VIROLOGY

190,

527-530

Herpesvirus

(1992)

Saimiri Has a Gene Specifying

a Homologue

J.-C. ALBRECHT,*~’ J. NiCHotAS,t~2 K. R. CAMERON,t *Institut

fijr Klinische und Molekulare Virologie, Germany; and tNational Institute

of the Cellular Membrane

C. NEWMAN,t

B. FLECKENSTEIN,*

Glycoprotein

AND R. W. HONES&

Friedrich-Alexander Universita’t Erlangen-NUmberg, Loschgestrasse 7, 8520 for Medical Research, The Ridgeway, Mill Hill, London NW7 1AA. U.K.

Received

April

15, 1992;

accepted

May

26,

CD59

Erlangen,

1992

Herpesvirus saimiri (HVS) is a T-lymphotropic tumor virus that causes fulminant lymphomas and leukemias in various New World primates other than its natural host, the squirrel monkey (Saimirisciureus). In the course of completing the nucleotide sequence of its genome, we identified an open reading frame of 363 nucleotides, designated HVS-15, that has no detectable homology to any other viral sequences to date. HVS-15 encodes a 121 -amino-acid protein which shows significant similarities to human CD59, a phosphatidyl-inositol-glycan-anchored glycoprotein involved in T-cell activation and restriction of complement-mediated lysis. The predicted HVS-15 gene product is more similar to human CD59 than to the related murine Ly-6 antigens. A nucleotide sequence identity of 64% was found between HVS-15 and the CD59 reading frame, and a 48% identity exists between the corresponding protein sequences. The comparison of the amino acid sequences revealed a number of conserved structural features such as a similar pattern of hydrophobic o 1992 Academic Press, I~C. termini and an identical cysteine skeleton.

defense by expression of glycoproteins related to complement control proteins encoded by the regulators of complement activation (RCA) gene cluster on human chromosome I (19). In this communication, we describe the first example of a viral homologue of human CD59, an antigen involved in restriction of complement-mediated lysis (20) and signal transduction for T-cell activation (2 1). The reading frame 15 of HVS group A strain 11 consists of 363 nucleotides (nt) (Fig. 1). It is located at position 29,231-29,593 of the genome and has been sequenced on both strands as described elsewhere (22). The search in current databases (72,208 entries; 92,912,352 residues) using the BLASTN program (23) implemented in the GCG program package (24) at the European Molecular Biology Laboratory (EMBL) revealed nucleotide sequence identity of 67% between position 33-323 of HVS-15 and the corresponding region of human CD59 cDNA. When the whole reading frames were compared, the sequence identity decreased to 64% (Figs. 2 and 3A). These data indicate that HVS-15 is as similar to the cellular CD59 gene as the HVS thymidylate synthase gene is to its cellular counterparts (25, 26). The amino acid sequence deduced from HVS-15 consist of 121 residues. We predict a leader peptide of 19 amino acids by the method of Heijne (27) and by comparison with human CD59 (21, 28, 29). The nascent polypeptide chain of HVS-15 may be processed at the equivalent residue of human CD59 since the environment of the cleavage site is identical (Figs. 2 and 3B). Both the predicted viral poly-

Herpesvirus saimiri (HVS) is the prototype of the y2subfamily of herpesviruses (Rhadinoviruses) (I, 2). The left-terminal region has been shown to be required for immortalization of marmoset lymphocytes in vitro (3) but is dispensable for growth of virus in permissive cell culture. Spontaneous or constructed deletions in this region (4, 5) had provided evidence that a predicted polypeptide of 164 amino acids (STP-A, saimiri-transformation-associated protein of group A) is required for transformation (6). Based on sequence diversity at the left end of L-DNA in different HVS isolates (7, 8), these can be divided into the three subgroups A, B, and C (9). A unique feature of this genomic region is the occurrence of genes for the first known virally encoded URNAs (8, IO- 73) and a gene for a dihydrofolate reductase (14). Viruses of groups A, B, and C have also been shown to have distinct transformation capacities (3, 9, 15). Potential oncogenes have been identified for group A and C (16, 17) but the mechanisms of T-lymphocyte transformation remain to be elucidated. Several genes have been identified as candidates for being involved in cell proliferation control. They were found by sequence comparison with entries of current databases and included homologues of members of the cyclin and the G protein-coupled receptor family (18). Additionally, cells infected with HVS, and the virus itself, may have the ability to evade the host’s immune

