Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Susan M. Slanina is active.

Publication


Featured researches published by Susan M. Slanina.


Journal of Virology | 2000

The Latency-Associated Transcript Gene Enhances Establishment of Herpes Simplex Virus Type 1 Latency in Rabbits

Guey Chuen Perng; Susan M. Slanina; Ada Yukht; Homayon Ghiasi; Anthony B. Nesburn; Steven L. Wechsler

ABSTRACT The latency-associated transcript (LAT) gene the only herpes simplex virus type 1 (HSV-1) gene abundantly transcribed during neuronal latency, is essential for efficient in vivo reactivation. Whether LAT increases reactivation by a direct effect on the reactivation process or whether it does so by increasing the establishment of latency, thereby making more latently infected neurons available for reactivation, is unclear. In mice, LAT-negative mutants appear to establish latency in fewer neurons than does wild-type HSV-1. However, this has not been confirmed in the rabbit, and the role of LAT in the establishment of latency remains controversial. To pursue this question, we inserted the gene for the enhanced green fluorescent protein (EGFP) under control of the LAT promoter in a LAT-negative virus (ΔLAT-EGFP) and in a LAT-positive virus (LAT-EGFP). Sixty days after ocular infection, trigeminal ganglia (TG) were removed from the latently infected rabbits, sectioned, and examined by fluorescence microscopy. EGFP was detected in significantly more LAT-EGFP-infected neurons than ΔLAT-EGFP-infected neurons (4.9% versus 2%, P < 0.0001). The percentages of EGFP-positive neurons per TG ranged from 0 to 4.6 for ΔLAT-EGFP and from 2.5 to 11.1 for LAT-EGFP (P = 0.003). Thus, LAT appeared to increase neuronal latency in rabbit TG by an average of two- to threefold. These results suggest that LAT enhances the establishment of latency in rabbits and that this may be one of the mechanisms by which LAT enhances spontaneous reactivation. These results do not rule out additional LAT functions that may be involved in maintenance of latency and/or reactivation from latency.


Journal of Virology | 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

Guey Chuen Perng; Barak Maguen; Ling Jin; Kevin R. Mott; Nelson Osorio; Susan M. Slanina; Ada Yukht; Homayon Ghiasi; Anthony B. Nesburn; Melissa Inman; Gail Henderson; Clinton Jones; Steven L. Wechsler

ABSTRACT After ocular herpes simplex virus type 1 (HSV-1) infection, the virus travels up axons and establishes a lifelong latent infection in neurons of the trigeminal ganglia. LAT (latency-associated transcript), the only known viral gene abundantly transcribed during HSV-1 neuronal latency, is required for high levels of reactivation. The LAT function responsible for this reactivation phenotype is not known. Recently, we showed that LAT can block programmed cell death (apoptosis) in neurons of the trigeminal ganglion in vivo and in tissue culture cells in vitro (G.-C. Perng et al., Science 287:1500–1503, 2000; M. Inman et al., J. Virol. 75:3636–3646, 2001). Consequently, we proposed that this antiapoptosis function may be a key to the mechanism by which LAT enhances reactivation. To study this further, we constructed a recombinant HSV-1 virus in which the HSV-1 LAT gene was replaced by an alternate antiapoptosis gene. We used the bovine herpes virus 1 (BHV-1) latency-related (LR) gene, which was previously shown to have antiapoptosis activity, for this purpose. The resulting chimeric virus, designated CJLAT, contains two complete copies of the BHV-1 LR gene (one in each viral long repeat) in place of the normal two copies of the HSV-1 LAT, on an otherwise wild-type HSV-1 strain McKrae genomic background. We report here that in both rabbits and mice reactivation of CJLAT was significantly greater than the LAT null mutant dLAT2903 (P < 0.0004 and P = 0.001, respectively) and was at least as efficient as wild-type McKrae. This strongly suggests that a BHV-1 LR gene function was able to efficiently substitute for an HSV-1 LAT gene function involved in reactivation. Although replication of CJLAT in rabbits and mice was similar to that of wild-type McKrae, CJLAT killed more mice during acute infection and caused more corneal scarring in latently infected rabbits. This suggested that the BHV-1 LR gene and the HSV-1 LAT gene are not functionally identical. However, LR and LAT both have antiapoptosis activity. These studies therefore strongly support the hypothesis that replacing LAT with an antiapoptosis gene restores the wild-type reactivation phenotype to a LAT null mutant of HSV-1 McKrae.


