当前位置: 首页 > 医学版 > 期刊论文 > 基础医学 > 病菌学杂志 > 2005年 > 第22期 > 正文
编号:11201996
The Herpes Simplex Virus Type 1 Locus That Encodes
     Department of Veterinary and Biomedical Sciences, Nebraska Center for Virology, University of Nebraska, Fair Street at East Campus Loop, Lincoln, Nebraska 68583-0905

    Department of Ophthalmology, College of Medicine, University of California at Irvine, UCIMC, Bldg. 55, Room 204, 101 The City Drive, Orange, California 92868-4380-02

    School of Biological Sciences, Nebraska Center for Virology, University of Nebraska, Lincoln, Nebraska

    ABSTRACT

    Herpes simplex virus type 1 (HSV-1) is the leading cause of virus-induced encephalitis; however, the viral genes that regulate encephalitis have not been well characterized. In this study, we tested whether the LAT (latency-associated transcript) locus regulates the frequency of encephalitis in male or female mice. Male BALB/c mice are more susceptible to HSV-1-induced encephalitis than age-matched female BALB/c mice. Deletion of LAT coding sequences reduced the frequency of encephalitis. A recombinant virus containing the first 1.5 kb of the LAT coding sequence induces levels of encephalitis in male BALB/c mice similar to those induced by wild-type HSV-1.

    TEXT

    Approximately 90% of the population is infected with herpes simplex virus type 1 (HSV-1) (29, 55). Following acute infection, latency is established in sensory neurons in trigeminal ganglia or sacral dorsal root ganglia (19, 50). Reactivation from latency periodically occurs, resulting in virus transmission and recurrent disease (19, 20).

    The latency-associated transcript (LAT) is abundantly transcribed in latently infected neurons (8, 10, 11, 22, 27, 44, 48, 51, 52). An 8.3-kb LAT transcript is spliced, yielding the stable 2-kb LAT intron that localizes to the nucleus (2, 31, 49) and an unstable 6.3-kb transcript (11, 44, 57). Proper splicing of LAT is necessary for establishment and maintenance of latency (21). The HSV-1 McKrae strain is frequently shed in tears of latently infected rabbits because of spontaneous reactivation (39-43), and LAT gene expression is crucial for spontaneous reactivation. LAT inhibits apoptosis in cultured cells and trigeminal ganglia of infected rabbits or mice (1, 14, 17, 18, 21, 37). The ability of LAT to interfere with apoptosis correlates with spontaneous reactivation (17, 18).

    HSV-1-induced encephalitis is a severe form of focal necrotizing encephalitis that affects approximately 2,000 individuals each year in the United States (12, 24, 54, 55). Without antiviral therapy, the mortality rate can be 70% (45, 46). Approximately 2/3 of HSV-1-induced encephalitis cases are due to reactivation from latency (56); consequently, high morbidity and long-term complications occur despite antiviral treatment (23, 26, 45).

    In general, females induce a stronger immune response because they express higher levels of lymphocytes and cytokines (3, 5, 16). Susceptibilities to encephalomyocarditis virus (9), vesicular stomatitis virus (4), coxsackievirus B3 (15), and HSV-1 infections in mice (13) differ based on gender. Furthermore, a HSV-2 vaccine trial in humans succeeded in females but not in males (47). In female mice, a HSV resistance locus (sex modifier locus) has been identified (25). HSV-1 hematogenous infection is more pathogenic to females (6), and estrogen influences pseudorabies virus infections in the central nervous system (53), suggesting that gender-specific factors, male as well as female, influence host-pathogen interactions.

    Based on the observations that gender can influence the pathogenic potential of certain viruses, we tested whether LAT influences the frequencies of encephalitis in male and female mice. Adult female or male BALB/c mice were infected with the wild-type (wt) McKrae strain or the LAT locus mutant, dLAT2903 (Fig. 1). As previously described for 129 Sv//Ev or RGKO male mice (13), male BALB/c mice infected with wt HSV-1 or dLAT2903R (rescued dLAT2903) (Fig. 1A) had frequencies of survival significantly lower than those of female BALB/c mice (Fig. 2A and B). A Tukey-Kramer multiple-comparison post-analysis of variance test revealed that survival rates for male and female mice infected with wt HSV-1 or dLAT2903R were significantly different (P = 0.004). Male mice infected with dLAT2903 had significantly higher (P = 0.003) levels of survival than those infected with wt HSV-1 or dLAT2903R. As previously reported (38), female mice infected with dLAT2903, wt HSV-1, or dLAT2903R exhibited similar survival rates.