’ To whom reprint requests should * Present address: John Hopkins Bond Street, Baltimore, MD 21231.

be addressed. Oncology Center,

418

North

527

0042-6822/92

$5.00

Copyright 0 1992 by Academic Press, Inc. All rights of reproduction in any form reserved.

528

SHORT c

Hind111

dyad

COMMUNICATIONS -t

symmetry

AAGCTTCTATTTATACTACATTAGAGGCATTTTTTCAAAAGCAAAAATGCCTCTAATTATATACACTGTACTATTTACCTCTATTACACATTTTCTATTT

100

TAAGTCTGATAGTGATTAATCAAGAAAAAAGTTTGTGGTTCTCAGGGGATTAGTTCACAAGCTGTCTGAGGTTAAGGGTGTTTCTTTGGCACTGACACAG

200

Hind111

MYILFTLVLT

AAGTTGCTATAAGAATTGAAGCTTGCTTTACAAAAAGTTACTTGTGATTAATTACTATAACAAGAAAGGTAATGTATATTTTGTTTACGTTGGTACTGAC F

V

F

C

K

P

I

H

S-~DL

Q

C

Y

N

C

S

H

ST

H

Cl

C

T

T

ST

S

C

T

S

N

300

L

D

TTTTGTTTTTTGCAAGCCAATACACAGCTTGCAATGCTACAACTGTTCTCACTCAACTATGCAGTGTACTACATCTACTAGTTGTACATCTAATCTTGAC

400

SCLIAKAGSGVYYRCWKFDDCSFKRISNQLSETQ TCTTGTCTCATTGCTAAAGCTGGGTCAGGAGTATATTACAGGTGTTGGAAGTTTGATGACTGTAGCTTT~ACGTATCTCAAATCAATTGTCTGAAACAC L

KY

H

CC

K

K

N

L

C

NW&'

N

K

G

I

E

NIK

R

T

1

SD

KA

L

L

L

500 LA

AGTTAAAGTATCATTGTTGTAAGAAGAACTTGTGTAATGTGAACAAAGGGATTGAAAATATTAAAAGAACAATATCAGATAAAGCTCTTTTACTATTAGC

600

LFLVTAUNFPL* ATTGTTTTTAGTAACTGCTTGGAACTTTCCTCTTTAAAAGTCAACAACAAAACTATATTGTAACATTTATTTTTGTGTAGCTTATTTGTATTGCTATTAC

700

AAGTTAAAATATTGTGTTTTTTAAACTATAATTTTTAAAAAGATAAAATGAGATGTAGTATACTACATAGTCAAAATTAAAGTGCTAAATATTATTAGCA

800

PONY-A ATTTTTTATCAACAACGCAAATAAAAGTTAAGCTACTTTATTTTTTCTGTTATCTAAATCATTACGCGCTTCTTAGCATGTGTTAAAAGTTTTATGTGAT

900

TTTATTCTTACATATATAAAGCTAAATTTTAAAGCAAATTATCAGTAGCATCTTATCTTCTAATCTGTACAGACCTATATAATATGGGATTATCCTTAAG

1000

Hind111 1039

AAAAAACAGCGGAGAAAAAGAAAACACAGTGCCAAGCTT

FIG. 1. Nucleotide sequence and deduced amino acid sequence of the CD59 homologue of,HVS strain 11. Two HindIll fragments 815 bp, respectively, are displayed. The 5’region of reading frame 15 bears a 38-bp dyad symmetry, the 3’region has a polyadenylation indicated. Above the sequence the translation of HVS-15 is given in one-letter code. The predicted leader peptide of 19 amino acids arrowheads as is the putative mature peptide. The consensus sequence for N-linked glycosylation is underlined and a possible attachment at amino acid position 89 is marked by an double arrowhead.