Antiviral Research | 1995

Vaccination of mice with herpes simplex virus type 1 glycoprotein D DNA produces low levels of protection against lethal HSV-1 challenge

Homayon Ghiasi; Steve Cai; Susan M. Slanina; Anthony B. Nesburn; Steven L. Wechsler

The herpes simplex virus type 1 (HSV-1) glycoprotein D (gD) gene was inserted into vectors pSVL or pRc/CMV under control of the SV40 late promoter or the human cytomegalovirus major immediate-early promoter, respectively. Intramuscular injection of mice with these gD-containing plasmids appeared to induce low levels of serum anti-gD antibody, as judged by the appearance of low levels of anti-HSV-1-neutralizing antibody and anti-gD ELISA responses in the serum of gD-DNA-vaccinated mice. As previously reported in other virus systems, vaccination with vector DNA also induced ELISA and neutralizing antibody titers. However, these titers were lower than those induced by the gD-containing plasmids. The ELISA and neutralization titers induced by the vectors appeared to be non-specific rather than directed at specific HSV-1 proteins, since serum from mice vaccinated with plasmid-gD immunoprecipitated significant amounts of gD from extracts of HSV-1-infected cells, while serum from mice vaccinated with vectors was unable to immunoprecipitate gD or any other obvious HSV-1 proteins. Neither pSVL-gD nor pRc/CMV-gD induced detectable lymphocyte proliferative or CTL responses. Vaccination with pSVL-gD provided a significant (P = 0.04, Fishers exact test), but low level of protection against lethal challenge with HSV-1. Vaccination with pRc/CMV-gD also appeared to provide a low level of protection against challenge, that was statistically significance at the 10% level (P = 0.054, Fishers exact test). Reports from numerous laboratories (including ours) have shown that vaccination with recombinantly expressed gD can provide very high levels of protection against HSV-1 lethal challenge. Thus, the results reported here suggest that vaccination with HSV-1 gD-DNA is not yet a useful alternative to a gD subunit vaccine.


Journal of Virology | 2001

Three Herpes Simplex Virus Type 1 Latency-Associated Transcript Mutants with Distinct and Asymmetric Effects on Virulence in Mice Compared with Rabbits

Guey Chuen Perng; Daniel Esmaili; Susan M. Slanina; Ada Yukht; Homayon Ghiasi; Nelson Osorio; Kevin R. Mott; Barak Maguen; Ling Jin; Anthony B. Nesburn; Steven L. Wechsler

ABSTRACT Herpes simplex virus type 1 latency-associated transcript (LAT)-null mutants have decreased reactivation but normal virulence in rabbits and mice. We report here on dLAT1.5, a mutant with LAT nucleotides 76 to 1667 deleted. Following ocular infection of rabbits, dLAT1.5 reactivated at a lower rate than its wild-type parent McKrae (6.1 versus 11.8%; P = 0.0025 [chi-square test]). Reactivation was restored in the marker-rescued virus dLAT1.5R (12.6%;P = 0.53 versus wild type), confirming the importance of the deleted region in spontaneous reactivation. Compared with wild-type or marker-rescued virus, dLAT1.5 had similar or slightly reduced virulence in rabbits (based on survival following ocular infection). In contrast, in mice,dLAT1.5 had increased virulence (P< 0.0001). Thus, deletion of LAT nucleotides 76 to 1667 increased viral virulence in mice but not in rabbits. In contrast, we also report here that LAT2.9A, a LAT mutant that we previously reported to have increased virulence in rabbits (G. C. Perng, S. M. Slanina, A. Yuhkt, B. S. Drolet, W. J. Keleher, H. Ghiasi, A. B. Nesburn, and S. L. Wechsler, J. Virol. 73:920–929, 1999), had decreased virulence in mice (P = 0.03). In addition, we also found that dLAT371, a LAT mutant that we previously reported to have wild-type virulence in rabbits (G. C. Perng, S. M. Slanina, H. Ghiasi, A. B. Nesburn, and S. L. Wechsler, J. Virol. 70:2014–2018, 1996), had decreased virulence in mice (P < 0.05). Thus, these three mutants, each of which encodes a different LAT RNA, have different virulence phenotypes. dLAT1.5 had wild-type virulence in rabbits but increased virulence in mice. In contrast, LAT2.9A had increased virulence in rabbits but decreased virulence in mice, anddLAT371 had wild-type virulence in rabbits but decreased virulence in mice. Taken together, these results suggest that (i) the 5′ end of LAT and/or a gene that overlaps part of this region is involved in viral virulence, (ii) this virulence appears to have species-specific effects, and (iii) regulation of this virulence may be complex.


Journal of General Virology | 2001

The effect of latency-associated transcript on the herpes simplex virus type 1 latency-reactivation phenotype is mouse strain-dependent.