    The deletion within dLAT2903 prevents expression of at least four genetic elements (Fig. 1C): (i) the stable 2-kb LAT; (ii) the first 1.5 kb of LAT, which contains only part of the stable 2-kb LAT and is sufficient for high levels of spontaneous reactivation in the rabbit eye model (41) or for trigeminal ganglia explant-induced reactivation in mice (38); (iii) the AL transcript (36); and (iv) the UOL transcript (30). To localize regions of LAT necessary for enhancing encephalitis in male BALB/c mice, we tested LAT3.3A (Fig. 1D) and LAT1.8A (Fig. 1E). LAT3.3A expresses UOL, AL, and one copy of the first 1.5 kb of LAT but does not express the intact stable 2-kb LAT. LAT1.8A contains one copy of the minimal UOL coding sequences but not AL or LAT. Male, but not female, BALB/c mice infected with LAT3.3A had a frequency of encephalitis significantly higher than that for age-matched male mice infected with LAT1.8A (P = 0.003) (Fig. 2A and B). Furthermore, LAT3.3A, wt HSV-1, and dLAT2903R induced similar levels of encephalitis in male BALB/c mice, indicating that expression of the intact stable 2-kb LAT does not regulate gender-specific encephalitis. Finally, LAT1.8A induced levels of encephalitis in male BALB/c mice similar to those induced by dLAT2903.

    Following infection of male or female mice with the respective viruses, the mice that died typically succumbed to fatal encephalitis between 7 and 10 days after infection (Fig. 2C). Similar virus titers were also detected in brain homogenates at 2, 4, or 8 days after infection, regardless of the virus used to infect male (Fig. 3) or female BALB/c mice (data not shown). Consistent with previous studies (28, 35, 38), we also found similar levels of infectious virus in ocular swabs from male and female mice following infection with the virus strains. These studies suggested that the reduced frequency of encephalitis in male BALB/c mice infected with dLAT2903 was not due to reduced viral titers in the brain.

    Since the LAT locus inhibits interferon (IFN) RNA expression in neuronal cell types (34), we tested whether IFN was differentially expressed in brains of mice during acute infection (2 to 8 days after infection) or after latency was established (14 days after infection). Using primers that specifically amplify mouse gamma IFN (IFN-) (Fig. 4C), male mice infected with dLAT2903 contained readily detectable levels of IFN- RNA at 6, 8, and 14 days after infection, but mock-infected mice did not (Fig. 4A). In contrast, male mice infected with the McKrae strain or dLAT2903R expressed IFN- RNA at 6 and 8 days, but not at 14 days, after infection. Viral glycoprotein (gC) was detected on days 6 and 8 regardless of the virus used for infection. At 4 days after infection, low levels of gC RNA were detected in male mice infected with the McKrae strain or with dLAT2903R but not in those infected with dLAT2903. gC RNA expression levels were similar in brains of male mice at 6 and 8 days after infection, regardless of which virus was used. As expected, LAT was not detected in mice infected with dLAT2903 (Fig. 4A). Female mice infected with dLAT2903, dLAT2903R, or wt HSV-1 expressed IFN- RNA 6 and 8 days after infection (Fig. 4B). These findings suggested that a correlation exists between survival of male mice infected with dLAT2903 and prolonged IFN- RNA expression in brains. Our results are also consistent with studies demonstrating that IFN- protects against HSV infection (7, 13).

    In summary, these studies indicated that the ability of HSV-1 to induce high levels of fatal encephalitis in male BALB/c mice was dependent on expression of the first 1.5 kb of LAT, expression of UOL, and/or expression of AL. We further suggest that the male mouse model may be useful for identifying sequences in the LAT locus that regulate the latency-reactivation cycle and neuropathogenesis. The first 1.5 kb of LAT promotes spontaneous reactivation (40, 41) and inhibits caspase 8-induced apoptosis efficiently (33). The functions of AL and UOL are not known. Additional studies are required to prove whether AL, UOL, or the 1.5-kb LAT sequence regulates neurovirulence in male BALB/c mice. Since expressions of the 1.5-kb LAT, of UOL, and of AL are regulated by the same promoter and their coding sequences overlap one another, preparing recombinant viruses that express just one of the respective gene products is a challenge.

    ACKNOWLEDGMENTS

    This work was supported by Public Health Service grant 1P20RR15635 and by two USDA grants (2002-35204 and 2003-02213).

    REFERENCES

    Ahmed, M., M. Lock, C. G. Miller, and N. W. Fraser. 2002. Regions of the herpes simplex virus type 1 latency-associated transcript that protect cells from apoptosis in vitro and protect neuronal cells in vivo. J. Virol. 76:717-729.

    Ahmed, M., and N. Fraser. 2001. Herpes simplex virus type 1 2-kilobase latency-associated transcript intron associates with ribosomal proteins and splicing factors. J. Virol. 75:12070-12080.

    Aida, T., N. Ishikawa, and K. Shinkai. 1998. Sex differences in immune responses to cephalothin in guinea pigs. J. Toxicol. Sci. 23:87-91.

    Barna, M., T. Komatsu, Z. Bi, and C. S. Reiss. 1996. Sex differences in susceptibility to viral infection of the central nervous system. J. Neuroimmunol. 67:31-39.

    Barrat, F., B. Lesourd, H. J. Boulouis, D. Thibault, N. S. Vincent, B. Gjata, A. Louise, T. Neway, and C. Pilet. 1997. Sex and parity modulate cytokine production during murine aging. Clin. Exp. Immunol. 109:562-568.

    Burgos, J. S., C. Ramirez, I. Sastre, J. M. Alfaro, and F. Valdivieso. 2005. Herpes simplex virus type 1 infection via the bloodstream with apolipoprotein E dependence in the gonads is influenced by gender. J. Virol. 79:1605-1612.

    Cantin, E., B. Tanamachi, H. Openshaw, J. Mann, and K. Clarke. 1999. Gamma interferon (IFN-) receptor null-mutant mice are more susceptible to herpes simplex virus type 1 infection than IFN- ligand null-mutant mice. J. Virol. 73:5196-5200.

    Croen, K. D., J. M. Ostrove, L. J. Dragovic, J. E. Smialek, and S. E. Straus. 1987. Latent herpes simplex virus in human trigeminal ganglia. Detection of an immediate early gene "anti-sense" transcript by in situ hybridization. N. Engl. J. Med. 317:1427-1432.

    Curiel, R. E., M. H. Miller, R. Ishikawa, D. C. Thomas, and N. J. Bigley. 1993. Does the gender difference in interferon production seen in picornavirus-infected spleen cell cultures from ICR Swiss mice have any in vivo significance J. Interferon Res. 13:387-395.

    Deatly, A. M., J. G. Spivack, E. Lavi, and N. W. Fraser. 1987. RNA from an immediate early region of the type 1 herpes simplex virus genome is present in the trigeminal ganglia of latently infected mice. Proc. Natl. Acad. Sci. USA 84:3204-3208.

    Deatly, A. M., J. G. Spivack, E. Lavi, D. R. D. O'Boyle, and N. W. Fraser. 1988. Latent herpes simplex virus type 1 transcripts in peripheral and central nervous system tissues of mice map to similar regions of the viral genome. J. Virol. 62:749-756.

    Gesser, R. M., and S. C. Koo. 1997. Latent herpes simplex virus type 1 gene expression in ganglia innervating the human gastrointestinal tract. J. Virol. 71:4103-4106.

    Han, X., P. Lundeberg, B. Tanamachi, H. Openshaw, J. Longmate, and E. Cantin. 2001. Gender influences herpes simplex virus type 1 infection in normal and gamma interferon-mutant mice. J. Virol. 75:3048-3052.

    Henderson, G., W. Peng, L. Jin, G.-C. Perng, A. B. Nesburn, S. L. Wechsler, and C. Jones. 2002. Regulation of caspase 8- and caspase 9-induced apoptosis by the herpes simplex virus latency-associated transcript. J. Neurovirol. 8:103-111.

    Huber, S., and B. Pfaeffle. 1994. Differential Th1 and Th2 cell responses in male and female BALB/c mice infected with coxsackievirus group B type 3. J. Virol. 68:5126-5132.

    Huppert, F. A., W. Solomou, S. O'Connor, K. Morgan, P. Sussams, and C. Brayne. 1998. Aging and lymphocyte subpopulations: whole-blood analysis of immune markers in a large population sample of healthy elderly individuals. Exp. Gerontol. 33:593-600.

    Inman, M., G. C. Perng, G. Henderson, H. Ghiasi, A. B. Nesburn, S. L. Wechsler, and C. Jones. 2001. Region of herpes simplex virus type 1 latency-associated transcript sufficient for wild-type spontaneous reactivation promotes cell survival in tissue culture. J. Virol. 75:3636-3646.

    Jin, L., W. Peng, G.-C. Perng, A. B. Nesburn, C. Jones, and S. L. Wechsler. 2003. Identification of herpes simplex virus type 1 latency associated transcript sequences that both inhibit apoptosis and enhance the spontaneous reactivation phenotype. J. Virol. 77:6556-6561.

    Jones, C. 1998. Alphaherpesvirus latency: its role in disease and survival of the virus in nature. Adv. Virus Res. 51:81-133.

    Jones, C. 2003. Herpes simplex virus type 1 and bovine herpesvirus 1 latency. Clin. Microbiol. Rev. 16:79-95.

    Kang, W., R. Mukerjee, and N. F. Fraser. 2003. Establishment and maintenance of HSV latent infection is mediated through correct splicing of the LAT primary transcript. Virology 312:233-244.

    Krause, P. R., K. D. Croen, S. E. Straus, and J. M. Ostrove. 1988. Detection and preliminary characterization of herpes simplex virus type 1 transcripts in latently infected human trigeminal ganglia. J. Virol. 62:4819-4823.

    Lahat, E., J. Barr, G. Barkai, G. Paret, N. Brand, and A. Barzilai. 1999. Long term neurological outcome of herpes encephalitis. Arch. Dis. Child. 80:69-71.

    Lohr, J. M., J. A. Nelson, and M. B. Oldstone. 1990. Is herpes simplex virus associated with peptic ulcer disease J. Virol. 64:2168-2174.

    Lundberg, P., P. Welander, H. Openshaw, C. Nalibandian, C. Edwards, L. Moldawer, and E. Cantin. 2003. A locus on mouse chromosome 6 that determines resistance to herpes simplex virus also influences reactivation, while an unlinked locus augments resistance of female mice. J. Virol. 77:11661-11673.

    McGrath, N., N. E. Anderson, M. C. Croxson, and K. F. Powell. 1997. Herpes simplex encephalitis treated with acyclovir: diagnosis and long term outcome. J. Neurol. Neurosurg. Psychiatry 63:321-326.

    Mitchell, W. J., R. P. Lirette, and N. W. Fraser. 1990. Mapping of low abundance latency-associated RNA in the trigeminal ganglia of mice latently infected with herpes simplex virus type 1. J. Gen. Virol. 71:125-132.

    Mott, K., N. Osorio, L. Jin, D. Brick, J. Naito, J. Cooper, G. Henderson, M. Inman, C. Jones, S. L. Wechsler, and G.-C Perng. 2003. The bovine herpesvirus 1 LR ORF2 is crucial for this gene's ability to restore the high reactivation phenotype to a herpes simplex virus-1 LAT null mutant. J. Gen. Virol. 84:2975-2985.

    Nahmias, A. J., and B. Roizman. 1973. Infection with herpes-simplex viruses 1 and 2. 3. N. Engl. J. Med. 289:781-789.

    Naito, J., R. Mukerjee, K. R. Mott, W. Kang, N. Osorio, N. W. Fraser, and G.-C. Perng. 2005. Identification of a protein encoded in the herpes simplex virus type 1 latency associated transcript promoter region. Virus Res. 108:101-110.

    Nicosia, M., J. M. Zabolotny, R. P. Lirette, and N. W. Fraser. 1994. The HSV-1 2-kb latency-associated transcript is found in the cytoplasm comigrating with ribosomal subunits during productive infection. Virology 204:717-728.

    Peng, W., G. Henderson, G.-C. Perng, A. B. Nesburn, S. L. Wechsler, and C. Jones. 2003. The gene that encodes the herpes simplex virus type 1 latency-associated transcript influences the accumulation of transcripts (Bcl-xL and Bcl-xS) that encode apoptotic regulatory proteins. J. Virol. 77:10714-10718.

    Peng, W., L. Jin, G. Henderson, G.-C. Perng, D. J. Brick, A. B. Nesburn, S. L. Wechsler, and C. Jones. 2004. Mapping herpes simplex virus type 1 (HSV-1) LAT sequences that protect from apoptosis mediated by a plasmid expressing caspase-8. J. Neurovirol. 10:260-265.

    Peng, W., G. Henderson, M. Inman, L. BenMohamed, G.-C. Perng, S. L. Wechsler, and C. Jones. 2005. The locus encompassing the latency-associated transcript of herpes simplex virus type 1 interferes with and delays interferon expression in productively infected neuroblastoma cells and trigeminal ganglia of acutely infected mice. J. Virol. 79:6162-6171.

    Perng, G.-C., B. Maguen, L. Jin, K. R. Mott, N. Osorio, S. M. Slanina, A. Yukht, H. Ghiasi, A. B. Nesburn, M. Inman, G. Henderson, C. Jones, and S. L. Wechsler. 2002. A gene capable of blocking apoptosis can substitute for the herpes simplex virus type 1 latency-associated transcript gene and restore wild-type reactivation levels. J. Virol. 76:1224-1235.

    Perng, G.-C., B. Maguen, L. Jin, K. R. Mott, J. Kurylo, L. BenMohamed, A. Yukht, N. Osorio, A. B. Nesburn, G. Henderson, M. Inman, C. Jones, and S. L. Wechsler. 2002. A novel herpes simplex virus type 1 transcript (AL-RNA) antisense to the 5' end of latency-associated transcript produces a protein in infected rabbits. J. Virol. 76:8003-8010.

    Perng, G.-C., C. Jones, J. Ciacci-Zanella, M. Stone, G. Henderson, A. Yukht, S. M. Slanina, F. M. Hoffman, H. Ghiasi, A. B. Nesburn, and S. Wechsler. 2000. Virus-induced neuronal apoptosis blocked by the herpes simplex virus latency-associated transcript (LAT). Science 287:1500-1503.

    Perng, G.-C., D. Esmail, S. Slanina, A. Yukht, H. Ghiasi, N. Osorio, K. R. Mott, B. Maguen, L. Jin, A. B. Nesburn, and S. L. Wechsler. 2001. Three herpes simplex virus type 1 latency-associated transcript mutants with distinct and asymmetric effects on virulence in mice compared with rabbits. J. Virol. 75:9018-9028.

    Perng, G. C., K. Chokephaibulkit, R. L. Thompson, N. M. Sawtell, S. M. Slanina, H. Ghiasi, A. B. Nesburn, and S. L. Wechsler. 1996. The region of the herpes simplex virus type 1 LAT gene that is colinear with the ICP34.5 gene is not involved in spontaneous reactivation. J. Virol. 70:282-291.

    Perng, G. C., E. C. Dunkel, P. A. Geary, S. M. Slanina, H. Ghiasi, R. Kaiwar, A. B. Nesburn, and S. L. Wechsler. 1994. The latency-associated transcript gene of herpes simplex virus type 1 (HSV-1) is required for efficient in vivo spontaneous reactivation of HSV-1 from latency. J. Virol. 68:8045-8055.

    Perng, G. C., H. Ghiasi, S. M. Slanina, A. B. Nesburn, and S. L. Wechsler. 1996. The spontaneous reactivation function of the herpes simplex virus type 1 LAT gene resides completely within the first 1.5 kilobases of the 8.3-kilobase primary transcript. J. Virol. 70:976-984.

    Perng, G. C., S. M. Slanina, H. Ghiasi, A. B. Nesburn, and S. L. Wechsler. 1996. A 371-nucleotide region between the herpes simplex virus type 1 (HSV-1) LAT promoter and the 2-kilobase LAT is not essential for efficient spontaneous reactivation of latent HSV-1. J. Virol. 70:2014-2018.

    Perng, G. C., S. M. Slanina, A. Yukht, B. S. Drolet, W. Keleher, Jr., H. Ghiasi, A. B. Nesburn, and S. L. Wechsler. 1999. A herpes simplex virus type 1 latency-associated transcript mutant with increased virulence and reduced spontaneous reactivation. J. Virol. 73:920-929.

    Rock, D. L., A. B. Nesburn, H. Ghiasi, J. Ong, T. L. Lewis, J. R. Lokensgard, and S. L. Wechsler. 1987. Detection of latency-related viral RNAs in trigeminal ganglia of rabbits latently infected with herpes simplex virus type 1. J. Virol. 61:3820-3826.

    Skoldenberg, B. 1991. Herpes simplex encephalitis. Scand. J. Infect. Dis. Suppl. 80:40-46.

    Skoldenberg, B., M. Forsgren, K. Alestig, T. Bergstrom, L. Burman, E. Dahlqvist, A. Forkman, A. Fryden, K. Lovgren, and K. Norlin. 1984. Acyclovir versus vidarabine in herpes simplex encephalitis. Randomised multicentre study in consecutive Swedish patients. Lancet ii:707-711.

    Stanberry, L. R., S. L. Spruance, A. L. Cunningham, D. I. Bernstein, A. Mindel, S. Sacks, S. Tyring, F. Y. Aoki, M. Slaoui, M. Denis, P. Vandepapeliere, and G. Dubin, for the GlaxoSmithKline Herpes Vaccine Efficacy Study Group. 2002. Glycoprotein-D-adjuvant vaccine to prevent genital herpes. N. Engl. J. Med. 347:1652-1661.

    Stevens, J. G., E. K. Wagner, G. B. Devi-Rao, M. L. Cook, and L. T. Feldman. 1987. RNA complementary to a herpesvirus alpha gene mRNA is prominent in latently infected neurons. Science 235:1056-1059.

    Thomas, D. L., M. Lock, J. M. Zabolotny, B. R. Mohan, and N. W. Fraser. 2002. The 2-kilobase intron of the herpes simplex virus type 1 latency-associated transcript has a half-life of approximately 24 hours in SY5Y and COS-1 cells. J. Virol. 76:532-540.

    Wagner, E. K., and D. C. Bloom. 1997. Experimental investigation of herpes simplex virus latency. Clin. Microbiol. Rev. 10:419-443.

    Wagner, E. K., G. Devi-Rao, L. T. Feldman, A. T. Dobson, Y. F. Zhang, W. M. Flanagan, and J. G. Stevens. 1988. Physical characterization of the herpes simplex virus latency-associated transcript in neurons. J. Virol. 62:1194-1202.

    Wagner, E. K., W. M. Flanagan, G. Devi-Rao, Y. F. Zhang, J. M. Hill, K. P. Anderson, and J. G. Stevens. 1988. The herpes simplex virus latency-associated transcript is spliced during the latent phase of infection. J. Virol. 62:4577-4585.

    Weiss, M. L., M. E. Dobbs, P. S. MohanKumar, S. I. Chowdhury, K. Sawrey, R. Guevara-Guzman, and J. Huang. 2001. The estrous cycle affects pseudorabies virus (PRV) infection of the CNS. Brain Res. 893:215-226.

    Whitley, R. 1991. Herpes simplex virus infections of the central nervous system. Encephalitis and neonatal herpes. Drugs 42:406-427.

    Whitley, R. 1997. Herpes simplex virus. Lippincott-Raven Publishers, Philadelphia, Pa.

    Yamada, S., T. Kameyama, S. Nagaya, Y. Hashizume, and M. Yoshida. 2002. Relapsing herpes simplex encephalitis: pathological confirmation of viral reactivation. J. Neurol. Neurosurg. Psychiatry 74:262-264.

    Zwaagstra, J. C., H. Ghiasi, S. M. Slanina, A. B. Nesburn, S. C. Wheatley, K. Lillycrop, J. Wood, D. S. Latchman, K. Patel, and S. L. Wechsler. 1990. Activity of herpes simplex virus type 1 latency-associated transcript (LAT) promoter in neuron-derived cells: evidence for neuron specificity and for a large LAT transcript. J. Virol. 64:5019-5028.(Clinton Jones, Melissa In)