peptide and human CD59 have a single consensus sequence for N-linked glycosylation (N X T/S, here in both cases NCS) and CD59 has been demonstrated to be glycosylated at this asparagine residue (30). HVS15 and CD59 have hydrophobic carboxy-terminal sequences (Fig. 3B); for CD59 it is known that this se-

452

T:T GUT GI~C TGT A&

of 218 and signal as is flanked by site for PIG

quence is replaced by a phophatidylinositol-glycan (PIG) anchor (20). The processing site for PIG linkage of CD59 is not known but is assumed to be&n70 based on comparison with the related mouse Ly-6 antigens of which the carboxyl-termini have been predicted (29, 31). This suggests that HVS-15 may also be a PIG-an-

TTT AE CGT A& TCA A!T CAA T+G TCT GAA ACA CAG TTA AAG TAT CAT TGT TGT AAG AAG AAC TTG TGT AAT GTG AAC AAA GGG ATT GAA 556

262 HTTT G G CAT ;a'Nu': TGC AAT TTC AAC GAC 10 GTC AC !i

CGC T CGC $-j TTG AGG ;['T~$jT;C~~ GAA AAT GAG CT

CG T'

I

I'

'I

118 116 Gk

C#

!b!

ii,

TIT 11: Iii

611, Cd:n C!!

bil,

366

649 460

FIG. 2. DNA and amino acid homology between HVS-I 5 and human CD59. Vertical lines indicate nucleotide sequence identity. The nucleotide positions refer to the EMBL Accession Numbers X64293 (HVS-15, upper row) and Xl 6447 (CD59, lower row). The putative signal peptide cleavage site is shown as a small arrow, possibly glycosylated asparagine residues are circled, conserved cysteines are marked by solid triangles and the proposed carboxy-termini by a solid circle. Identical amino acids and conservative substitutes are boxed. The nucleotide sequence identity is 64% and the amino acid sequence identity is 48% (62% similarity allowing conservative replacements). The alignments were generated using the GAP option of the GCG program package with the parameters for gap weight and length weight set to 5.0 and 0.3, respectively.

SHORT

A

o

500

COMMUNICATIONS

1000

B

529

o

50

1M

121 128

FIG. 3. Dotplot comparison of HVS-15 and human CD59. (A) DNA homology. Open arrows represent the translation range of each gene. The window/stringency parameters of the COMPARE program were set to 21/14. The scales indicate numbers of nucleotides. (6) Comparison of peptide sequences with the respective parameters set to 21 and 15, respectively. A plot displaying the hydropathic profiles of the proteins is integrated. Filled areas correspond to hydrophobic regions. The scales indicate numbers of residues.

chored protein since there is an asparagine residue at position 70 and the domain surrounding the proposed PIG modification site is highly conserved (Fig. 2). Processing of the HVS-15 precursor peptide may result in a mature peptide of 70 amino acids. The structure of the viral and the cellular protein is expected to be very similar based on the observation that(i) both predicted mature proteins share an overall identity of 53% (66% in consideration of conservative replacements), (ii) both have a single N-linked glycosylation site of the same context, (iii) all cysteine residues are at identical positions, and (iv) both may consist of 70 residues (Figs. l-3). The common features of their sequences suggest that both proteins may also share functional properties. Human CD59 is an 18- to 20-kDa glycoprotein that has been characterized as an inhibitor of the membrane attack complex of complement (20, 28, 32) and as a signal-transducing molecule for human T-cell activation complexed to a protein tyrosine kinase (PTK) (21, 33). Comparison of its amino acid sequence had revealed significant homology with murine Ly-6 anti-

TABLE SEQUENCE

1

HOMOLOGY BETWEEN HVS-15, HUMAN CD59, AND MURINE LY-6 PROTEINS CD59

Ly-6C.

47.9”

21.9 24.2

1

gens including conservation of 10 cysteine residues and terminal hydrophobic segments. Similar homologies were observed when Ly-6 protein sequences were compared with HVS-15 (Table 1). Ly-6 antigens have been implicated in augmentation of response to T-cell receptor stimulation (34) and are also PIG-linked cell membrane molecules associated with PTKs (33). Proteins of the Ly-6 family have not been considered to be functional counterparts of CD59 because they have several distinct characteristics (28, 29). A possible evolutionary relationship between a squid glycoprotein and Ly-6 antigens has also been suggested (35). However, HVS-15 is the most closely related molecule to human CD59 identified to date (Table 1). This implies a possible function for HVS-15 either as an additional protein involved in evading host immune defense mechanisms by preventing HVS-bearing cells from complement-mediated lysis or as a novel candidate contributing to proliferation control of persistently infected cells or T-lymphocytes transformed by HVS. The DNA sequencing work was supported by the Medical Research Council, U.K., and its analysis by the Deutsche Forschungsgemeinschaft, Forschergruppe “DNA-Viren des haematopoetischen Systems.” The nucleotide sequence data will appear in the EMBL, GenBank and DDBJ Nucleotide Sequence Databases under the Accession Number X64273.

Ly-GE.1

REFERENCES HVS-15 CD59 Ly-6C. 1

20.3 23.4 66.4

a Numbers represent percentage identity. Comparisons of precursor protein sequences were done using the GAP program, the parameters for gap weight and length weight were set to 3.0 and 0.1, respectively.

1. ROIZMAN, B., In B. Roizman (ed.), The Herpesviruses, Vol. 1. Plenum Press, New York/London, p. l-23 (1982). 2. ROIZMAN, B., DESROSIERS, R. C., FLECKENSTEIN, B., LOPEZ, C., MINSON, A. C., and STUDDERT, M. J., Arch. Vim/. 123,425-449 (1992). 3. DESROSIERS, R. C., SILVA, D. P., WALDRON, L. M., and LET~IN, N. L., J. L’M. 57, 701-705 (1986).

530 4. 5. 6. 7. 8. 9. 10. 7 1. 12. 13. 74. 16. 16. 17.

18. 19. 20.

SHORT

COMMUNICATIONS

KOOMEY, J. M., MULDER, C., BURGHOFF, R. L., FLECKENSTEIN, B., and DESROSIERS, R. C., /. Viral. 50, 662-665 (1984). DESROSIERS, R. C., BAKKER, A., KAMINE, J., FALK, L. A., HUNT, R. D., and KING, N. W., Science 228, 184-l 87 (1985). MURTHY, S. C. S., TRIMBLE, I. J., and DESROSIERS, R. C., J. Viral. 63,3307-3314(1989). DESROSIERS, R. C., and FALK, L. A., J. Viral. 43, 352-356 (1982). BIESINGER, B., TRIMBLE, J. J., DESROSIERS, R. C., and FLECKENSTEIN, B., virologyl76, 505-514(1990). MEDVECZKY, P., SZOMO~ANYI, E.. DESROSIERS, R. C., and MULDER, C., /. Viral. 52, 938-944 (1984). MURTHY, S., KAMINE, J., and DESROSIERS, R. C., EMBOI. 5, 16251632 (1986). LEE, S. I., MURTHY, S. C. S., TRIMBLE, J. J., DESROSIERS, R. C., and STEITZ, J. A., Cell54, 599-607 (1988). WASSARMAN, D. A., LEE, S. I., and STEITZ, J. A., Nucleic Acids Res. 17, 1258 (1989). ALBRECHT, J.-C.. and FLECKENSTEIN, B., lvucleic Acids Res. 20, 1810 (1992). TRIMBLE, I. J., MURTHY, S. C. S., BAKKER, A., GRASSMANN, R., and DESROSIERS, R. C., Science 239, 1 145-l 147 (1988). SZOMOLANYI, E., MEDVECZKY, P., and MULDER, C., /. viral. 61, 3485-3490 (1987). JUNG, J. U., and DESROSIERS, R. C., J. Viral. 65, 6953-6960 (1991). JUNG, J. U., TRIMBLE, J. J., KING, N. W., BIESINGER, B., FLECKENSTEIN, B. W., and DESROSIERS, R. C., Proc. Nat/. Acad. Sci. USA 88,7051-7055(1991). NICHOLAS, J., CAMERON, K. R.. and HONESS, R. W., Nature (London) 355, 362-365 (1992). ALBRECHT, J.-C., and FLECKENSTEIN, B., /. \/ire/. 66, 3937-3940 (1992). DAVIES, A., SIMMONS, D. L., HALE, G., HARRISON, R. A., TIGHE, H., LACHMANN, P. J., and WALDMANN. H., J. Exp. Med. 170, 637654 (1989).

21. 22.

23. 24. 25. 26.

27. 28.

29.

30. 31. 32.

33. 34. 35.

KORTY, P. E., BRANDO, C., and SHEVACH, E. M., 1. Immunol. 146, 4092-4098(1991). ALBRECHT, J.-C., NICHOLAS, J., BILLER, D., CAMERON, K. R., BIESINGER, B., NEWMAN, C., WITTMANN, S., CRAXTON, M. A., COLEMAN, H.. FLECKENSTEIN, B., and HONESS, R. W., J. Viral., in press (1992). ALTSCHUL, S., GISH, W., MILLER, W., MYERS, E. W., and LIPMAN, D. J., /. Mol. Biol. 215, 403-410 (1990). DEVEREUX, J., HAEBERLI, P., and SMITHIES, O., Nucleic Acids Res. 12,387-395(1984). BODEMER, W., NILLER, H.-H., NITSCHE, N., SCHOLZ, B., and FLECKENSTEIN, B., 1. t’irol. 60, 1 14-123 (1986). HONESS, R. W., BODEMER, W., CAMERON, K. R., NILLER, H.-H., FLECKENSTEIN, B., and RANDALL, R. E., froc. Nat/. Acad. Sci. USA 83,3604-3608 (1986). HEIJNE, G. VON., Nucleic Acids Res. 14, 4683-4690 (1986). SUGITA, Y., TOBE, T., ODA, E., TOMITA, M., YASUKAWA, K.. YAMAII, N., TAKEMOTO, T., FURUICHI, K., TAKAYAMA, M., and YANO, S.,/. Biochem. 106,555-557(1989). SAWADA, R., OHASHI, K., ANAGUCHI, H., OKAZAKI, H., HATTORI, M., MINATO, N., and NARUTO, M., DNA Cell Biol. 9, 213-220 (1990). STEFANOV~, I., HILGERT, I., KRISTOFAVA, H., BROWN, R., Low, G. M., and HOREJSI, V., Mol. lmmunol. 26, 153-161 (1989). FERGUSON, M. A. J., and WILLIAMS, A. F., Annu. Rev. Biochem. 57,285-320(1988). MERI, S., MORGAN, B. P., DAVIES, A., DANIELS, R. H., OLAVESEN, M. G., WALDMANN, H.. and LXHMANN, P. J., lmmunology71, l-9 (1990). STEFANOVA, I., HOREJSI, V.. ANSOTEGUI, I. J., KNAPP, W., and STOCKINGER, H., Science 254, 1016-1019 (1991). SHEVACH, E. M., and KORTY, P. E., Immunol. Today 10, 195-200 (1989). WILLIAMS, A. F., TSE, A. G. D., and GAGNON, J.. Immunogenetics 27,265-272(1988).

Herpesvirus saimiri has a gene specifying a homologue of the cellular membrane glycoprotein CD59.

Herpesvirus saimiri (HSV) is a T-lymphotropic tumor virus that causes fulminant lymphomas and leukemias in various New World primates other than its n...
400KB Sizes 0 Downloads 0 Views