Guey-Chuen Perng; Susan M. Slanina; Homayon Ghiasi; Anthony B. Nesburn; Steven L. Wechsler

Herpes simplex virus type 1 (HSV-1) latency-associated transcript (LAT) null mutants reactivate poorly in the rabbit ocular model. The situation in mice is less clear. Reports concluding that LAT null mutants reactivate poorly in the mouse explant-induced reactivation (EIR) model are contradicted by a similar number of reports of normal EIR of LAT(-) mutants in mice. To determine if the EIR phenotype might be mouse strain-dependent we infected BALB/c and Swiss Webster mice with LAT(-) or LAT(+) virus and assessed EIR in individual trigeminal ganglia. Compared to LAT(+) virus, LAT(-) virus reactivated poorly in Swiss Webster mice (P<0.05). In contrast, the EIR phenotype of these viruses was similar in BALB/c mice (P>0.1). Thus, LAT appeared to have a much greater impact on the EIR phenotype in Swiss Webster mice than in BALB/c mice. The mouse strain therefore appeared consequential in the HSV-1 EIR phenotype in mice.


Immunology | 1997

The importance of MHC-I and MHC-II responses in vaccine efficacy against lethal herpes simplex virus type 1 challenge.

H. Ghiasi; Derry C. Roopenian; Susan M. Slanina; Steve Cai; Anthony B. Nesburn; Steven L. Wechsler

To investigate the importance of major histocompatability complex (MHC) class I‐ and MHC class II‐dependent immune responses in herpes simplex virus‐1 (HSV‐1) vaccine efficacy, groups of β2m°/° (MHC I–) and Ab°/° (MHC II–) mice were inoculated with various vaccines, and then challenged intraperitoneally with HSV‐1. Following vaccination with either live avirulent HSV‐1, expressed HSV‐1 glycoprotein D (gD), or a mixture of seven expressed HSV‐1 glycoproteins (7gPs), Ab°/° (MHC‐II–) mice developed no enzyme‐linked immunosorbent assay (ELISA) or neutralizing antibody titres. In contrast, significant ELISA and neutralizing antibody titres were induced in β2m°/° (MHC‐I–) mice by all three vaccines. The neutralizing antibody titres were similar for all three vaccines, but were only ≈ 1/4 to 1/3 of that developed in C57BL/6 (parental) mice vaccinated with the same antigens. All three vaccines protected 100% of the wild‐type C57BL/6 mice against lethal challenge with 2×107 plaque‐forming units (PFU) of HSV‐1. The live virus vaccine and the 7gPs vaccine also protected 80% of the β2m°/° mice against the same lethal HSV‐1 challenge dose. In contrast, in Ab°/° mice, none of the vaccines provided significant protection against the same lethal challenge dose of HSV‐1. However, at a lower challenge dose of 2×106 PFU, all three vaccines protected 70–80% of the vaccinated Ab°/° mice (compared to only 10% survival in mock vaccinated controls). Thus, vaccination provided some protection against lethal HSV‐1 challenge in both β2m°/° and Ab°/° mice; however, the protection was less than that seen in the parental C57BL/6 mice. In addition, Ab°/° mice were less well protected by vaccination than were β2m°/° mice. Our results suggest that (1) both MHC‐I and MHC‐II are involved in vaccine efficacy against HSV‐1 challenge; (2) both types of responses must be present for maximum vaccine efficacy; and (3) the MHC‐II‐dependent immune response appeared to be more important than the MHC‐I‐dependent immune response for vaccine efficacy against HSV‐1 challenge.


The Journal of Infectious Diseases | 1999

The Role of Interleukin (IL)-2 and IL-4 in Herpes Simplex Virus Type 1 Ocular Replication and Eye Disease

Homayon Ghiasi; Steve Cai; Susan M. Slanina; Guey-Chuen Perng; Anthony B. Nesburn; Steven L. Wechsler

To assess the relative effect of interleukin (IL)-2- and IL-4-dependent immune responses on herpes simplex virus (HSV)-1 infection, naive, vaccinated, and mock-vaccinated IL-20/0 and IL-40/0 knockout mice were challenged ocularly with HSV-1. Naive IL-20/0 mice were significantly more susceptible to lethal infection than IL-40/0 or parental BALB/c mice. Vaccinated, IL-20/0, IL-40/0, and BALB/c mice induced similar neutralizing antibody titers and were completely protected against HSV-1-induced death and corneal scarring. Vaccinated and mock-vaccinated IL-20/0 mice had significantly higher HSV-1 titers in their eyes than BALB/c mice, while vaccinated and mock-vaccinated IL-40/0 mice had significantly lower HSV-1 titers in their eyes than BALB/c mice. Recombinant (r) IL-2 treatment of the IL-20/0 mice significantly reduced ocular HSV-1 replications, but rIL-4 treatment of IL-40/0 mice significantly increased ocular HSV-1 replications. Th1 (IL-2) cytokine responses may help protect mice against ocular HSV-1 challenge and reduce ocular HSV-1 replication.


