyces pombe] 40 C606720.1 RIKEN cDNA 2310003C10 [Mus musculus] XP_130224 1. 0E-134 41 C608591.1 BRG1/brm-associatedfactor 53A [Mus XP124032 1. 0E-131 musculus] 42 C610802.1 CG7954-PB [Drosophila melanogaster] AAN14315 1. OE-127 43 C603698.1 DnaJ-likeprotein [Cercopithecusaethiops] AAK81721 1. 0E-126 44 C606400.1 CG2246-PB [Drosophila melanogaster] AAN14244 1. 0E-126 45 C602922.1 fibrillarin[Caenorhabditis elegans] NP_506691 1. 0E-125 46 C600207.1 Zmpste24 [Mus musculus] AAK38172 1. 0E-125 47 C611213.1 similar toUroporphyrinogen decarboxylase XP_046565 1.0E-125 (URO-D (UPD [Homo sapiens] 48 C610415.1 cyclin-dependent kinase 5 EC 2.-human JE0374 1. 0E-124 49 C603862.1 pyruvate dehydrogenase type 11 alpha subunit P26268 1. 0E-123 [Ascaris suum] 50 C604689.1 ribosomal protein L [Argopectenirradians] AAN05596 1. 0E-120 51 C609291.1 FX protein [Cricetulus griseus] AAM91926 1. 0E-117 52 C608022.1 CG12030 gene product [Drosophila NP_612044 1. OE-116 melanogaster] 53 C606474.1 casein kinase I [Ciona savignyi] BAC05520 1. 0E-116 54 C612207.1 unnamed protein product [Homo sapiens] BAA91542 1. 0E-115 55 C604091.1 ribosomal protein L5 [Bombyx mori] 076190 1. 0E-112 56 C601324.1 ribosomal protein S2 [Urechis caupo] P49154 1. 0E-111 57 C607146.1 ribosomal protein S4 [Gallus gallus] P47836 1. 0E-109 58 C605600.1 agCP2962 [Anophelesgambiae str. PEST] EAA08424 1. 0E-109 59 C606295.1 agCP4242 [Anophelesgambiae str. PEST] EAA04901 I. OE-108 60 C609913.1 GPTHUMAN N-Q9H3H5 1. OE-107 acetylglucosaminephosphotransferase GPT (G1 PT (N acetylglucosamine-1-phosphate transferase (GlcNAc-1-P transferase 61 C612017. 1 prohibitin [Mus musculus] NP_032857 1. 0E-106 62 C603844.1 RIKEN cDNA 1700029A20 [Mus musculus] NP_081563 1. 0E-104 63 C611019.1 SelD protein [Drosophila melanogaster] 018373 1. 0E-103 64 C601609.1 casein kinase 11, betasubunit [Mus musculus] NP 034105 1. 0E-102 65 C604197.1 ebiP8896 [Anophelesgambiae str. PEST] EAA00062 1. 0E-101 66 C601067.1 2-oxoglutarate carrier protein [Rattus 2116232A 1. 0E-100 norvegicusr] 67 C607183.1 regulatory subunit ofcAMP-dependent histone BAA11899 5. 0E-99 kinase [Hemicentrotus pulcherrimus] NP 004716 5. 0E-99 68 C606163.1 BUB3 buddinguninhibited by benzimidazoles 3 homolog (yeast; BUB3 (budding Uninhibited by benzimidazoles 3, yeast homolog ; mitotic checkpoint component [Homo sapiens] 69 C609011. 1 QM protein [Bombyx mandarina] 096647 3. 0E-98 70 C610016.1 RIKEN cDNA 2610016C12 [Mus musculus] NP_080678 4. 0E-98 71 C603392.1 TfilB-P1 [Drosophilamelanogaster] NP_476888 3. 0E-97 72 C605473.1 Sop2-P1 ; lethal (2 34Dd [Drosophila NP_476596 4. 0E-97 melanogaster] 73 C608700.1 CG10907 gene product [Drosophila NP_648508 6. 0E-97 melanogaster] 74 C603425.1 ribosomal protein L7a [Gallus gallus] P32429 2. 0E-96 75 C610963.1 agCP7321 [Anophelesgambiae str. PEST] EAA06918 2. 0E-96 76 C601703.1 ribosomal protein S6 [Branchiostoma floridae] 001727 6. 0E-96 77 C611490.1 Unknown (proteinfor MGC: 27286 [Homo AAH15848 1. 0E-95 sapiens] 78 C602076.1 alpha 4 subunit of 20S proteasome [Carassius Q9PTW9 2. 0E-95 auratus] 79 C612574.1 actin capping protein beta subunit, isoform 2 U07826 2. 0E-94 [Gallus gallus] 80 C600673.1 Unknown (protein for MGC: 33928 [Homo AAH31096 1. 0E-93 sapiens] 81 C606297.1 proliferating cellnuclear antigen subtype1 BAA92700 1. 0E-93 [Xenopus laevis] 82 C601367.1 A Chain A, The Structure Of Alpha-N-1 KTB 4. 0E-93 Acetylgalactosaminidase 83 C610177.1 proteasome (prosome, macropain subunit, NP002780 1. 0E-92 alpha type, 4 [Homo sapiens] 84 C603622.1 N-ethylmaleimidesensitive fusion protein NP_080174 5. 0E-92 attachment protein alpha [Mus musculus] 85 C610458.1 KdelR-P1 ; ER lumenprotein retaining receptor NP_477296 7. 0E-92 2 [Drosophila melanogaster] 86 C601590.1 cathepsin L precursor [Sarcophaga peregrina] Q26636 4. 0E-91 87 C606034.1 gene trap locus 3 [Mus musculus] XP_125117 3. OE-90 88 C603706.1 RIKEN cDNA 2510008H07[Mus musculus] NP_079862 4. OE-90 89 C608993.1 rho protein [Aplysia californica] P01122 1. 0E-89 90 C606027.1 mRNA cleavage factor I 25 kDa subunit [Homo NP008937 3. 0E-89 sapiens] 91 C605905.1 lin-10 protein homolog[Rattus norvegicus] NP_620609 4. 0E-89 92 C600233.1 RIKEN cDNA 2410004J02 [Mus musculus] NP_598517 2. 0E-88 93 C605165.1 hypothetical protein FLJ10702 [Homo sapiens] NP060654 5. 0E-88 94 C608805.1 RAB2 [Lymnaea stagnalis] Q05975 2. 0E-87 95 C605341.1 CG6838 gene product [altl] [Drosophila AAF51848 4. 0E-86 melanogaster] 96 C607090.1 multicatalytic endopeptidase complex EC3 9 6 S38529 2. 0E-85 chain XC8-clawed frog 97 C610193.1 data source: MGD, sourcekey: MG1 : 1350917, BAB27110 6. 0E-85 evidence: ISS-putative# ribosomal protein S3 [Mus musculus] 98 C608036.1 ribosmal protein smallsubunit [Homo sapiens] AAA35682 3. 0E-84 99 C609520.1 integrin beta 4 bindingprotein; p27BBP; NP002203 3. 0E-84 eukaryotic translation initiation factor 3A [Homo sapiens] 100 C607425.1 CsCDC42 [Ciona savignyi] BAA25400 3. 0E-83 101 C601598.1 agCP2424 [Anophelesgambiae str. PEST] EAA08621 3. 0E-83 102 C610490.1 Arf4 [Xenopus laevis] P51644 2. 0E-82 103 C606688.1 myophilin antigen [Echinococcus granulosus] CAA82316 4. 0E-81 104 C604777.1 CG8031 gene product [Drosophila NP650252 2. 0E-80 melanogaster] 105 C604306.1 Hypotheticalprotein Y48G8AL 4 AAM15614 4. 0E-80 [Caenorhabditis elegans] 106 C612269.1 ras-related C3botulinum toxin substrate 1 (rho AAH04247 1. 0E-79 family, small GTP binding protein Rac1 [Homo sapiens] 107 C607608.1 BAP 1 [Bos taurus] P23196 1. 0E-78 108 C600256.1 calmodulin [Metridiumsenile] BAB61794 2. 0E-78 109 C610509.1 similar toNADH-ubiquinone oxidoreductase 23 XP 129107 2. 0E-78 kDa subunit, mitochondrial precursor (Complex 1-23KD (Cl- 23KD (TYKY subunit 110 C612582.1 ribosomal protein L10a [Rattus norvegicus] NP 112327 4. 0E-78 111 C605252.1 transcriptionelongation factor B polypeptide 1-NP_003188 5. 0E-78 like ; organ of Corti protein 2 [Homo sapiens] 112 C601587.1 annexin !-human LUHU8 9. 0E-78 113 C610528.1 SET translocation [Mus musculus] NP076360 1. 0E-77 114 C611350.1 phosphomannomutase 2[Mus musculus] NP_058577 5. 0E-77 115 C602648.1 vacuolar proton pump delta polypeptide [Homo NP057078 2. 0E-76 sapiens] 116 C610375.1 agCP5675 [Anophelesgambiae str. PEST] EAA11918 5. 0E-76 117 C609509.1 agCP10880 [Anophelesgambiae str. PEST] EAA12363 3. 0E-75 118 C602928.1 ADP-ribosylation factor-like protein 3 [Xenopus AAL77055 7. 0E-75 laevis] 119 C608961.1 similar to TARDNA-binding protein-43 (TDP-43 XP110497 9. 0E-75 [Mus musculus] 120 C608299.1 ribosomal protein L7 [Argopecten irradians] AAN05591 1. 0E-74 121 C608238.1 carboxy terminus ofHsp70p-interacting protein XP083939 2. 0E-74 [Homo sapiens] 122 C609961.1 14-3-3 protein zeta chain-bovine S65013 1. 0E-73 123 C600243.1 RAS-related protein[Caenorhabditis elegans] NP_503397 3. 0E-72 124 C604924.1 agCP14033 [Anophelesgambiae str. PEST] EAA09167 3. 0E-72 125 C610487.1 Ubc7 gene product [Drosophila melanogaster] AAF48626 1. OE-71 126 C606337.1 ribosomal protein L13a [Ictalurus punctatus] AAK95140 1. 0E-71 127 C608646.1 putative prostatecancer tumor suppressor AAH10370 2. 0E-71 [Homo sapiens] 128 C609538.1 I-isoaspartyl proteincarboxyl methyltransferase NP_571540 6. 0E-71 [Danio rerio] 129 C607601.1 G10 protein [Branchiostoma belcheri] AAK81863 7. 0E-71 130 C611535.1 CG3061 gene product [Drosophila NP_650328 8. 0E-71 melanogaster] 131 C601526.1 ribose 5-phosphateisomerase A [Mus NP_033101 3. 0E-70 musculus] 132 C607429.1 SNAP25 fusion protein [Loligo pealei] AAM18191 1. OE-69 133 C610579.1 Unknown (proteinfor IMAGE : 3505010 [Homo AAH05148 2. 0E-69 sapiens] 134 C604598.1 histone gene complex 1 [Mus musculus] NP038576 4. 0E-69 135 C605248.1 ribosomal proteinL18a ; 60S ribosomal protein NP000971 1. 0E-68 L18a [Homo sapiens] 136 C610236.1 CG6056 gene product [Drosophila AAF55874 2. 0E-68 melanogaster] 137 C601999.1 diphthine synthase[Caenorhabditis elegans] NP_496427 5. 0E-68 138 C603765.1 mago-nashi homolog [Homo sapiens] NP002361 6. 0E-68 139 C605050.1 RTC domain containing1 ; RNA 3'-terminal NP003720 8. 0E-68 phosphate cyclase [Homo sapiens] 140 C601420.1 ribosomal protein S13; 40S ribosomal protein NP001008 1. 0E-67 S13 [Homo sapiens] 141 C612194.1 quinoiddihydropteridine reductase [Rattus NP071785 2. 0E-67 norvegicus] 142 C611908.1 agCP8207 [Anophelesgambiae str. PEST] EAA13967 2. 0E-67 143 C609770.1 agCP13846 [Anophelesgambiae str. PEST] EAA06004 3. 0E-67 144 C611251.1 vacuolar H+ ATPase E subunit [Homo sapiens] NP001687 4. 0E-67 145 C606060.1 40S ribosomal proteinS23 [Lumbricus rubellus] CAC14789 5. 0E-67 146 C609431.1 RE63412p [Drosophila melanogaster] AAL49196 1. OE-66 147 C606264.1 EBF3-S [Homo sapiens] AAK07557 3. 0E-66 148 C602561.1 data source: MGD, sourcekey: MGl : 1928344, BAB23170 9. 0E-66 evidence : ISS-putative#vacuolar protein Sorting 29 (S. pombe [Mus musculus] 149 C607084.1 CG4593 gene product [Drosophila AAF46235 1. 0E-65 melanogaster] 150 C612233.1 centrin 1; calciumbinding protein; Centrin-1; NP004057 2. 0E-65 EF-hand protein [Homo sapiens] 151 C600674.1 ubiquitin-conjugating enzyme [Drosophila P25867 4. 0E-65 melanogaster] 152 C601902.1 RAD23 homolog B (S. cerevisiae [Homo AAH20973 1. 0E-64 sapiens] 153 C608652.1 LD44221 p [Drosophila melanogaster] AAM11189 2. 0E-64 154 C612343.1 HSPC177 [Homo sapiens] AAF29140 9. 0E-64 155 C605206.1 clathrin coat assembleprotein [Caenorhabditis NP504559 2. 0E-63 elegans] 156 C611248.1 dehydrogenase [Caenorhabditis elegans] NP_498146 3. 0E-63 157 C607538.1 CG9911 gene product [Drosophila NP_573111 4.0E-63 melanogaster] 158 C611049.1 endless ; nonjumper-262 [Drosophila NP_511150 9. 0E-63 melanogaster] 159 C607040.1 ebiP7010 [Anophelesgambiae str. PEST] EAA12698 I. OE-62 160 C604244.1 muscle RAS oncogenehomolog ; muscle and NP036351 2. 0E-62 microspikes RAS [Homo sapiens] 161 C612360.1 RP42 homolog [Homo sapiens] AAH09478 4. 0E-62 162 C608607.1 agCP10213 [Anophelesgambiae str. PEST] EAA05757 4. 0E-62 163 C718961.1 ribosomal protein L27; 60S ribosomal protein NP000979 9. 0E-62 L27 [Homo sapiens] 164 C602326.1 CGI-35 protein [Homo sapiens] NP057046 1. 0E-61 165 C609343.1 solute carrier family25 (mitochondrial NP_068380 2. 0E-61 deoxynucleotide carrier, member 19; mitochondrial uncoupling protein 1 ; deoxynucleotide carrier, mitochondrial [Homo sapiens] 166 C605379.1 Elongation factor 1 beta ; Elf 1 beta [Drosophila NP524808 1. 0E-60 melanogaster] 167 C602131.1 ribosomalprotein S15a [Taenia solium] AAK29203 2. 0E-60 168 C608722.1 agCP13402 [Anophelesgambiae str. PEST] EAA06529 5. 0E-60 169 C610185.1 calcyphosine [Halichondria okadai] BAB89357 1. 0E-59 170 C600946.1 agCP10825 [Anophelesgambiae str. PEST] EAA09841 3. 0E-59 171 C601092.1 mammalian A1,A2 /B1 hnRNP homologue P21522 2. 0E-58 [Schistocerca americana] 172 C608940.1 H2A protein (AA 1-124 [Platynereis dumerilii] P19178 3. 0E-58 173 C612409.1 similar to U2 small nuclear ribonucleoprotein B XP 149169 3. 0E-58 [Mus musculus] dbj 174 C612191.1 hypothetical proteinMGC4093 [Homo sapiens] NP085055 8. 0E-58 175 C602816.1 ebiP6131 [Anophelesgambiae str. PEST] EAA14354 4. 0E-57 176 C603503.1 ribosomal protein S11[Gallus gallus] BAB40319 6. 0E-57 177 C601643.1 general transcriptionfactor IIE, polypeptide 2 NP002086 8. 0E-57 (beta subunit, 34kD [Homo sapiens] 178 C603372.1 centrin 3-datasource : MGD, source BAB27862 8. 0E-57 key: MG1 : 1097706, evidence: ISS- putative [Mus Musculus] 179 C608166.1 CG3203 gene product [Drosophila NP_572346 1. 0E-56 melanogaster] 180 C612635.1 vacuolar ATP synthase 16kDa proteolipid AAM28211 2. 0E-56 subunit [Danio rerio] 181 C600706.1 Unknown (protein for MGC: 21169 [Homo AAH14842 5. 0E-56 sapiens] 182 C609647.1 MCT-1 protein [Homo sapiens] NP054779 8. 0E-56 183 C600716.1 scavenger receptor class Btype I [Bos taurus] AAB70920 5. 0E-55 184 C603353.1 ribosomalprotein S16 [Spodoptera frugiperda] AAL26583 2. 0E-54 185 C603336.1 uridine monophosphatekinase; uridine-cytidine NP036606 2. 0E-54 kinase 2; uridine kinase; nucleosidephosphate Kinase; NMP-kinase [Homo sapiens] 186 C608325.1 RIKEN cDNA 1500016L11 [Mus musculus] XP_133183 3. 0E-54 187 C608523.1 growth factor receptorbound protein 2 [Mus NP032189 1. 0E-53 musculus] 188 C609720.1 CG8891 gene product [Drosophila NP_608890 1. 0E-53 melanogaster] 189 C611688.1 similar to dCMP deaminase [Mus musculus] AAH31719 2. 0E-53 190 C608635.1 similar to putative transmembrane protein; XP170670 6. 0E-53 homolog of yeast Golgi membrane protein Yif1p (Yip1p-interacting factor [Homo sapiens] 191 C602695.1 ribosomal protein L9 [Mus musculus] NP_035422 9. 0E-53 192 C602003.1 agCP1472 [Anophelesgambiae str. PEST] EAA07465 2. 0E-52 193 C607870.1 similar to germinal histone H4 gene [Homo XP172562 2. 0E-52 sapiens] 194 C603818.1 Unknown (protein for MGC: 12282 [Homo AAH05248 3. 0E-52 sapiens] 195 C607191.1 hypothetical proteinFLJ21087 [Homo sapiens] XP_166303 3. 0E-52 196 C610089.1 Roc1a gene product[Drosophila melanogaster] NP_569852 5. 0E-52 197 C602021.1 RIKEN cDNA 1110014C03[Mus musculus] XP_127008 1. 0E-51 198 C600596.1 dUTP pyrophosphatase[Homo sapiens] NP_001939 3. 0E-51 199 C606144.1 histone H2B; histone [Drosophila NP_478141 4. 0E-51 melanogaster] 200 C606214.1 agCP6233 [Anophelesgambiae str. PEST] EAA11799 6. 0E-51 201 C603784.1 Chmp1 gene product [Drosophila NP_649051 1.0E-50 melanogaster] 202 C606558.1 ribosomal protein S7 [Argopecten irradians] AAN05602 2. 0E-50 203 C605605.1 RIKEN cDNA 2510010F15[Mus musculus] NP_080730 4. 0E-50 204 C605166.1 agCP6124 [Anophelesgambiae str. PEST] EAA11346 4. 0E-50 205 C610102.1 testis enhanced gene transcript-like protein Q91A79 7. 0E-50 [Paralichthys olivaceus] 206 C603157.1 agCP8097 [Anophelesgambiae str. PEST] EAA14494 8. 0E-50 207 C610125.1 similar to RIKEN cDNA0610042E07 [Mus XP_145307 3. 0E-49 musculus] 208 C612230.1 Dxpa protein [Drosophila melanogaster] BAA06690 3. 0E-49 209 C600581.1 agCP2729 [Anophelesgambiae str. PEST] EAA13201 5. 0E-49 210 C608732.1 LP06328p [Drosophila melanogaster] AAM27524 9. 0E-49 211 C605237.1 mapmodulin ; anon-fast-evolving-3A2 NP523780 3. 0E-48 [Drosophila melanogaster] 212 C603065.1 Tetraspanin 86D[Drosophila melanogaster] NP_524309 3. 0E-48 213 C601065.1 nucleoside diphosphate kinase [Columba livia] Q90380 9. 0E-48 214 C604360.1 GABA (A receptor associated protein AAG13318 1. 0E-47 [Gillichthys mirabilis] 215 C612438.1 ibosomalprotein L26 [lctalurus punctatus] AAK95152 2. 0E-47 216 C611634.1 CG1349 gene product [Drosophila NP651825 4. 0E-47 melanogaster] 217 C610110. 1 ribosomal protein L44 [Chlamys farreri] AAM94276 6. 0E-47 218 C610151. 1 ebiP5478 [Anophelesgambiae str. PEST] EAA05859 1. 0E-46 219 C605978.1 chargedmultivesicular body protein AAG01448 1. 0E-46 1/chromatin modifying protein 1 [Homo sapiens] 220 C611347.1 hypothetical proteinMGC10540 [Homo NP_115729 2. 0E-46 sapiens] 221 C603431.1 carbonic anhydrase 2 ; CA II [Mus musculus] NP_033931 3. 0E-46 222 C607079.1 ubiquitin carboxy-terminal hydrolase-6EC 3-JC7117 8. 0E-46 chicken 223 C605972.1 ebiP2778 [Anophelesgambiae str. PEST] EAA08176 1. 0E-45 224 C600998.1 similar to RIKEN cDNA5630401J11 gene; XP129588 2. 0E-4. 5 expressed sequence AA409698 [Mus musculus] 225 C602037.1 6-phosphogluconolactonase [Mus musculus] NP_079672 3. 0E-45 226 C611827.1 translocase of innermitochondrial membrane NP006326 4. 0E-4. 5 17 homolog A (yeast; preprotein translocase [Homo sapiens] 227 C604044.1 ribosomalprotein L27 [Ictalurus punctatus] AAK95153 9. 0E-45 228 C600072.1 data source: SPTR, sourcekey: Q15701, BAB24776 3. 0E-44 evidence: ISS-putative-similar to DYNEIN LIGHT CHAIN 1, CYTOPLASMIC (PROTEIN INHIBITOR OF NEURONAL NITRIC OXIDE SYNTHASE (PIN [Mus musculus] 229 C600677.1 ubiquitin-conjugating enzyme E2 variant 1 CAC16954 3. 0E-44 [Homo sapiens] 230 C601555.1 homologous to animal glutathione peroxidases P30708 4. 0E-44 [Nicotiana sylvestris] 231 C600543.1 H20J04. p [Caenorhabditis elegans] NP_494762 4. 0E-44 232 C604691.1 similar to retinalshort chain dehydrogenase XP143619 4. 0E-44 reductase [Mus musculus] 233 C611202.1 RIKEN cDNA 1500006009 [Mus musculus] NP 079711 1. 0E-43 234 C605347.1 agCP2127 [Anophelesgambiae str. PEST] EAA10491 3. 0E-43 235 C606117.1 RIKEN cDNA2400003C14 [Mus musculus] XP_134531 5. 0E-43 236 C602699.1 8 kDa outer arm dynein light chain Q39580 4. 0E-42 [Chlamydomonas reinhardtii] 237 C710782.1 small nuclearribonucleoprotein D1 polypeptide NP_008869 4. 0E-42 (16kD [Homo sapiens] 238 C604704.1 db83 [Rattusnorvegicus] NP067703 6. 0E-42 239 C610152.1 putative enhancer ofrudimentary CAC18546 2. 0E-41 [Echinococcus multilocularis] 240 C610073.1 T26A5. p [Caenorhabditis elegans] NP_498422 2. 0E-41 241 C608012.1 ubiquitin [Ciona savignyi] BAC06474 2. 0E-41 242 C601298.1 ribosomal protein L23a [Argopecten irradians] AAN05592 2. 0E-41 243 C603081.1 agCP9917 [Anophelesgambiae str. PEST] EAA00657 4. 0E-41 244 C608609.1 Porin 31 HM [human, skeletal AAB20246 6. 0E-41 musclemembranes, Peptide, 282 aa] 245 C710527.1 KIAA1665 protein [Homo sapiens] BAB33335 1. 0E-40 246 C612312.1 Tetraspanin 97E [Drosophila melanogaster] NP_524524 2. 0E-40 247 C601637.1 CG11858 gene product [Drosophila NP_651364 3. 0E-39 meianogaster] 248 C604751.1 CGI-99 protein [Homo sapiens] NP057123 1. 0E-38 249 C608248.1 GABA (A receptor-associated protein-like 2; NP009216 3. 0E-38 ganglioside expression factor 2; MAP1 light chain 3 related protein [Homo sapiens] 250 C612139.1 similar toTranscription factor BTF3 (RNA XP038290 1. 0E-37 polymerase B transcription factor 3 [Homo sapiens] 251 C612163.1 CG7949 gene product [Drosophila Q9VTC4 1. 0E-37 melanogaster] 252 C606671.1 voltage-dependent anion channel [Gallus AAF73513 6. 0E-37 gallus] 253 C611164.1 hypothetical protein [Homo sapiens] CAB66619 3. 0E-36 254 C602607.1 hypothetical proteinFLJ12960 [Homo sapiens] NP078914 3. 0E-36 255 C606773.1 putativeU6-snRNA-associated protein CAC18540 8. 0E-36 [Echinococcus multilocularis] 256 C604744.1 snRNP C [Xenopus laevis] Q03369 1. 0E-35 257 C601559.1 proteasome (prosome, macropain26Ssubunit, NP002805 1. 0E-35 non-ATPase, 10 [Homo sapiens] 258 C606336.1 agCP11479 [Anophelesgambiae str. PEST] EAA12342 1. 0E-35 259 C600698.1 40S ribosomal proteinS26 [Caenorhabditis elegans] Nu-493571 8. 0E-35 260 C606681.1 suppressor of Ty 4homolog 1 (S. cerevisiae; NP003159 1. 0E-34 suppressor of Ty (S. cerevisiae 4 homolog 1 [Homo sapiens] 261 C602954.1 similar tohypothetical protein FLJ13448 [Mus XP_109592 2. 0E-34 musculus] 262 C606776.1 Adeninephosphoribosyltransferase ; Apt1 NP013690 3. 0E-34 [Saccharomyces cerevisiae] 263 C600222.1 hypothetical protein MGC10198 [Homo NP689895 1. 0E-33 sapiens] 264 C610210. 1 data source: SPTR, sourcekey: Q9ZUU2, BAB23870 1. 0E-33 evidence: ISS-putative-related to F3G5 PROTEIN [Mus musculus] 265 C709996.1 CG11985 gene product [Drosophila Q9VH14 3. 0E-33 melanogaster] 266 C611744.1 CT28MOUSE Protein C20orf28 homolog Q99JL1 3. 0E-33 267 C606251.1 hypothetical protein [Taenia solium] CAD21525 5. 0E-33 268 C603749.1 heat shock 10 kDaprotein 1 (chaperonin 10; NP032329 7. 0E-33 mitochondrial chaperonin 10 [Mus musculs] 269 C603494.1 kappa-ras 1 [Homo sapiens] NP065078 2. 0E-32 270 C607582.1 40S ribosomal protein S27 [Lumbricus rubellus] CAB58439 2. 0E-32 271 C608956.1 c-myc binding protein [Homo sapiens] BAA14006 2. 0E-32 272 C608627.1 agCP3588 [Anophelesgambiae str. PEST] EAA04034 5. 0E-32 273 C606722.1 expressed sequenceAA545216 [Mus XP_129630 2. 0E-31 musculus] 274 C605974.1 14 kDa perchloric acidsoluble protein [Capra AAC72281 2. 0E-31 hircus] 275 C605458.1 translation initiation factor eIF4E [Aplysia 077210 2. 0E-31 californica] 276 C610929.1 agCP7916 [Anophelesgambiae str. PEST] EAA08835 2. 0E-31 277 C601661.1 synaphin A [Loligopealei] BAB62069 5. 0E-31 278 C606808.1 cleavage stage histone H2A [Psammechinus AAB48831 5. 0E-31 miliaris] 279 C610909.1 actin related protein2/3 protein complex CAC14790 2. 0E-30 subunit p16 [Lumbricus rubellus] 280 C608152.1 mevalonatekinase [Hevea brasiliensis] AAL18925 2. 0E-30 281 C607922. 1 mitochondrialribosomal protein L13; L13 NP054797 3. 0E-30 protein [Homo sapiens] 282 C604386.1 ribosomal protein S10 [Spodoptera frugiperda] Q962R9 1. OE-29 283 C608770.1 ribosomal protein L35 [Choristoneura parallela] AAM53950 8. 0E-29 284 C601581. 1 similar to CD151 antigen [Mus musculus] AAH12236 6. 0E-28 285 C710392.1 similar to RIKEN cDNA0610041E09 gene AAH26285 3. 0E-27 [Homo sapiens] 286 C611252.1 TATA box binding protein (TBP-associated NP005636 3. 0E-27 factor, RNA polymerase 11, K, 18kD [Mus musculus] 287 C602979.1 ribosomal protein rpa6 [Schizosaccharomyces NP_594358 2. 0E-26 pombe] 288 C605175.1 agCP14222 [Anophelesgambiae str. PEST] EAA09174 2. 0E-26 289 C606346.1 cut up; Cytoplasmicdynein light chain 1; NP525075 3. 0E-26 Dynein light chain; dynein light-chain 1; Cytoplasmic dynein 8kD light chain [Drosophila melanogaster] 290 C604065.1 agCP7468 [Anophelesgambiae str. PEST] EAA06802 3. 0E-26 291 C608958.1 cytochrome b-5 [Oryctolagus cuniculus] P00169 5. 0E-26 292 C603073.1 general transcriptionfactor Ila, 2 (12kD subunit NP_445797 9. 0E-26 [Rattus norvegicus] 293 C606430.1 similar to RIKEN cDNA0610038F07 gene [Mus AAH29630 1. OE-25 musculus] 294 C601141.1 similar to a putativeprotein with homology to a XP059329 2. 0E-25 sequence between V2 and V3P Segments within T cell gamma receptor region [Homo sapiens] 295 C607925.1 similar to putativecyclin G1 interacting protein AAH26751 1. 0E-24 [Mus musculus] 296 C605590.1 RpP2-P1 ; Minute (2 21C ; Ribosomal protein A2; NP_476630 2. 0E-24 Ribosomal-protein-A2 [Drosophila Melanogaster] 297 C610224.1 RS28-ICTPU 40S ribosomal protein S28 gb Q90YP3 7. 0E-24 298 C606050.1 expressed sequenceAW545363 [Mus NP_598899 8.0E-24 musculus] 299 C609512.1 similar to RIKEN cDNA 2700023B17 gene XP_133381 2. 0E-23 [Mus musculus] 300 C607176.1 agCP14721 [Anophelesgambiae str. PEST] EAA03693 2. 0E-23 301 C607470.1 CG4692 gene product [Drosophila NP_611940 4. 0E-23 melanogaster] 302 C610278.1 smt3-P1 [Drosophila melanogaster] NP_477411 4. 0E-23 303 C604342.1 Y106G6H. p [Caenorhabditis elegans] NP_492733 6. 0E-22 304 C605395.1 CG7053 gene product [Drosophila NP_573326 2. 0E-21 melanogaster] 305 C606832.1 agCP6366 [Anophelesgambiae str. PEST] EAA11945 2. 0E-21 306 C601649.1 CG9117 gene product [Drosophila NP_608983 3. 0E-21 melanogaster] 307 C607453.1 RIKEN cDNA 0610007L03 [Mus musculus] NP_079591 7. 0E-21 308 C608661.1 agCP5437 [Anophelesgambiae str. PEST] EAA11271 9. 0E-21 309 C606005.1 ZC395 O. p [Caenorhabditis elegans] NP_498126 2. 0E-20 310 C603255.1 eukaryotic translationinitiation factor 4E NP004087 2. 0E-20 binding protein 2; Eukaryotic Translation initiation factor-4E binding protein- 1 [Homo sapiens] 311 C600672.1 mitochondrial carrierhomolog 2 [Xenopus BAB03397 2. 0E-20 laevis] 312 C603433.1 agCP6950 [Anophelesgambiae str. PEST] EAA05220 3. 0E-20 313 C609703.1 unnamed protein product [Homo sapiens] BAC04897 5. 0E-20 314 C601062.1 similar to PTD015protein [Mus musculus] XP 133651 6. 0E-20 315 C707332.1 presenilin enhancer [Danio rerio] AAM88324 7. 0E-20 316 C600118.1 unnamed protein product [Homo sapiens] BAC11188 1. 0E-19 317 C612287.1 agCP3889 [Anophelesgambiae str. PEST] EAA04483 2. 0E-19 318 C704594.1 putative 42-9-9 protein [Mus musculus] CAC51438 3. 0E-19 319 C604741.1 A Chain A, Nuclear Transport Factor 2Ntf2 1ASK 4. 0E-19 H66a Mutant 320 C605704.1 similar tohypothetical protein FLJ10342 [Mus XP_131324 9. 0E-19 musculus] 321 C600556.1 Mi-2 histonedeacetylase complex protein 66 AAD55392 1. 0E-18 [Xenopus laevis] 322 C610457.1 hypotheticalprotein FLJ20442 [Homo sapiens] AAH01140 3. 0E-18 323 C602925.1 CG3192 gene product [Drosophila AAF46185 3. 0E-18 meianogaster] 324 C607025.1 unnamed protein product [Homo sapiens] BAB71083 4. 0E-18 325 C714068. 1 CG8860 gene product [Drosophila AAF58563 7. 0E-18 melanogaster] 326 C606434.1 hypothetical protein [Schizosaccharomyces NP_595698 1. 0E-17 pombe] 327 C606258.1 CG13551-PC [Drosophila melanogaster] AAM68246 1. OE-17 328 C602211. 1 agCP5435 [Anophelesgambiae str. PEST] EAA11710 1. 0E-17 329 C612425.1 RIKEN cDNA 2410003B13 [Mus musculus] XP_124082 1. 0E-17 330 C607105.1 V-ATPase G subunit (Clonorchis sinensis] AAN04090 3. 0E-17 331 C601554.1 signal recognitionparticle 9 kDa [Mus NP036188 3. 0E-17 musculus] 332 C610465.1 prefoldin 4 [Homo sapiens] NP002614 1. 0E-16 333 C606090.1 ubiquinol-cytochrome c reductase complex NP079628 4. 0E-15 ubiquinone-binding protein QP-C; Complex III subunit Vil [Mus musculus] 334 C607598.1 RIKEN cDNA 1110005A23[Mus musculus] NP_079640 5. 0E-15 335 C603891.1 similar toevidence: NAS#hypothetical XP059423 6. 0E-15 protein-putative [Homo sapiens] 336 C603824.1 CG14235-PB [Drosophila melanogaster] AAN09513 1.0E-14 337 C602007.1 SH3 domain bindingglutamic acid-rich protein NP003013 2. 0E-14 like ; SH3-binding domain glutamic acid-rich protein like [Homo sapiens] 338 C606704.1 hypothetica ! proteinFLJ10511 [Homo sapiens] NP060590 2. 0E-14 339 C605235.1 mitochondrialribosomal protein S21 [Mus NP_510964 4. 0E-14 musculus] 340 C610794.1 agCP9394 [Anophelesgambiae str. PEST] EAA00178 2. 0E-13 341 C605957.1 nuclear DNA-bindingprotein [Homo sapiens] NP006324 3. 0E-13 342 C612322.1 RNA polymerases I, lland III shared NP501593 3. 0E-13 polypeptide (approx 7kd [Caenorhabditis Elegans] 343 C603923.1 GM16138p [Drosophila melanogaster] AAM50712 4. 0E-13 344 C607486.1 CG32069-PA [Drosophila melanogaster] AAF50111 2. 0E-12 345 C610946.1 calcitonin gene-relatedpeptide-receptor NP055293 2. 0E-11 component protein [Homo sapiens] 346 C606849.1 putative vacuolar ATPase membrane sector CAD21533 2. 0E-11 associated protein [Taenia solium] 347 C605796.1 RE27904p [Drosophila melanogaster] AAL48861 3. 0E-10 348 C610040.1 RIKEN cDNA 1700127H04 [Mus musculus] NP-598458 3. 0E-10 349 C610531.1 G protein gamma 1 [Drosophila melanogaster] NP_523662 3. 0E-10 350 C612149.1 Taf24-P1 [Drosophila melanogaster] NP_477463 4. 0E-10 351 C601059.1 fast skeletal troponin C beta [Xenopus laevis] BAA19735 7. 0E-10 352 C600131.1 hypothetical proteinMGC14353 [Homo NP_116120 5. 0E-09 sapiens] 353 C602539.1 similar to RIKENcDNA 1110001J03 gene AAH08467 5. 0E-09 [Homo sapiens] 354 C604199.1 Jra-P1 ; lethal (2 IA109 ; Jun oncogene Nu-476586 2. 0E-08 [Drosophila melanogaster] 355 C608275.1 hypothetical proteinMGC14126 [Homo NP_116287 4. 0E-08 sapiens] 356 C610132.1 anon2C9 [Drosophilayakuba] AAB81489 2. 0E-07 357 C603164.1 unknown [Clonorchis sinensis] AAM55183 8. 0E-07 358 C712405.1 profilin I [Dictyostelium discoideum] P26199 3. 0E-06 359 C608339.1 hypothetical proteinMGC14697 [Homo Nu_116136 4. 0E-06 sapiens] 360 C715968.1 RIKEN cDNA 1110003H18 [Mus musculus] XP_131076 9. 0E-06 EXAMPLE 3 [0039] Following the experiments described supra, putative protein products were assigned to SmAEs, based on BLASTX hits to the publicly available NCBI NR database. In accordance with Ashburner, et al., Nat. Genet., 25: 25-29 (2000), incorporated by reference, gene ontology terms were assigned to the SmAEs, based on BLASTX hits against a database that had been locally built from public sequences, associated with GO terms. Public GO annotated datasets were available for H. sapiens, D. melanogaster, A. thaliana, O. sativa, C. elegans, S. cervisiae, S. pombe, and V. cholera, plus a curated sequence database, at www. geneontolog. org.. The BLASTX e- value cut off for this work was 10-6. The deduced amino acid sequences were in accordance with Iseli, et al., Proc. Int. Conf. Intell. Syst. Mol. Biol., 138-148 (1999), and were used as queries against Pfam database 7.8.

[0040] A total of 8,001 SmAEs received GO classifications. SmAEs were distributed amongst various families, as presented below, and summarized in Table 3, which follows.

[0041] Protein metabolism was the most abundant biological process identified from this analysis. Protein kinases were most abundant when conserved domains were searched (180), suggesting a more compact set of protein kinases than any other fully sequenced metazoa.

[0042] Most of the top 15 Pfam domains are from proteins that are involved in either intracellular communication, or transcriptional regulation processes. For a parasite with multiple tissues and organs, this is to be expected.

TABLE 3 Biological process (5,463 distinct SmAEs) Distinct % SmAEs in Category SmAEs in each sub- each sub-category category 1) Cell communication 1. a) Cell adhesion 127 1.3 1. b) Signal transduction 535 5.6 1. c) Response to external stimulus 166 1.7 1. d) Cell communication others 74 0.8 2) Development 2. a) Reproduction 141 1.5 2. b) Morphogenesis 162 1.7 2. c) Development others 244 2.6 3) Cell growth and/or maintenance 3. a) Transport 875 9.2 3. b) Cell cycle 293 3.1 3. c) Metabolism 3. c. 1) Carbohydrate metabolism 258 2.7 3. c. 2) Energy pathways + electron transport 315 3. 3 3. c. 3) Nucleobase, nucleoside, nucleotide and nucleic 1,174 12. 4 acid metabolism 106 1.1 3. c. 4) Amino acid and derivative metabolism 155 1.6 3. c. 5) Lipid metabolism 464 4.9 3. c. 6) Phosphorus metabolism 478 5.0 3. c. 7) Catabolism 828 8. 7 3. c. 8) Biosynthesis 1,513 15.9 3. c. 9) Protein metabolism 586 6.2 3. c. 10) Metabolism others 448 4.7 3. d) Cell organization and biogenesis 327 3.4 3. e) Cell growth and/or maintenance others 228 2.4 4) Biological process others 9,497 100.00 Total SmAEs in Biological process categories Cellular component (4,329 distinct SmAEs) Distinct % SmAE in Category SmAEs in each sub- each sub-category category 1) Cell 4,043 91.7 2) Extracellular 201,40 4.5 3) External protective 3 0.9 4) Obsolete 120 0.0 5) Unlocalized 4,407 2.7 Total SmAEs in Cellular component categories 100. 0 Molecular function (6,577 distinct SmAEs) Distinct % SmAEs in Category SmAEs in each sub- each sub-category category 1) Transcription regulator 267 2.4 2) Chaperone 212 1.9 3) Motor 159 1.4 4) Enzyme 4. a) Kinase 549 5.0 4. b) Phosphatase 147 1.3 4. c) Oxidoreductase 340 3.1 4. d) Transferase 1,013 9.2 4. e) Hydrolase 1,357 12.3 4. f) Enzyme others 877 8.0 5) Translation regulator 160 1.5 6) Signal transducer 515 4.7 7) Structural molecule 502 4.6 8) Transporter 867 7.9 9) Binding 3,625 33.0 10) Molecular function unknown 107 1.0 11) Molecular function others 304 2.8 Total SmAEs in Molecular function categories 11, 001 100. 0 EXAMPLE 4 [0043] Phylogenic analysis of the data reported supra was carried out, using unambiguous sequence information for the deduced amino acid sequences and the Clustal X multiple sequence alignment program. In addition, phylogenetic trees were constructed using the Phylip program version 3.5, described by Felsenstein, et al., Cladistics, 5: 164-166 (1989), incorporated by reference. Parsimony analysis was carried out using Protpars, and distance calculations were performed using the neighbor-joining method of the Phylips program described supra. Bootstrap analysis used 1,000 iterations.

The commercially available Treeview program was used to print the trees.

[0044] A total of 55% of the SmAEs showed no significant matches to sequences in Genbank, which is a high fraction. These results support the proposal of e. g., Hausdorf, Syst. Biol 49 : 130-142 (2000), that the platyhelminth acoelomates like S. mansoni diverged from other eubilaterian metazoa more than a billion years ago, and lie somewhere between unieukaryotes, like S. cervisiae and P. falciparum and more advanced invertebrates like C. eleaans, D. melanogaster and C. intestinalis.

[0045] In a follow up to this, SmAEs which code for proteins conserved amongst eukarya or metazoa were selected by comparing the SmAEs with known proteomes of completely sequenced organisms. Those SmAEs which had orthologs with H. sapiens, D. melanogaster, C. elegans and C. intestinalis (all multicellular eukaryotes) but none with S. cervisiae or P. falciparum (unicellular eukoryotes), or any prokaryotes, constituted a dataset of 1598 sequences, or about 645 genes.

[0046] Eukarya conserved sequences were those which had at least one ortholog in all of the listed eukaryotes. This set contains 3,194 SmAEs, constituting about 1443 genes.

[0047] These data sets represent, respectively, genes important for metazoan cell function and eukaryotic cell function.

EXAMPLE 5 [0048] These experiments were designed to study differential expression of genes in different life stages of the parasite. Frequency of reads within SmAE clusters, when obtained from different life cycle stages, can reflect differential gene expression, when the same set of primers is used to generate mini-libraries across stages.

[0049] ORESTES sequences obtained from 6 primers, which had been used along all six life cycle stages were assembled. The number of reads per stage for each cluster was considered an indirect inference of expression levels within stages. The results were validated experimentally via RT-PCR. In brief, reverse transcription was carried out using standard, commercially available reagents, together with 300 mg of cercariae, miracidial, or egg total RNA, and was followed by PCR using 50 ul samples, together with DNA polymerase, 10 pmol of each primer, and a cycling protocol of 94°C, 8 minutes, followed by cycles of 94°C, 1 minute, 55°C, 1 minute, 72°C, 1 minute. For each stage, total cDNA was normalized using actin, which was found to be expressed at comparable levels, in all stages. Following the PCR, a 2.5 ml of each sample was subjected to electrophoresis, on a 1.2% agarose gel, followed by transfer to a nylon membrane. Amplicions were quantified via hybridization of the membrane with a radiolabelled probe, followed by autoradiography.

[0050] A total of 5172 sequences thus obtained were analyzed and generated 2058 SmAEs. Of these, 82 were found to exhibit conspicuously different expression patterns among the stages at a confidence level of 99.8%, following Stekel, et al., Genome Res. , 10: 2055-2061 (2000), incorporated by reference. Several of the genes were predominant in a single stage. For example, in the germ ball stage, overexpression of elastase 2a (a component of cercarial secretions for host invasion) troponin I and tropomyosin 2 (related to development of cercaria muscle cells), centrin3 and S-rex/Nsp, which are related to differentialion and embryogenesis was observed. This highlights complex developmental processes which occur at this stage.

EXAMPLE 6 [0051] These experiments describe the identification of"single nucleotide polymorphisms"or"SNPs"in S. mansoni.

[0052] Identification of polymorphisms was carried out in accordance with Marth Nat. Genet. 23: 452-6 (1999), incorporated by reference, using"Polybayes,"together with filters so as to reduce false polymorphisms resulting from nucleotide misincorporation introduced by Taq polymerase.

[0053] A fraction of the putative SNPs were then selected for experimental validation. This was carried forward by using DNA from pooled adult worms, and first confirming the polymorphisms within the BH isolate, referred to supra, and then to DNA from pooled worms obtained from 10 different regions, including Lebanon, Senegal, Kenya, Mali, Sao Tome, Egypt, and two areas of Brazil (Pernambuco and Bahia). PCR primers were used which flank the putative polymorphism site, and the DNA was amplified using proof reading DNA polymerases. PCR products were cloned, and at least 30 alleles were sequenced for each isolate. A total of 601 alleles were amplified, from the 10 regions referred to supra. Within the BH isolate, polymorphisms were found in, e. g., paramyosin, GST28 and TPI. These 3 molecules exhibited 8 polymorphisms, 5 of which were non-synonymous. GST28 was most polymorphic, exhibiting 5 of the 8 polymorphisms.

[0054] The sampling from the various regions confirmed the TPI SNP, and one of the GST 28 SNPs. Specifically, in GST28, Asp61 was replaced by Asn. Frequency of the polymorphic allele ranges from 6.6% in BH, to 93.4% in Bahia strain. In the TPI SNP, Amg 85 was replaced by Lys, with a frequency varying from 24 to 42% in two, distinct, Senegalese isolates.

EXAMPLE 7 [0055] The material which follows relates to analysis of the data developed in the examples set forth supra.

[0056] Distribution of SmAEs in GO categories was described, supra. When the relative distributions in the eukarya conserved and metazoa specific databases were compared, it was noted that the latter category contained a higher proportion of sequences in several categories of proteins, including those involved in cell to cell interaction for multicellular organisms (e. g., cell adhesion), developmental processes such as morphogenesis, including muscle and nerve generation, as well as embryo formation (including homeobox orthologs) responses to external stimuli, and signal transduction, including receptors believed to be involved in tissue signaling.

[0057] The distribution of sequences is emblematic of metazoa. For example, intercellular adhesion was undoubtedly the most basic feature required for metazoan evolution. As triploblastic acoelomates with three germ layers, bilateral symmetry and dorso-ventral patterning, schistosomes stand on the threshold of organization beyond mere tissues, into organs such as gut, nephridia, nervous system and muscle. The SmAE dataset permits evaluation of the molecular basis of the structural organization of these acoelomates. Schistosomes possess at least two protocadherins, which usually function in homotypic cell adhesion. More localized regions of cell contact, exemplified by adherins junctions, are indicated by the presence of 3 catenin, vinculin and VASP homology protein (homer), to link the cytoplasmic portion of the protocadherins with actin in the cytoskeleton. The small G proteins Rho, Ras and Rac plus afadin, involved in actin polymerization, are all present. The existence of organized tight junctions, which would be important in maintaining the integrity of gastrodermal and protonephridial epithelia, can be inferred from the occurrence of transcripts for OapIlTspan3, and several Maguk orthologs, including Zo2. Evidence for gap junctions is provided by two pannexins/innexins; these form channels to assist the integration of cell metabolism in both neuronal and non-neuronal tissues.

[0058] The extracellular matrix is represented by at least 10 collages, 4 laminins and X and C forms of tenascin, to which cell attachment may be facilitated by V3 and 32 integrins, that could potentially heterodimerize. The intracellular links (talin, focal adhesion kinase, vinculin and actinin) between integrins and the actin cytoskeleton are also evident.

[0059] The ability to undergo remodelling is a feature of organized tissues, but evidence for apoptosis in S. mansoni is fragmentary. Four transcripts with homology to caspases were found, plus orthologs of two death-associated protein kinases, the apoptosis-inducing factor (AIF), Bcl-2-interacting protein (beclin-1) and Bax inhibitor; however, other components such as Bax, the Bcl-2 family or endonuclease G were not evident. Flatworms do not possess wandering phagocytes to engulf apoptotic corpses and therefore need other ways of eliminating redundant cells. In this context, mechanisms of autophagy were revealed by Apg proteins 2-9 and 16, many Apg1p orthologs, and the autophagins Aut-1, -2, and-3. Gene fragments of Tor (Target of rapamycin, which phosphorylates Apg13p, inhibiting activation of Apg1p) were identified, but not Apg13p itself, nor Apg12p, the initiator of autophagy. The almost complete autophagy pathway concurs with ultrastructural studies on irradiated schistosomes, as described by Mastin, et al., Parasitology, 91: 101-110 (1985).

[0060] S. mansoni has several axis-determining components in common with other metazoa. The presence of nanos, pumilio and the knirps gap-gene strongly suggests parallels with the mechanisms in Drosophila, described by Lall, et al., Curr. Biol., 13: 224-229 (2003), in which maternal factors segregating to one pole of the egg determine the antero-posterior axis. The polycomb group transcripts, enhancer of zeste, polyhomeotic distal and extra sex combs, which are responsible for the maintenance of pattern, were detected in the SmAEs, but none of the archetypal Hox cluster sequences were. Orthologs of putative S. mansoni homeotic transcription factors included LIM- homeodomain, double homeobox protein 4 and homeotic protein Msx1 were observed.

[0061] Dorso-ventral patterning may be dictated by an analogue of the TGF-3 pathway. Activin/TGF-3 receptor (but not TGF-3) orthologs were identified, together with Smad 4, and Smad 8 and Medea, plus Smad 1 and Smad 2 as described by Osman, et al., J. Biol. Chem., 276: 10072-10082 (2001). The R-Smads (Smad 1, Smad 2 and Smad 8) are anchored to the plasma membrane by SARA, also newly identified. Specification of the dorso-ventral axis may also involve the Wnt pathway; two Wnts and frizzled, their transmembrane receptor, were identified, plus the cytosolic components of the intracellular signaling cascade, disheveled, axin, Gsk3 and 3-catenin.

[0062] S. mansoni adult schistosomes have three epithelia, the surface tegument, gastrodermis and protonephridial canals, which control the transport of material into and out of their bodies. Transcripts of villin family members, supervillin and archvillin were found, and may cap and bundle actin filaments to provide an internal scaffold for cellular extensions cross-braced at their base by spectrin, which was also present.

Functional studies have revealed mediated transport of sugars, amino acids and nucleotides (Pappas, et al., Exp. Parasitol., 37: 469-530 (1975) ). At least 9 SmAEs for sugar transporters (some ATP-driven) can be added to the already cloned Sgtp1, 2 and 4, taught by Skelly, et al., J. Biol. Chem., 269: 4247-4253 (1994). Several transporters for lipids, amino acids, nucleotides and ions were identified.

[0063] Endocytosis is prominent in the gastrodermis but caveolin-type lipid rafts have also been postulated as being present in the tegument surface. See Racoosin, et al., Mol. Biochem. Parasitol., 104: 285-297 (1999). Caveolin transcripts were not identified although the raft-associated flotillin was found. Paradoxically, transcripts for many components of clathrin-mediated endocytosis, described by Conner, et al., Nature, 422: 37-44 (2000), not previously reported in schistosomes, were conspicuous. These included the clathrin heavy chain, the assembly protein Ap180, and the adaptor complex Ap2, which together encode all the functions to select cargo and form a vesicle. Dynamin, the master regulator of endocytosis was present, along with phospholipid-interacting endophilin, Eps15 and epsin. In addition to the previously characterized GPI-linked low- molecular weight LDL binding proteins, transcripts for serotransferrin, LDL and VLDL receptors attest to the importance of receptor-mediated endocytosis in schistosome nutrition.

[0064] All life cycle stages of S. mansoni possess an extensive and intricately organized musculature comprised of smooth fibers, (Mair, et al., Prasitology, 121: 163-170 (2000) ), whilst only the rapidly oscillating cercarial tail has a form of striated muscle (Nuttman, Parasitol, 68: 147-154 (1974) ). Transcripts were obtained for several myosins, two actins, plus tropomyosin, paramyosin and troponins C, I and T, involved in the regulation of contraction, besides the filament attachment proteins, V-actinin, vinculin and titin, many of which are novel paralogs. Germ ball/cercaria-specific transcripts encoding striated muscle proteins were not evident.

[0065] Platyhelminths are the first metazoan group to possess a central nervous system (Halton, et al., Parasitol, 113: 547-572 (1996)), whilst a variety of sensory structures (Dorsey, et al., Micron., 33: 279-323 (2002) ) transduce a wide range of stimuli.

Notch signaling is involved in neurogenesis; Notch receptor, its transcription factor partner (suppressor of hairless), and its membrane-bound ligand, delta, suggest a role for this pathway in S. mansoni. The identification of transcripts for axon guidance molecules to direct nerves to their synaptic partners, such as netrin and its membrane receptor Unc5, two semaphorin-like and two plexin-like molecules, all document the molecular repertoire for a sophisticated neural circuitry. With respect to sensory structures, components of the light detection system (rhodopsin, rhodopsin kinase, arrestin and transducin) were identified, the first two in eggs and germ balls respectively, consistent with the responsiveness of miracidia and cercariae to light. Interestingly, the rhodopsin transcript is a paralog of previously described by Santos, Parasitol., 103: 79-97 (1999), and Hoffmann, et al., Mol. Biochem. Parasitol., 112: 113-123 (2001).

[0066] Knowledge of schistosome neurotransmitter/receptor systems derived from cytochemistry has now been given a molecular basis by transcriptome analysis.

Examples of ligand-gated channels present in the SmAEs include three versions of the nicotinic Acetylcholine (Ach) receptor, as well as choline o-acetyltransferase for synthesis, and Ach esterase for breakdown of this inhibitory neurotransmitter. A glutamate receptor was also detected, plus transcripts for the (-amino butyric acid (GABA) transporter and GABA receptor-associated protein, but not the inhibitory GABA receptor itself.

[0067] G-protein-coupled receptors for glutamate and the excitatory transmitter serotonin, plus its transporter, were found, as well as a putative muscarinic Ach receptor.

While catecholamine responsiveness has been reported (Pax, et al., Parasitol., 102 Suppl. S31-9 (1991) ), no transcripts for the relevant receptors were evident. Primitive neuroendocrine processes are known to be mediated by FaRP (FMRP amide-like) type peptides (Smart, et al., J. Comp. Neuro., 347: 426-432 (1994) ), but only a transcript for allatostatin precursor protein was found. Nevertheless, orthologs of hormone proprotein convertase 2 (PC2), which processes the precursors of bioactive peptides, and its regulatory neuroendocrine protein 7B2, were present, as well as glycine peptidyl V-amide monooxygenase, required for the C-terminal amidation of the resulting peptides. PC2 generates the opioid peptides and enkephalin in higher animals, and might have the same function in schistosomes since these peptides have previously been reported by Pryor, et al., Acta. Biol. Hung., 51: 331-341 (2000).

[0068] It is difficult to envisage how hormone signaling might operate in coelomate animals, except over a short distance or via the neuroendocrine route.

Nevertheless, two members of the nuclear receptor superfamily (retinoid-X and fushi tarazu factor 1) have been characterized by deMendonca, et al., Parasitol. Today, 16: 233- 240 (2000), and SmAEs for a retinoic acid receptor (RAR- (), a thyroid hormone (TH) receptor family member, a nuclear receptor 1 and a nuclear orphan receptor Tr2/4 have been identified. The absence of ligands for known receptors led to speculation that ligand-binding domains are a later evolutionary addition, and characterization of the molecules as orphan receptors; however, detection of transcripts for TH interactor proteins 4,12, 13 and 15 and TH receptor-associated proteins Trap240 and Trap80, together with the reported effect of TH on schistosome development (Saule, et al., J.

Parasitol., 88: 849-855 (2002), suggest that at least one nuclear receptor may have a functional ligand. An ortholog of thyroid peroxidase, required to synthesize TH, is present, but not thyroglobulin, its vertebrate substrate. If there is endogenous TH, perhaps S. mansoni uses an alternative tyrosine-rich protein as a precursor.

[0069] The possibility that schistosomes synthesize steroid hormones from cholesterol is suggested by the presence of transcripts for a series of cytochrome P450 enzymes, testosterone 6-beta-hydroxylase and 17b-hydroxysteroid dehydrogenase, which catalyses the final step in testosterone biosynthesis. Schistosomes also appear to have some receptor elements (progesterone receptor membrane component 2 and estrogen- related receptor), which could bind endogenous steroids, or mediate the supposed action of exogenous steroids on their maturation. Other receptors identified for insulin and FGF but not their ligands, reinforce the concept that host molecules act on parasite receptors.

The presence of SmAEs encoding neurotensin and natriuretic peptide receptors is intriguing but more difficult to place in context.

[0070] Most platyhelminths are hermaphroditic, but sexual dimorphism has apparently evolved separately on at least 8 occasions, arguing for a relatively simple underlying mechanism. See Snyder, et al., The Interrelationships of Platyhelminthes, (Taylor & Frances, 2002), pgs. 194-199. Determination of sex is inherent, while envelopment by the male is a prerequisite for female maturation, (Basch, Compl.

Biochem. Physio. B. , 990: 389-392 (1988) ), revealing the need for"cross-talk". Orthologs of Fox1, Mog1, Mog4, Tra2 and Fem1, involved in the determination of sex in C. elegans, were detected. The ortholog of mago-nashi, which in C. elegans (mag1) specifies female development by inhibiting the hermaphrodite phenotype, was also obtained. The presence of the above transcripts in S. mansoni confirms their evolutionary ancient role in sex determination, but the way they contribute to the dioecious state is unclear.

[0071] Schistosomes have a prolonged association with their hosts, and they should possess distinct features that represent specific adaptations to the parasitic way of life. Adult worms are bathed in, and feed on host blood. Transcripts were found for echicetin-like molecules that affect haemostasis and prevent thrombosis, probably by inhibiting platelet aggregation and activation through glycoprotein IB. Adults expressed apyrase (CD39/ATP-diphosphohydrolase), an enzyme involved in platelet aggregation and thromboregulation that has been localized to the tegument (de Marco, et al., Biochem. Biophys. Res. Commun., 307: 831-838 (2003), possibly indicating the capacity to inhibit platelet activation. Such anticoagulant properties are probably essential for parasite survival in the definitive host.

[0072] In contrast to the lifespan of 10 days for C. elegans, or a few weeks for Drosophila, the longevity of schistosomes is remarkable, with dynamic models predicting from 6 to 10 years (Fulford, et al., Parasitology, 110 (Pt3): 307-316 (1995). In yeast and C. elegans, an extra copy of the Sir2/Sir2. 1 genes, implicated in chromatin silencing through histone deacetylase activity, can increase lifespan, and orthologs to Sir2. 1, Sir2. 2, Sir2. 5, Sir2. 6 and Sir2. 7 were identified in S. mansoni. SmAEs from the insulin-signaling pathway, associated with longevity in C. elegans, were identified, including Daf2, an insulin-like receptor, Age1, a phosphatidylinositol-3-OH kinase and Daf16. This last is a transcription factor regulating many genes that affect lifespan including enzymes that protect against or repair oxidative damage. See, e. g. , Murphy, et al., Nature, 424: 277-283 (2003). In addition, Pdk1 and PTEN, proteins that regulate the Daf2 pathway, were identified.

[0073] The rapid transitions between different environments experienced by S. mansoni are accompanied by temperature and osmotic stresses. The list of previously described heat shock genes was extended (23 SmAEs, 12 possibly new), including an HtrA ortholog, a stress-regulated serine protease. Uroplakin is believed to limit the permeability of membranes to water and small non-electrolytes (Hu, et al., Am. J. Physiol.

Renal. Physio, 283: F1200-1207 (2002), and an ortholog was found in egg, miracidium and cercaria stages. Parasites also encounter oxidative stress during host immune attack which is dealt with by antioxidant enzymes, both previously characterized (superoxide dismutases, thioredoxin and glutathione reductases and peroxidases) and novel, the latter including mitochondrial thioredoxin 2 (Mt-TRX), a PKC-interacting thioredoxin, a thioredoxin-like 2, an ortholog of Plasmodium yoelEi thioredoxin, and glutaredoxin 3 (GRX- 3).

[0074] The innate immune response comprises primitive mechanisms used by metazoa in defence against infection. See Dehal, et al., Science, 298: 2157-2167 (2002), The C. elegans Sequencing Consortium Science, 282: 2012-2018 (2002). The Toll pathway plays an important role and several components were identified, such as Toll interacting protein (Tollip), pellino and NF-kB kinase (NEMO), implying the capacity to respond to extracellular pathogens. The presence of transcripts for adenosine deaminase, Dicer and Piwi/argonaute indicates that S. mansoni can also deal with intracellular attack mediated by viral dsRNA. By extension, the last two genes indicate that post-transcriptional gene silencing could occur, and the use of RNA interference to suppress schistosome gene function was recently reported. Skelig, et al., Int J.

Parasitol., 33: 363-369 (2003); and Boyle, et al., Mol. Biochem. Parasitol., 128: 205-215 (2003).

[0075] Strategies proposed for the parasite to evade host immune responses include protection of the tegument surface by a secreted membranocalyx (Wilson, et al., Parasitol., 74: 61-71 (1977)), molecular mimicry, antigenic variation and immunomodulation. As an example of molecular mimicry, the convergent evolution of S. mansoni and Biomphalaria glabrata (snail intermediate host) tropomyosins (TPM) 1 and 2, has been suggested by Dissous, et al., Parasitol Today, 11: 45-46 (1995) on the basis of immunological cross-reactivity and amino acid sequence identity (-63%). A new isoform, TMP3, was detected in adults, eggs and germ balls, with only 35% amino acid identity to B. glabrata, suggesting a different tissue location not subjected to the same selective pressure.

[0076] In the context of antigenic variation, no evidence of highly variable gene families was found (such as var, rif and stevor in Plasmodium), but analysis revealed 449 putative novel paralogs to known S. mansoni genes. Further, 33 paralogs with high identity and >30% coverage are listed in Table 4.

TABLE 4 [0077] This multiplicity of isoforms would allow the parasite to use paralogs of an # a mnsonæ genB SiSE Gene Accessiai % c§lFew9 ifl e Uffi S. mans-enigene 1 ! 14-3-aprstBinCEOSS61. 1AAC4@a83,. 11. 0E-71 CB10694. 1 AAS468S3. 1 36. 1 S. OE-3B -ST131B5. 1AaS4BS33. 1 35. 73. 0E-43. 2 actin CE04453. 1 &aC4SSE@. 1 1BQ. O 0 C7l7S25. 1 AAS4Sgas. 1 B@. S S. QE-96 0715439. 1 AAC4S9BS. 1 54. S 3. 0E-91 C7Q83SB. 1 AAC4@3B@. 1 53. S 1. QE-78 CE0470E. 9 AAC4@aBE. 1 37. 0 S. QE-4S C7CS ? aa. 1 AAG4S8B6. 1 SQ. 8 2. OE-33 z u 3 Ellphffl tubulin C ; 60w. 1 MA2991S. t 99 6j Q 4 a ! phatubut ! r) GB03SS7. 1 ASSB1S. 1 B 0 CEbi50SB. 1 MA29918. 1 45. 2 tOE-115 Cai03§0. 1 AAASEB13. 1 33. 3 e. OE-SS C6eS6E3. 1 AAA2S61S. 1 37. 7 2. QE-72 C711E. 1 AAA3SS1S. 1 33. 7 5. BE-M C71SB3S. 1 AMSSaiS. t31. SS. BE-SI C ATP : guanidinBhina6etSfne749C712Ma. 1 AMSS7. 131. Ea. QE-2S 8 ea) paninhomo) Eg CBOSMS. I AAB47S3E. 1 99. E T. kE9S s_H cob04248. 1. 647536. 153. 3S. OE-3S 10 eyclopM 7 calhepsin BC7C467S. 1 AAAESSBS. 141. 30. 0E-47 a chanen prntsinC71E9. 1/tAASa6'BS. 133S. QE-17 cl hilin FO,. F.'1 ? 48B1. h 7 2. E-4 10 oophMnACS02142. 1 AAC47317. 1BS. 13. QE-S3 11 e. ydopMnB CT1B072. 1 Aj% 04Sa8S. 1 71. 4 4. BE-48 C601 457. 1 AA046985. 1 OB. 1 1. OE-34 CEOõ340. 1 A-84FSD7. 1 1DO. D 4. 0E29 GBOCa72. 1 AS4T3B7. 1 10S. O 2. 0E-3E Court143. 1 AAC4T3Q7. 1- ! QB. O 1. DUE-39 GOO6345. 1 SC47307. 1 BB. E t. OE-27 CB1M73. 1 AA0473Q7. 1 37. 8 S. OE-34 G7176') S.'t AAC4T. 3Q7. 1 aS. B 1. 0E-a7 CBOSBa. l AM473S7. 1 8S. B 1. BE-33 C7EB41S. 1 AAC47SD7. 1 ß 7. 0E21 CEB3S17. 1 AM473B7. 1 73. 0 S. OE-25 G7CB415. 1 AM473B7. 1 S1. S 7. GE-21 C7E3. S7S. 1 AC473B7. 1SS. 4ME-12 13 ebsteG718@OS. 1 S4e8E7. 145. BS. OE-44 14 ferntin) tght chain CE10B48. 1. AAAZSSS1. 1 88. 4 1. 0E-31 GE0027Q. 1. AAA2SS8Q. 1 T1. 7 S. OE-35 07SDF. t AAA299FO. 1 48. D l. OE21 C7B74S7. 1 A28SB0. 138. 3ME-34 IS giutathicne paroMdassCEniS55. 1 AAG144Ea. 282. 3S. 8E-41 18 heat shade pm) Bin7B B8054T2. 1. AAASSSBa. 1 SS. 3 1. 0E-ia2 CSGTO. I AAASSSaS. 1 SS. 8 0 0 hastshck priain7í3 Cõ0$472. 1. AM29998. 1 g59 tOE-102 a_ m t7 nudeeBidarncnochaschatsMnasa CS'1') a03. 1 AMS07. 134. E5. 0E-14 18 nuelacsidemonophosphata liinm 19 p4a aggahe)) prctainS ? 1igSO.'t MAS§9aa. 173.. aa. OE-35 20 potassium ohannatpmiain S7Q4T47. 1 AC37227. 1 31 J5 1. 0E-7B S7S ! NSS. 1. MC37SS7. 13B. 3a. OE-B6 21 Pernreal CEQB43B. 1 AE0943S. 1 aa. a 3. BE-54 S PemraCBC4S44. 1 AABBS438. 187. 89. 0E-4S 2S Psmr3 C6D7gO2. t AAB09439. 1 B. 4 7. 0ES9 24 Bb-ra) GTP4indingprctain CE0024'i. t 9t3B. 1 9F. t 2. DEo GB1S551. 1 AM. 7913S.-t 53. 7 1. 0E-S7 CE1SS70. 1 A7913S. 151. 7S. BE-SO 25 facsptartbraetiwatadPKS CB12054. 1 A-a4173. 191. 8a. OE-S7 .. C71S34S. 1 AMJS4173. 1S4. 81. 0E-3e 38 similar humancarbnnduose CB12144. 1 AaC468Sa. 1 100. 0 B. BE-S4 (tADPH) CB11SBB. 1 AaC4SB8&. 147. 13.. HE-56 27 simi ! art9mitaahondna ! ATPa8e C60@SS8. 1 Aj% C4@aoo. 1 100. 0 4. 8E-14 inhibitarCB03111. 1MC4S8ao. 135. 2S. OE-13 29 Bimi ! artoBynaptobresin CEOQS1S. 1 AAC4S8S1. 1 BS. 7 . 0&. 33 1555. 9 A69'I. 1 . S : E'I CE1SS5S. 1 AAC46BB1. 14S. SS. QE-16 SmadiCEOSa44. 1AAG35S@E. 162. 8ME-SO 31 Smad1 Coos 1 AA63E2SS. 1 43. 3 ME-58 31 SMQR2 CBOS154. 1 AAA86477. 1 39. 1 1. 0E-S8 33 Y-0DOX bindirg prdbin homolcg cæQass581 MC4n01. 1 S3. 13 1. OE4S SS If-box bind ing prcteinhamoteg CSOSE. 1 AAC4T7B1. 1 SS. B 1. 0E-4a C70B071. 1 AAC477B1. 1S7. 17. 0E-38 essential enzyme targeted by the immune system, to avoid loss of function, thus making vaccine development more difficult. Indeed, several paralogs of previously investigated vaccine candidates were identified. See Table 5.

TABLE 5 Pamsnitrasa (S86aa) SBStM ? {4S2ss-M-} esmina taHbRtarpretan (idenfiveoverage) Far, ya : Ja Pammyosm (Safta) Cpsn (758aa} ptin e CM167. cisi ral prtarr-_ Ci9D {S4%} 14 Catponin {3S1 aa) caseteiScn prntEn ; s% n traasductitm as ntuaets JV% J277as-N-leminall combacean ; hindn to aEn, aEgmadAn, troponin. 13 andl » yuiln. Cl0424@, < [S3%/12C ! sa-0m ! nat} Sm226 (1, Waa) TegumenW pmialn ; mwgked by anesera anl- SS. %. f ISSaa) Bt) S ! <B ! e ! ! iract Snt2&. S-m !) <mtB anSgen {1&1aa} 's tast e@MSM. antgen (191 as) CM-944, 1 (45%) 17&a) Cßy944st {45 CSI2l l2. § {40% f l@ Pa3 SìgnagDs CJ Sing ß irargspoi steiF dcium G6tS2 28% 17S CE 2sj4 % { CT Ei 142% . 1 ia @ '34aear CTMa. 3% 83sa C5<Bt88 eapedsss, ear abc {otmaEan M Ci 1 (45% 1. 100aa) L.. W.- [0078] Non-synonymous single nucleotide polymorphisms (nsSNPs) are another source of variation. Analysis of redundant EST coverage of genes identified 8 putative polymorphisms, two of which were validated in isolates from different regions of the world.

Alternative splicing was detected in several genes, including a recently determined exon skipping in Sm14, (Ramos, et al., J. Biol. Chem., 278: 12745-12751 (2003) ), present in germ balls, schistosomula and adults.

[0079] Modulation of mammalian host immune responses by a schistosome infection is well documented, but the agents and mechanisms remain to be fully defined.

The presence of transcripts for pro-inflammatory phospholipase A2-activating protein in several stages, adds weight to the documented effect of lyso-phosphatidylserine as an inducer of T-regulatory cells and Th2 polarization. See van der Kleis, et al., J. Biol.

Chem, 277: 48122-48129 (2002). It is unclear whether this deliberate Th2 polarization is less deleterious to some stages of the parasite than a Th1 response. S. mansoni infection induces a characteristic allergic response, with increased IgE formation, mediated by allergens from eggs and adult worms. See Damonneville, et al., Int. Arch. Allergy Appl.

Immunol., 73: 248-255 (1984); and Cutts, et al., Parasite Immunol., 19: 91-102 (1997). A family of wasp venom allergen 5 orthologs with 33-38% identity to different wasp species has been identified. Production of such allergens raises the question of what benefits the parasite would gain from amplifying allergic and other inflammatory responses.

[0080] A major benefit from our project should be the identification of novel proteins amenable to rational drug design that could add to the previously defined collection of molecular targets. Effective anthelminthics such as levamisole and pyrantel target nematode nicotinic Ach receptors, and the identification of three novel receptor orthologs (Table 3) should aid the development of new compounds. Piperazine is a GABA receptor inhibitor in nematodes and the avermectins target glutamate-gated channels. The presence of both systems (Table 3), and a serotonin receptor, in S. mansoni provides further new targets.

[0081] Praziquantel is believed to exert its effects via tegument voltage- dependent Ca++ channels; novel paralogs of L-type calcium channel alpha subunit were found, besides several transcripts encoding the previously described channels. The immunosuppressive drug cyclosporin is known to have a pronounced antischistosomal effect and three paralogs of its target, cyclophyllin, were evident. These paralogs of vulnerable proteins may provide new targets. The invertebrate innexins, gap junction proteins involved in embryonic development and in wiring of the nervous system, represent promising new targets (Table 3); they are cell-surface proteins functionally analogous to the vertebrate connexins (proposed as targets in cancer chemotherapy), with a similar predicted secondary structure, but no sequence identity. Another example is DNA polymerase delta (an anti-viral target) which, due to the long evolutionary time since the split from the human ortholog (see Supplementary Fig. 4b), may possess different catalytic and substrate properties.

[0082] Members of the Multidrug Resistance (MDR) family of ABC transporters prevent drug accumulation in humans. As well as transcripts for the previously identified S. mansoni MDR2 (Ref. 54), new paralogs of this transporter were identified plus MDR7 (Table 3). In addition, orthologs of M. musculus MRP2, and also a RND MDR efflux transporter, were found. The existence of these proteins could well complicate the development of new drugs.

EXAMPLE 8 [0083] This example merely describes the nucleic acid molecules and proteins which are the subject of the examples which then follow.

[0084] SEQ ID NO: 2141 consists of 1635 nucleotide bases, encoding an amino acid sequence as set forth in SEQ ID NO: 2142, with 545 predicted amino acids. It is an Apyrase, or ATP-diphosphohydrolase that hydrolyzes ATP, ADP and other nucleoside tri- phosphate substrates and is localized on the tegumental membrane of S. mansoni, according to (DeMarco, et al., Biochem. Biophys. Res. Commun., 307 (4): 831-838 (2003)).

[0085] SEQ ID NO: 2143 is 519 nucleotide bases long, and encodes an amino acid sequence as set forth in SEQ ID NO: 2144, with 173 predicted amino acids. It is an ortholog of LIN-7, putative involved in exocitosis processes of synaptic vesicles and is secreted by schistosomula, but is mainly localized on the tegument membrane of S. mansoni adult worms, according to (Butz, et al., Cell, 94 (6): 773-782 (1998); Harrop, et al., Parasitol., 118 (pt6): 583-594 (1994); Harris, et al., J. Biol. Chem., 277 (38): 34902-34908 (2002)).

[0086] SEQ ID NO: 2145 is 837 bases in length, and encodes an amino acid sequence as set forth in SEQ ID NO: 2146 with 279 predicted amino acids. It is an integral membrane oligosacharyl transferase (STT3) involved in the N-glycosylation of proteins of the endoplasmic reticulum and a source of immunodominant MHC-associated peptides. (McBride, et al., Immunogenetics, 54 (8): 562-569 (2002)).

[0087] SEQ ID NO: 2147 consists of 1077 nucleotide bases which encode an amino acid sequence as set forth in SEQ ID NO: 2148, with 359 predicted amino acids. It is an ortholog of Stonatin, putative involved in lipid transport, mechanoreception or organization of lipid rafts and calcium permeability control (Wang, et al., J. Biol. Chem., 275 (11): 8062-8071 (2000)).

[0088] SEQ ID NO: 2149 is a polynucleotide sequence of 666 bases encoding an amino acid sequence as set forth in SEQ ID NO: 2150, with 222 predicted amino acids.

It is an ortholog of Wasp venom allergen 5, or Antigen 5, putative involved in the regulation of immune response by antibody class switch under control of Antigen 5- specific T-cells. In wasps, Antigen 5 would be an exported protein (Lu, et al., J. Immunol., 150 (7): 2823-2830 (1993)).

[0089] SEQ ID NO: 2151 is a polynucleotide sequence of 375 bases encoding an amino acid sequence as set forth in SEQ ID NO: 2152, with 125 predicted amino acids.

This gene encodes a protein named Dif-5 which shows increased expression in schistosomula in comparison to cercaria of S. mansoni (Verjovski-Almeida, et al., supra).

EXAMPLE 9 [0090] The DNA molecules of the invention were obtained by polymerase chain reaction (PCR) using a cDNA library from adult stage S. mansoni worms as template, and primers capable of annealing to the cDNA sequence of interest as described in Table 5, which follows. The oligonucleotide primers typically contain 18-28 nucleotides. A sequence of four nucleotides, CACC, was introduced in the forward primer to allow a directional cloning in the vector used herein. TABLE 5 shows the primer sequences employed to obtain the target nucleic acid sequences.

TABLE 5 Name Primers | SEQID NO : Apyrase ApyraseF : CACC ATG GTT TTT GGA AAA CGC AA 2153 ApyraseR : TTA TAC CTG ACT AGT TGA TTT 2154 Lin-7 Lin-7F : CACC ATG AGA TGT CCC GAA ATA CAA CC 2155 Lin-7R : TTA ATT AAG TTG TCG ACG TCG 2156 STT3 STT3F : CACC ATG GGG AGT AAT CTG CGT GCA G 2157 STT3R : TTA AAT AGG TGT ATT GAG TCG ACC 2158 Stomatin StomatinF : CACC ATG ATT CGT AGT ATC ATT GG 2159 StomatinR : CTA TTC TTG TTT ATC GCT ATC 2160 Antigen5 Antigen5F : CACC ATG ACT GTG AAA TGT CAG TC 2161 Antigen5R : TTA TCT TTC GTT TGT ATG ATT ACA 2162 Dif-5 Dif-5F : CACC ATG AAA GTA TTG GGA ATT T 2163 Dif-5R : TCA ATA TAT GAA TTT ATT AGT 2164 [0091] Experimental conditions used to obtain the target DNA molecules as reagents, include template DNA (1.0 ng), deoxynucleoside triphosphates, (0.4 mM), an agent for polymerization, such as Taq DNA polymerase or Pfx polymerase (0.5 unit from 1 unit/Ell stock solution), complementary primers as set forth in Table 5 (0. 2 uM) at pH 8.0, in a total volume of 50 pI. PCR was performed in a thermocycler machine by setting the following cycle of temperatures: DNA denaturation at 94°C for 3 minutes, primer-DNA template annealing at 55°C for 20 seconds and nucleotide polymerization at 68°C for 4 minutes. The cycle was repeated 45 times, except that the time for the denaturation step was 20 seconds; after these, the reaction mixture was kept at 68°C for 20 minutes. The primers have sufficient complementarity with the flanking sequences to hybridize therewith and permit amplification of the nucleotide sequence. Typically, one primer is complementary to the negative (-) strand of the template nucleotide sequence and the other is complementary to the positive (+) strand. Annealing the primers to denatured nucleic acid sequence followed by extension with DNA polymerase and nucleotides, results in newly synthesized + and-strands containing the target nucleic acid molecules.

Because these newly synthesized nucleic acids are also templates, repeated cycles of denaturing, primer annealing, and extension resulted in an exponential production of the target defined by the primers.

EXAMPLE 10 [0092] The polynucleotide sequences described supra maybe inserted into a cloning or recombinant expression vector, for example a bacterial plasmid. Such expression vectors contain an origin of replication, a promoter sequence to facilitate the transcription of the inserted genetic sequence, and specific genes to permit the selection of transformed cells. Example of suitable expression vectors are: T7-based vectors for expression in bacteria, such as pRSET and pDEST or vectors for expression in mammalian cells, such as pDEST12.2 (SEQ ID NOS: 2143,2145, and 2147) and pTARGET (SEQ ID NOS: 2141,2149, and 2151), and vectors for expression in live recombinant vaccine carriers, such as Salmonella, Lactobacillus or BCG. A recombinant construct in accordance with the invention includes an isolated nucleic acid molecule encoding a protein, of the invention operatively linked to one or more transcription control and a terminator sequences.

[0093] To elaborate, the DNA sequence of the invention were directional cloned into the commercially available, pENTR TOPO vector. This vector includes a kanamycin resistance gene for E. coli selection, a pUC origin of replication for maintenance of the plasmid in E coli, two attL1 and attL2 sites for further site-specific recombination reaction of the cloned polynucleotide sequences into the expression vector, and a TOPO cloning site for directional cloning of blunt-end PCR products. This specific site has nucleotide sequence CCCTT to which a topoisomerase I binds. The pENTR TOPO vector is not suitable for gene expression since it lacks the T7 promoter sequence of T7 RNA polymerase, an enzyme present in E coli BL21 (DE3).

[0094] The cloning reaction was performed according to the manufacture's instructions wherein 15 ng of the DNA obtained in the PCR reaction and 5 ng of pENTR TOPO vector were combined in 5 mM Tris buffer at pH 7.4 at a total volume of 10 ul. The reaction proceeded for 10 minutes at room temperature (~25°C). Positive clones were selected from Luria Berth agar plates containing 50 lug/ml of kanamycin, as described in Sambrook 1989. Once the nucleotide sequence of the invention was cloned into this pENTR TOPO vector, it was easy transferred to a specific expression vector by reaction at the attR1 and attR2 recombination sites, in the presence of the commercially available LR Clonase enzyme mix. The pDEST12.2 expression vector employed infra is a cytomegalovirus (CMV) -based eukaryotic expression vector for expression in mammalian cells. It is a shuttle-vector, i. e. , it has an origin of replication of E coli, to permit its genetic manipulation and amplification in bacteria prior to injection into mammalian cells.

[0095] For the recombination reaction, 2 ul of the cloning reaction mixture described supra, 15 ng of pDEST12.2 expression vector, and 4 ul of LR Clonase enzyme mix were combined in Tris-EDTA (10 mM Tris-HC1 ; 0.1 mM EDTA) buffer, pH 8.0 for 60 minutes at room temperature (~25°C) ; the enzyme was then inactivated by the addition of proteinase K (4 ug), for 30 minutes at 37°C. Positive clones were selected from Luria Broth agar plates containing 20 ug/ml of ampicillin, as described in Sambrook, supra.

[0096] In an alternative embodiment of the invention, the nucleic acid molecules encoding the proteins of the invention and obtained by PCR methodology were directly into pTARGET vector, a commercially available CMV-based eukaryotic expression vector.

[0097] Nucleotide sequences encoding these proteins can be expressed in other prokaryotes and eukaryotes as well. Hosts can include various microbes, yeast, insect and mammalian cells depending on the expression vector used. Methods that can be used to construct expression vectors containing the protein coding sequences are described in Sambrook supra or according to the other well known methods which it is not necessary to elaborate here.

[0098] The nucleic acid molecules of the invention encoding target proteins can be used to transform bacterial expression hosts, such as E coli, including strain BL21 (DE3). The E coli BL21 (DE3) host has the T7 RNA polymerase gene inserted into its chromosome, under the control of the/acUV5 promoter, which is inducible by IPTG, as described in Methods in Enzymology, vol. 185,1990. Once this bacteria host is transformed with the pDEST17 vector containing the target DNA sequence, IPTG (1mM) can be added to the culture medium. The/acUV5 promoter in the host will direct the transcription of T7 RNA polymerase within the bacteria host, which, in turn, will transcribe the DNA sequence target in the pDEST17, under the control of the T7 promoter. The recombinant proteins can then be purified by metal affinity chromatography or by commonly used chromatographic techniques described in Methods in Enzymology, supra.

EXAMPLE 11 [0099] This example presents data obtained from challenge experiments using DNA vaccines. Each of the 6 different target sequences described supra were used.

Control naive mice were injected intramuscularly on day 1 and day 21 with saline solution, and test mice were injected on day 1 and day 21 with a saline solution containing the DNA vaccine. The mice had been injected with cardiotoxin 5 days prior to day 1 of the immunization protocol. As a control, no cardiotoxin was used and DNA vaccine containing the gene for SEQ ID NO: 2149 in TDM-loaded PLGA microspheres as described by (Lima, et al., Gene Ther., 10 (8): 678-685 (2003) ) was injected intramuscularly on day 1. On day 49 all animals were infected by skin penetration with cercariae and on day 94 the mice were sacrificed, and followed by perfusion of the hepatic portal system and manual recovery of adult S. mansoni worms from the mesenteric vessels. Worm burden of individual mice were recorded as the sum of the worms perfused from the liver and the worms from the mesenteric blood. Vaccine protection was calculated by the formula: [average burden from naive mice] - [average burden from test mice]/ [average burden from naive mice] x 100.

[00100] The results clearly demonstrate that these novel genes or proteins can be used immunologically as vaccine antigens for the parasite S. mansoni. In addition, they could be useful in the diagnosis of schistosomiasis. The purified proteins can be used for the constitution of serological diagnostic kits, such as coating multi-well plates with the recombinant proteins, which can then be used to screen sera of diseased patients by ELISA, or for the formulation of vaccines. Figures 1-6 show the results obtained with each nucleic acid molecule.

EXAMPLE 12 [00101] In this experiment, apyrase protein was expressed in bacteria that were transformed with plasmid vector containing a portion of the apyrase sequence supra as an insert. Specifically, a sequence which encoded amino acids 65-509, was used. The recombinant apyrase protein portion thus obtained contained a hexahistidine tag and it was purified by affinity chromatography with a Nickel column, using standard techniques.

Purified recombinant apyrase was compounded with aluminum hydroxide, using 10 micrograms of protein and 1000 micrograms of aluminum hydroxide. Balb/C female mice (5-6 week-old) were immunized, subcutaneously, with 10 micrograms of recombinant protein per animal. Control naive mice were injected with aluminum hydroxide alone.

Three injections were performed on days 1,7, and 14. On day 70, the animals were challenged by exposure to cercariae, as described supra. On day 119, all animals were sacrificed and the liver was perfused as described supra, in order to recover and count the Schistosoma adult worms: [00102] On days 1,6, 13, and 60 a blood sample was collected from each mouse, the serum was separated and used in an ELISA assay for titration of specific antibodies against recombinant apyrase that had been induced by immunization of the mice. In brief, microtiter plates were covered with apyrase by incubating the plate for 16 hours at 4°C with 0.5 microgram of purified recombinant apyrase per platewell in a solution of 0.05 M carbonate-bicarbonate buffer pH 9.6. Plates were washed 3 times with phosphate saline buffer (PBS) plus 0.05% Tween 20, and blocked by incubation for 1 hour at 37°C with 200 microliters of a solution containing 10% milk in PBS. Plates were washed with PBS plus 0.05% Tween 20, and 100 microliters of diluted serum sample from naive control or from immunized mice were added per well. Serial dilutions of each serum were assayed, starting with a 1: 20 dilution and going to 1: 40,1 : 80 and so on. Plates were incubated with diluted serum at 37°C for 2 hours and washed 3 X with PBS plus 0.05% Tween 20.

Subsequently, 100 microliters of anti-mouse IgG secondary antibody conjugated to horseradish peroxidase was added per well at a dilution of 1: 15000 and incubated for 1 hour at 37°C. Plates were washed 3 X with PBS plus 0.05% Tween 20, and 100 microliters of peroxidase substrate was added per well. Plates were processed in an ELISA Labsystem reader, and absorbance was recorded at 492 nm.

[00103] Interims of naive mice, the worm burden was 3521. 7, whereas the mice which received apyrase injections showed a count of 2818. 2, or a protection rate of 19.05%.

[00104] In terms of immune response, 5 animals which received apyrase developed a high antibody titer, as well as reduced worm burden. Figure 7 shows these results.

[00105] The foregoing disclosure set forth various aspects of the invention, including the isolated, S. mansoni proteins, the amino acid sequences of which are set forth in the listing attached hereto, isolated nucleic acid molecules which encode these proteins such as those set forth herein as well as immunogenic portions of these proteins, and the nucleic acid molecules which encode them,. It will be understood that once an amino acid sequence is known, various degenerate nucleotide sequences can be provided which encode that sequence. All of those are encompassed by this invention.

[00106] "Protein"as used herein, refers to proteins, which are full length, and portions of full length proteins encoded by the listed sequences. With respect to portions of proteins, these may be any sequence of at least 20 concatenated amino acids found in any of the disclosed sequences as long as the amino acid sequence is immunogenic.

The determination of whether or not a peptide is immunogenic is an activity that is well within the skill of an artisan of average abilities.

[00107] The proteins of the invention may be used, alone or in combination with each other, as or as components of immunogenic compositions, such as vaccines.

Further, they can be used as immunogens, so as to generate an antibody response, either alone or when combined with a carrier. The antibodies thus generated can be used, e. g., as diagnostic tools to determine S. mansoni infection or presence, as well as components of vaccines used to generate passive immunity. Proteins and antibodies may be administered"neat"or"compounded"with other standard materials used in preparing immunogenic compositions vaccines, such as carriers, adjuvants, and other materials.

Intravenous formulations are one embodiment and, it will be understood that other formulations of immunogenic compositions, such as vaccines, are possible, including intradermal, subcutaneous, oral, such as sublingual forms, and others. The compositions may be in liquid or"dry"form, such as in lyophilized form. This type of composition is especially suitable when it must be carried"in the field,"and used as a vaccine at some point in time later than when carried.

[00108] The nucleic acid molecules of the invention, as will be understood by the skilled artisan, can be used to produce the proteins described supra, via any of the recombinant methodologies well known to the skilled artisan. They can be placed in, e. g., expression vectors, under control of a promoter or other regulatory element, or they can be used"as is"to transfect or to transform cells. Eukaryotic cells, such as yeast cells, CHO cells, fibroblasts, insect cells, etc. , are among the eukaryotes which can be transformed or transfected. Prokaryotes, such as E. coli or other bacteria can also be used. The choice of host cell will depend upon many factors, including whether or not glycosylation is desired, and to what end the transformant or transfectant will be used.

One way these recombinant cells can be used is as in the form of a cell based vaccine, such as a whole cell vaccine. If the cells are then processed so as to become non-proliferative, the cells present an ideal vaccine, especially if the host cell is one that is not normally the target of immune surveillance in the host.

[00109] Another aspect of the invention is the use of membrane preparations, or cellular"ghosts"of transformants or transfectants. Such approaches have been used with other bacterial species, so preparation of these is well known. Transformants or transfectants which express the proteins of the invention, especially surface proteins can be used to prepare these materials in a fashion taught by the art.

[00110] The proteins have been discussed as vaccines, supra. It is to be understood that the vaccines of the invention can be formulated for any subject. Human vaccines are, of course, included, but so are vaccines for livestock animals, such as sheep, bovine animals, goats, pigs and so forth, and domesticated animals such as pets, confined zoo animals, etcetera. The vaccines may be used prophylactically, e. g. , by administering them prior to possible exposure to S. mansoni, and may also be used post exposure, in order to treat a pre-existing infection.

[00111] As will be recognized by the skilled artisan, the use of the proteins of the invention, or portions thereof, may constitute vaccination, and the protein or portion of the protein may constitute the vaccine. Upon administration to the individual or subject, in any of the ways described supra, an immune response results which protects the individual or subject when confronted with the pathogen. Such an approach, i. e. , the immunization with one or more proteins or portions of proteins, can be used therapeutically or prophylactically. Passive immunization, as described supra, can also be used. In such a case, antibodies are developed against the proteins or protein portions, and via passive transfer serve a role in, e. g. , prophylaxis.

[00112] It is to be understood that, while immunization with the proteins or portions of proteins can serve to stimulate an antibody response, cellular immune responses are also a part of the response of the subject to the immunization. It is well within the skill of the artisan to determine which proteins or portions of proteins function as antibody or cellular immune vaccine agents and that artisan can then formulate, e. g., "cocktails"of appropriate mixes of proteins which have the desired, immune effect.

[00113] In addition to the use of the proteins as vaccines, it is to be understood that the nucleic acid molecules described herein can also be used as vaccines. Via targeted delivery of nucleic acid molecules, one can assure an"in vivo"supply of the desired protein or protein portion molecules at a site of relevance. The artisan of delivery systems, such as liposomes, adenoviruses, retroviruses, and other formats for the delivery of DNA as immunoprophylactic or a therapeutic agents, and all are envisioned as methods for administering the vaccine. It should be kept in mind that the nucleic acid molecules of the invention include those which encode the proteins of the invention, but differ in nucleotide sequence due to codon degeneracy. Indeed, due to patterns of codon usage, which vary from organism to organism, it may be desirable to alter the sequence to maximize expression of the desired vaccine in the treated subject.

[00114] To prepare a vaccine the purified polypeptide can be isolated, lyophilized and/or stabilized, as described supra. The protein may then be adjusted to an appropriate concentration, optionally combined with a suitable vaccine adjuvant, and packaged for use. Suitable adjuvants include but are not limited to: surfactants and pluronic polyols ; polyanions, e. g. , pyran, dextran sulfate, poly IC ; polyacrylics, carbopol ; peptides, e. g., muramyl dipeptide, dimethylglycine, oil emulsions, vitamins, cytokines, hormones, aluminum, calcium salts, and mixtures thereof, bacterial and plant products, e. g., Bacillus Calmette-Guerin (BCG), complete Freund's adjuvant, and threalose. Alternatively, the immunogenic protein may be incorporated into liposomes for use in a vaccine formulation, may be fused to other immunogenic proteins, or may be conjugated to polysaccharides or other polymers. See Edelman, in New Generation Vaccines (Marshall Decker, N. Y.

1997), incorporated by reference.

[00115] The weight of the immunogenic protein included in a given dosage of vaccine can vary widely, e. g. , from 5 ug-300 mg. , and depends on: the age, weight, and physical condition of the animal or the human subject considered for vaccination.

[00116] In addition, the nucleic acid molecules of the invention can be used as vaccines. The rational of this approach is that the DNA will produce the protein following injection thus in turn inducing the desired immune response.

[00117] For such vaccines, a pharmaceutical composition can include either a mammalian recombinant expression vector, such as TARGET, or a construct such as an expression vector, which includes a nucleic acid molecule encoding the protein, operatively linked to transcription control/terminator sequences, combined with a pharmaceutical acceptable carrier. These may include, but are not limited to, aqueous physiologically balanced solutions, artificial lipid-containing substrates, natural lipid- containing substrates, oils, esters, and glycols. Pharmaceutically acceptable carriers can also include a suitable delivery vehicle, such as liposomes, micelles, and cells. Adjuvants for DNA based vaccines can be used, such as CpG oligonucleotides and cytonkines. The vaccines can also be delivered by attenuated bacteria, such as Salmonella.

[00118] Another feature of the invention is the use of the proteins, or portions of the proteins of the invention, as well as nucleic acid molecules and portions of nucleic acid molecules, in the manufacture of kits useful for diagnosis of Leptospira infection, either"in the field"or in the laboratory. Such kits involve, e. g. , the protein, protein portion, nucleic acid molecule, or nucleic acid molecule portion, in combination with, e. g. , a solid phase, such as a bead, mutltiwell plate, etc. , to which the component is affixed. The kit can then be used by contacting a sample of interest thereto, followed by a second component, which can also be included in the kit, such as a labeled protein or proteins portion, or labeled nucleic acid molecule or portion of a nucleic acid molecule. When nucleic acid molecules are used in the diagnostic kits of the invention, it is desirable that each portion hybridize to a separate portion of a target nucleic acid molecule.

[00119] It may be desirable to screen the proteins and nucleic acid molecules of the invention in, e. g. , and animal or cellular model prior to administration to large animals or humans. Standard practice tests a potential vaccine in, e. g. , a hamster, mouse, rat or other rodent model to determine it efficacy and strength, and the molecules of this invention may be so tested as well. Testing of the DNA molecules in, e. g., microorganisms such as E. coli or other prokaryotes or eukaryote organisms can be carried out to deter mine which are, in fact, good producers, i. e., molecules which produce high yields, and/or produce transformants which react well to culture.

[00120] Another aspect of the invention are computer readable media which have recorded thereon all or a part of the nucleotide sequences, or degenerate variants thereof.

Exemplary of such computer readable media are floppy discs, hard discs, random access memory (RAM), read only memory (ROM), and CD-ROMs. Such computer readable media are useful, e. g. , in identifying whether a nucleic acid molecule of interest is from, or is homologous to, an S. mansoni nucleic acid molecule. Thus, they permit the skilled artisan to determine if a subject, such as a human is infected with S. mansoni, by analyzing, e. g. , a blood, serum, or other body fluid sample.

[00121] Other aspects of the invention will be clear to the skilled artisan and need not be set forth herein.

[00122] The terms and expressions which have been employed herein are used as terms of description and not of limitation, and there is no intention in the use of such terms and expressions of excluding any equivalents of the features shown and described or portions thereof, it being recognized that various modifications are possible within the scope of the invention.

[00123] For the experiment of Figure 7, Apyrase protein was expressed in bacteria that were transformed with plasmid vector containing Apyrase DNA Sequence ID 1 as an insert. This recombinant Apyrase protein thus obtained contained a hexa-histidine tag and it was purified by affinity chromatography with a Nickel column. Purified recombinant Apyrase was formulated with aluminum hydroxide, using 10 micrograms of protein and 1000 micrograms of aluminum hydroxide. Balb/C female mice (5-6 week-old) were immunized subcutaneously with 10 micrograms of recombinant protein per animal.

Control naive mice were injected with aluminum hydroxide alone. Three injections were performed, on days 1,7 and 14. On day 70 the animals were challenged by exposure to cercariae, as described elsewhere in this patent application. On day 119 all animals were sacrificed and the liver was perfused as described elsewhere in this patent application, in order to recover and count the Schistosoma adult worms.

[00124] On days 1,6, 13 and 60 a blood sample was collected from each mouse, the serum was separated and used in an ELISA assay for titration of specific antibodies against recombinant Apyrase that were induced by immunization of the mice. In brief, microtiter plates were covered with Apyrase by incubating the plate for 16 hours at 4 °C with 0.5 microgram of purified recombinant Apyrase per plate-well in a solution of 0.05 M carbonate-bicarbonate buffer pH 9.6. Plates were washed 3 times with phosphate saline buffer (PBS) plus 0.05% Tween 20, and blocked by incubation for 1 hour at 37°C with 200 microliters of a solution containing 10% milk in PBS. Plates were washed with PBS plus 0.05% Tween 20, and 100 microliters of diluted serum sample from naive control or from immunized mice were added per well. Serial dilutions of each serum were assayed, starting with a 1: 20 dilution and going to 1: 40,1 : 80 and so on. Plates were incubated with diluted serum at 37°C for 2 hours and washed 3 X with PBS plus 0.05% Tween 20.

Subsequently, 100 microliters of anti-mouse IgG secondary antibody conjugated to horseradish peroxidase was added per well at a dilution of 1: 15000 and incubated for 1 hour at 37 °C. Plates were washed 3 X with PBS plus 0.05% Tween 20, and 100 microliters of peroxidase substrate was added per well. Plates were processed in an ELISA Labsystem reader, and absorbance was recorded at 492 nm.