Virology | 1992

Baculovirus-expressed glycoprotein E (gE) of herpes simplex virus type-1 (HSV-1) protects mice against lethal intraperitoneal and lethal ocular HSV-1 challenge

Homayon Ghiasi; Ravi Kaiwar; Anthony B. Nesburn; Susan M. Slanina; Steven L. Wechsler

We have constructed a recombinant baculovirus expressing high levels of the herpes simplex virus type 1 (HSV-1) glycoprotein E (gE) in Sf9 cells. The expressed gE migrated on gels as a double band with apparent molecular weights of 68 and 70 kDa. The recombinant gE was glycosylated based on its susceptibility to tunicamycin treatment and was transported to the membrane of Sf9 cells based on indirect immunofluorescence. Mice vaccinated with gE developed high serum titers of HSV-1-neutralizing antibodies based on plaque reduction assays. gE vaccination also induced a strong delayed type hypersensitivity (DTH) response to HSV-1. In addition, mice vaccinated with the recombinant gE were protected from both intraperitoneal and ocular lethal HSV-1 challenge. To our knowledge, this is the first report in which vaccination with gE was shown to induce high neutralizing antibody titers, a DTH response, or protection against lethal HSV-1 challenge.


Diabetes | 1985

Insulin Suppresses Rat Growth Hormone Messenger Ribonucleic Acid Levels in Rat Pituitary Tumor Cells

Shlomo Melmed; Lorna Neilson; Susan M. Slanina

Insulin has been shown to suppress growth hormone (GH) secretion by rat pituitary tumor cells (GH3) independently of glucose utilization. The effects of physiologic doses of insulin were therefore tested on cytoplasmic levels of GH messenger ribonucleic acid (mRNA) sequences. Insulin (3.5 nM) treatment of cells for 5 days suppressed the hybridization of cytoplasmic GH mRNA with 32P-cDNA for rGH by 50%. The three- to fourfold induction of cytoplasmic GH mRNA by hydrocortisone (1 μM) was also suppressed by insulin (3.5 nM) by 40%. The results show a direct suppression of cytoplasmic rGH mRNA concentration by physiologic doses of insulin. These findings may be due to either decreased rate of GH gene transcription, increased intracellular breakdown, or decreased nuclear-cytoplasmic transport of GH mRNA caused by insulin.


Archives of Virology | 1994

Expression and characterization of baculovirus expressed herpes simplex virus type 1 glycoprotein L

H. Ghiasi; Ravi Kaiwar; Susan M. Slanina; Anthony B. Nesburn; Steven L. Wechsler

SummaryWe have constructed a recombinant baculovirus expressing high levels of the herpes simplex virus type 1 (HSV-1) glycoprotein L (gL) in Sf9 cells. Sf9 cells infected with this recombinant virus synthesized three polypeptides of 26–27 kDa 28 kDa, and 31 kDa. The 28 and 31 kDa species were sensitive to tunicamycin and N-glycosidase F (PNGase F) treatment, suggesting that they were glycosylated. As shown by both indirect immunofluorescence and Western blot analysis, using polyclonal antibodies to synthetic gL peptides indicated that the baculovirus expressed gL was abundant on the surface of baculovirus gL infected Sf9 cells. A small fraction of the 31 kDa polypeptide was secreted into the extracellular medium as judged by Western blot analysis. The secreted form of gL was completely resistant to Endoglycosidase H (Endo-H), while the membrane associated form of gL was only partially resistant to Endo-H treatment, suggesting that the secreted gL represented a subpopulation of the membrane bound gL. Mice vaccinated with baculovirus expressed gL produced serum antibodies that reacted with authentic HSV-1 gL. However, these mice produced no HSV-1 neutralizing antibody (titer <1: 10) and they were not protected from lethal intraperitoneal or lethal ocular challenge with HSV-1. Thus, when used as a vaccine in the mouse model, gL, similar to our findings with HSV-1 gH, but unlike our results with the other 6 HSV-1 glycoproteins that we have expressed in this baculovirus system, did not provide any protection against HSV-1 challenge.

Collaboration


Dive into the Susan M. Slanina's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Homayon Ghiasi

University of California

View shared research outputs
Top Co-Authors

Avatar

Guey Chuen Perng

National Cheng Kung University

View shared research outputs
Top Co-Authors

Avatar

H. Ghiasi

Cedars-Sinai Medical Center

View shared research outputs
Top Co-Authors

Avatar

Ada Yukht

Cedars-Sinai Medical Center

View shared research outputs
Top Co-Authors

Avatar

Ravi Kaiwar

Cedars-Sinai Medical Center

View shared research outputs
Top Co-Authors

Avatar

Shlomo Melmed

Cedars-Sinai Medical Center

View shared research outputs
Top Co-Authors

Avatar

Guey-Cheun Perng

Cedars-Sinai Medical Center

View shared research outputs
Top Co-Authors

Avatar

Steve Cai

Cedars-Sinai Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge