Preprint
Review

A Journey through the Minefield of the Discovery and Characterization of LR-RNA/LAT

Altmetrics

Downloads

57

Views

34

Comments

0

A peer-reviewed article of this preprint also exists.

This version is not peer-reviewed

Submitted:

05 September 2024

Posted:

06 September 2024

You are already at the latest version

Alerts
Abstract
Scientific knowledge evolves in small steps, with occasional backsteps to correct inaccuracies, all occurring within a competitive environment. This perspective for the first time looks at the history of latency-related RNA (LR-RNA) that was later renamed latency-associated transcript (LAT). At the 1986 International Herpesvirus Workshop (IHW) meeting in Leeds, England, Daniel L Rock and Anthony B Nesburn first reported the discovery of human herpes virus 1 (HSV-1) latency-related (LR)-RNA that is antisense to ICP0. Less than a month after the IHW meeting a paper was submitted to Science magazine and 8 months later appeared in print thanking “D. Rock for suggesting RNA complementary to the ICP0 message may be present in latently infected cells”. This perspective is not a review of LAT literature but intends to clarify the timeline of LAT discovery and subsequent breakthroughs such as reactivation, apoptosis, CD8+ T cell exhaustion, and LAT expression in different cell types detected during latency. While many review articles have been written about LAT since 1987, the most comprehensive and balanced review about LAT was written by Dr. David Bloom’s group (1). In this overview, I will discuss our original collaboration with Dr. Dan Rock and subsequent work that our group performed, which is still ongoing. Finally, I will discuss the controversies associated with LAT from its inception to current times.
Keywords: 
Subject: Biology and Life Sciences  -   Virology

Introduction

Herpes simplex virus (HSV) families are broadly studied, and Bernard Roizman continues to be the undisputed “father” of herpes virology. Regarding HSV latency and the subject of this perspective, Jack Stevens and colleagues were primary players in the field of neurovirulence and latent infection using classical virological approaches (2-5) . However, in the early 1980s and during the dawn of molecular biology, Nigel Fraser and associates were the first to use modern molecular biology techniques to understand HSV-1 latency (6-8).
Part 1: First reported detection latency-related RNA (LR-RNA) in trigeminal ganglia (TG) of latently infected rabbits. After joining North Dakota State University as an assistant professor and in collaboration with Anthony Nesburn, Dan Rock presented an abstract at the 11th International Herpesvirus Workshop (IHW) meeting in Leeds England (July 21-26, 1986). A copy of this abstract is shown below, describing the first reported detection of LR-RNA in TG of latently infected rabbits and this transcript was antisense to ICP0:
Preprints 117453 i001
The above abstract and poster presented at the IHW meeting clearly demonstrated that LR-RNA (now named LAT) is antisense to ICP0, and this study was later published in Journal of Virology (9). However, prior to publishing this study, Stevens et al. (10) published a paper in Science reporting that an RNA complementary to a herpesvirus alpha gene mRNA is prominently expressed in latently infected neurons. In their acknowledgment they thanked Dan Rock for “suggesting that RNA complementary to the ICP0 message may be present in latently infected cells” as shown in this statement copied from the Stevens et al. paper (10).
Preprints 117453 i002
The abstract and poster that Dan Rock presented at the 1986 IHW, as well as his published study (9), clearly indicated that LR-RNA is antisense to ICP0 using single-stranded DNA probes prepared from recombinant M13 clones containing HSV-1 fragments (see D. Rock abstract, above). The most intriguing part of the Stevens’ Science paper was that the 11th IHW ended on July 26th and the Stevens paper was submitted on August 25, 1986 (31 days after the end of the IHW meeting) (see submission date of the 1987 Stevens’ Science paper above). Stevens et al. (10) indicated that they cloned a fragment of HSV-1 into M13 phage vectors, which they named mp8 and mp9, each expressing one strand of the ICP0 region. Thus, in 31 days, Stevens et al. (10) constructed the insert and cloned it into two phage plasmids, prepared single-stranded DNA, performed, and replicated experiments, wrote the manuscript, and submitted it to Science.
Part 2: Role of LAT in apoptosis. We previously reported that significantly higher levels of apoptosis were detected in rabbits acutely infected with a LAT-null mutant virus (dLAT2903) than in rabbits similarly infected with the parental WT HSV-1 McKrae strain (11). This study also demonstrated LAT inhibited apoptosis in transient transfection studies, which indicates LAT directly impairs apoptosis. Our paper reporting this result was published in Science in 2000. One group wrote to Science to complain that our work was not correct. Science published this letter, which can be found in a search of PubMed (12), while our response letter is not available on PubMed but did appear at the end of their letter. In subsequent years, multiple groups have confirmed that LAT has anti-apoptotic functions (13-18). Arguments that LAT is unable to inhibit apoptosis were further proven by generating recombinant viruses expressing cellular anti-apoptosis genes in place of LAT and these viruses restored wt reactivation (19, 20).
Because these cellular genes have only one function, they inhibit apoptosis, this provides compelling evidence that LAT does inhibit apoptosis and this function is important for reactivation. Ultimately, the same group that claimed LAT is not involved in blocking apoptosis (12), reported that LAT promotes neuronal survival (14). Thus, it appears that for some, “half-empty” is different from “half-full”!
Another neurotropic herpesvirus, bovine herpesvirus 1 (BoHV-1), encodes a LAT-like transcript (LR gene) that is abundantly expressed during latency, which encodes a protein (ORF2) that inhibits apoptosis in transient transfection assays (21, 22). Additional studies revealed a mutation where ORF2 was mutated did not reactivate from latency in calves and this mutant induced higher levels of TG neurons that underwent apoptosis (23, 24). Finally, when the wt LR gene was inserted into a LAT null mutant (dLAT2903) restored wild-type levels of reactivation from latency (25, 26).
Part 3: The role of LAT in CD8+ T cell exhaustion. In a groundbreaking 2006 study, Rafi Ahmed and colleagues demonstrated the role of PD-1/PD-L1/PD-L2 in CD8+ T cell exhaustion (27). The exhausted CD8+ T cells had significantly reduced function, which is consistent with CD8+ T cell proliferation and exhaustion in response to continued long-term exposure to T cell specific antigens (28-30). Based on these studies, we too asked if, like LCMV, continuous LAT expression during latency influenced T cell exhaustion in TG of latently infected mice. We included our preliminary data in our grant renewal and the primary reviewer of our grant wrote two pages describing how our data is counter to their published studies suggesting a role for CD8+ T cells in maintaining latency (31, 32). However, we and others later used LAT(+) and LAT(-) viruses to show that the presence of LAT leads to the generation of dysfunctional CD8+ T cells in TG of latently infected mice (33, 34). Our published study using LAT(+) and LAT(-) viruses as well as PD-1-/, PD-L1-/-, and PD-L2-/- mice clearly demonstrated that LAT expression contributes to CD8+, but not CD4+, T cell exhaustion (33). Later, Benmohamed’s group used LAT(+) and LAT(-) viruses and extensive FACS analysis to confirm our results, showing LAT contributes to CD8+ T cell exhaustion (34). Two years later another group, confirmed both our studies and those of Benmohamed (35).
Part 4: Involvement of CD8α+ DCs and not CD8+ T cell in HSV-1 latency. In the above studies we and others clearly demonstrated that, consistent with the pioneering study using LCMV, CD8+ T cells are not completely functional in vivo (27, 33, 34). However, between 2000 and 2003 several papers were published showing that CD8+ T cells infiltrate into TG at the time of HSV-1 establishment of latency and inhibit reactivation from latency (31). Furthermore, a subset of CD8+ T cells remain in direct contact with infected neurons (32). Using ex vivo cultures of latent TG, the authors showed that CD8+ T cells can block HSV-1 reactivation from latency (31, 32). These reports showed the importance of CD8+ T cells in maintaining latency using ex vivo studies while published studies of ours and others suggested that CD8+ T cells are not functional in vivo. Thus, to evaluate the contribution of CD8+ T cells in maintaining latency we used CD8α-/-, CD8β-/-, and β2M-/- mice and demonstrated that CD8+α DCs, but not CD8+ T cells, promote maintenance of latency (36, 37). Overall, detection of infiltrates in TG of latently infected mice most likely indicates subclinical or unsuccessful reactivation by the virus when low levels of lytic cycle proteins are expressed. Regardless of which scenario is correct, CD8+α DCs, and not CD8+ T cells, play an essential role in regulating maintenance of latency. The discrepancy between our results and those of other studies, regarding the importance of CD8+α DCs versus CD8+ T cells in maintaining latency is probably due to the use of anti-CD8 antibody in ex vivo studies. Anti-CD8 antibody detects CD8α cells, thus depleting both CD8+α DCs and CD8+ T cells. However, our studies using a combination of knockout mice and transfer experiments that clearly demonstrated the importance of CD8+α DCs and not CD8+ T cells.
It was previously reported that blocking reactivation from latency in ex vivo TG cultures by CD8+ T cells is mediated at least in part by gamma interferon (IFNγ) (38). The authors showed that neutralization of IFNγ significantly enhanced the rate of HSV-1 reactivation from latency in ex vivo TG cultures. In contrast to this study, we previously did not detect any differences in latency levels using IFNγ or a recombinant HSV-1 expressing IFNγ (HSV-IFNγ) under control of the LAT promoter (39-41). Replication of HSV-IFNγ was wild type in tissue culture and mouse eyes and its expression did not affect latency levels despite IFNγ expression during latency. Thus, our published studies using knockout mice and a recombinant virus continuously expressing IFNγ in TG of latently infected mice rule out the involvement of IFNγ in latency.
We were the first group to show the presence of LAT in TG isolated from cadavers (42). Based on the role of CD8+ T cells in maintaining latency in mice (32), the presence of CD8+ T cells in TG of cadavers was later reported using human TG (43). However, these CD8+ T cells may be: 1) migratory due to the absence of perfusion; 2) associated with reactivation as shown in rabbit or subclinical reactivation as shown in mice; 3) associated with status of the individual before death; 4) trauma associated with death that is more severe than reactivation associated with stress or sun exposure; or 5) presence of other infectious agents (i.e., viral, bacterial etc.), also the prolonged time before the TG were obtained from cadavers could have complicated these studies. Thus, except for the relevant presence of HSV LAT or DNA in human TG, detection of infiltrates may not indicate what really happens during latency. In most published studies, different groups examined the role of CD8+ T cells in maintaining latency by FACS or IHC. Ironically, these studies used an anti-CD8α antibody that detects both CD8α+ DCs and CD8α+ T cells.
Part 5: Detecting LAT in immune cells. Previous studies using IHC, EM, and in situ nucleic acid hybridization showed that neurons of latently infected mice, rabbits, and humans express LAT (9, 42, 44-46). After the first report detecting LR-RNA in TG of latently infected rabbits in 1986, LAT/LR-RNA was generally believed to be the only HSV-1 transcript detected in TG of latently infected hosts. However, the development of more sensitive molecular techniques enabled detection of small amounts of other viral gene products in neurons of latently infected mice (47-49). We also showed that TG from LAT(+) infected mice had significantly higher viral antigen levels than LAT(-) TG during latency (33). These findings suggest either low level expression of viral antigens in a small number of neurons during latency, or that subclinical or abortive reactivations occur in mice at very low levels. These “subclinical” reactivation episodes could be due to virus reactivation in neurons of latently infected mice and/or other cell types in TG of latently infected mice. We tested this hypothesis by isolating CD45+ cells from TG of latently infected mice and using RT-PCR to compare LAT expression in these cells with neurons of latently infected mice (50). In this study, we detected LAT in immune cells, but at lower levels than in neurons of latently infected mice. Based on RT-PCR results and single-cell analysis, we examined various immune cells in TG of latently infected mice, showing the presence of LAT in B cells, DCs, fibroblasts, glial cells, innate lymphoid cells (ILCs), macrophages, microglia, monocytes, NK cells, neurons, neutrophils, CD4-CD8-, CD4+, and CD8+ T cells (50). Due to the uniqueness and novelty of our findings and our previous experience with unfounded accusations that LAT does not play a role in apoptosis or CD8+ T cell exhaustion, we performed a second set of single-cell transcriptomic experiments on TG of latently infected mice and obtained the same results. Previous studies using single-cell analysis, typically perform one experiment rather than multiple experiments because the number of cells used for analysis is a crucial factor (51-55). Thus, even though the normal standard is single-cell RNA-sequencing (sncRNAs) analysis, we performed a second sncRNAs analysis. Our Science Advances paper provides solid evidence that gB and gD DNA are present in CD4+, CD8+, DC, and ILC cells (50). Consistent with our findings, a 2022 paper published in the Cell Press Journal, Neuron, from a Harvard group detected HSV-1 LAT in immune cells and fibroblasts of human TG (56). Our published study in mice (50) and Yang’s study in humans (56) used LAT as marker of latent TG and we identified infected cells based on LAT expression. However, many isolated cells may be infected but do not express LAT, especially non-neuronal cells as previous reports showed that these cells establish latency in the presence of Acyclovir but do not express LAT (57).
Not to our surprise and with the nature of working with LAT, even before the ink had dried on our paper, a group contacted Science Advances questioning the validity of our results without performing any experiments but rather, based on questionable reanalysis of our RNA-seq data. This group’s concern with our detection of LAT in immune cells, as I was told by one of the authors of the Science Advances inquiry, was that our work would negatively impact their research. The authors of this inquiry failed to consider our data showing LAT RNA detection in isolated immune cells or the presence of gB and gD DNA in isolated CD4+, CD8+, DC+, and ILCs cells. They also ignored a publication showing the detection of HSV-1 LAT in human TG (56). Our Science Advance paper came online January 23, 2023. Reasonable approaches to prove or disprove our work and by extension the work of Yang et al. (56), would have been to repeat our experiments and independently test whether our studies are reproducible. The group that contested our results had over a year to repeat our experiments many times over however, their goal was not to prove our work by repeating our work. Although scientists do not always agree, this group could have performed the experiments and presented their data to support or negate our results rather than misrepresenting our work in the absence of alternative evidence. These “scientists” spent more than a year Journal-shopping to publish their opinion without experimental evidence. As history and current times have shown, people who lack new ideas, dislike “new ideas”, as we have seen in different forms and at different times.
Part 6: Detecting reactivation in isolated immune cells. The main characteristics of HSV-1 infection are: 1) establishment of latency; and 2) during the latent phase of infection, and in contrast to primary infection, LAT is the only gene product expressed abundantly in infected mice, rabbits, and humans (9, 42, 44-46). Although additional HSV-1 transcripts have been detected, they are expressed at significantly lower levels than LAT transcripts (47, 48, 58). The HSV-1 LAT gene is known to play a critical role in enhancing the reactivation phenotype (44, 59-61) and LAT deletion mutants have a significantly lower reactivation phenotype in mice and rabbits (44, 59-61). Although spontaneous reactivation occurs in rabbits at levels similar to that seen in humans (62). Spontaneous reactivation in a murine model of ocular HSV-1 is rare. Following ocular infection, latent HSV genomes express LAT in a portion of those neurons maintaining them, and virus can be recovered by co-cultivating explanted ganglia (63-73). An interesting variation on this method, which comes closer to an in vivo method, has been developed using virus that can be reactivated in latently infected mice after transient exposure to UV-B radiation (74) or hyperthermia (75).
To determine if reactivation occurs in isolated immune cells from infected TG, mice were infected with HSV-1 strain McKrae, as we described previously (50). On day 28 post-infection, infected TG were isolated, and TG from four mice were pooled, digested with collagenase D, and dissociated into single-cell suspensions. The cells were then separated using a Percoll density gradient. The top cell layer, containing neurons, was discarded while the middle layer, containing immune cells, was collected and co-cultured with Vero cells to detect CPE as a measure of reactivation. Of the four samples from 16 mice, all four were reactivated, demonstrating that HSV-1 can be reactivated from this cell layer in which neurons were removed by density gradient. We then used two separate approaches to measure reactivation in isolated immune cells from the TG of infected mice (unpublished data). 1) First, we determined which immune cells types can be reactivated, then stained cells from the middle layer of the density gradient with BV510-anti-CD45, BV421-anti-CD3, and 7-AAD. CD45+CD3+ T cells and CD45+CD3- non-T immune cells were sorted using a BD Aria 3 cell sorter. Each cell type was then co-cultured with Vero cells to assess HSV-1 reactivation. In two independent experiments (total of 19 mice, with 9 or 10 TG pooled for each group), no HSV-1 reactivation was seen in either the T cell group, or non-T cell group. The lack of reactivation in these sorted cells could be due to cell damage during sorting or insufficient cell numbers (approximately 20,000 cells per group). However, we previously detected LAT RNA, as well as LAT, gB, and gD DNA in isolated CD4, CD8, macrophage, and DC cells (50). Further investigation is needed to determine whether HSV-1 can be reactivated in specific immune cell types using a cell isolation column rather than FACS. Second, we isolated immune cells as described above using 15 mice (6 TG per sample). The collected middle layer cells were stained with anti-CD45 antibody, and positive cells were purified using beads per the STEMCELL Technologies protocol. Purified CD45+ cells were then co-cultured with Vero cells and monitored for 3-7 days for CPE. Four \of 5 (80%) samples reactivated in two independent experiments, indicating that HSV-1 can reactivate from purified immune cells. These results suggest that reactivation can occur in isolated immune cells, depending on how cells are isolated. Studies are in progress to determine reactivation in individual cell populations using density gradient and staining with each cell type. In the above explant reactivation of infected immune cells we made sure that the isolated immune cells did not contain neurons.
Part 7: In vitro versus in vivo models of latency and concerns about using neuronal cells to study the HSV-1 latency-reactivation cycle. After discovering LAT, Wilcox and Johnson (76) were the first to use an in vitro model of latency in the presence of acyclovir. Between the late 1980s and early 1990s, other groups used C1300, Neuro2A, PC12 and many other cell types to study HSV-1 latency in vitro in the presence of antivirals. With improved neuronal isolation and culturing techniques in late 2000, some groups began to again use the in vitro model of latency. Infection of neurons by HSV-1 produces low levels of virus and cell death unless acyclovir is added to cultures. However, acyclovir treatment causes gaps in replicating viral DNA, which may increase transcription and “reactivation” in latently infected neuronal cultures. Further, LAT is not abundantly expressed in most “latently” infected neuronal cell culture models of latency. This is a problem because it is well documented that LAT is required to establish and maintain latency in mouse and rabbit infection models (9, 42, 44-46). Only one published study has shown that LAT is important in neuronal models of latency using Lund human mesencephalic (LUHMES) cells in vitro (77). Most neuronal models of latency do not use sensory neurons and thus cannot be correlated with in vivo latency models. In contrast to in vitro latency models, in which infected cells die without antiviral treatment, in vivo, neurons survive infection and establish and maintain latent infection in the host surrounded by non-neuronal cells that play critical roles in signaling to neurons in vivo.
Non-neuronal cells are not present in in vitro neuronal cultures used to study latency, but infiltrating immune cells are readily detected in human and mouse latency models (50, 56). Non-neuronal cells secrete cytokines that impair reactivation from latency and impair viral gene expression to promote latency establishment and maintenance. The absence of these immune cells in neuronal models of latency reduces the biological relevance of these models. In one latency model, used by the Cliffe-Wilson groups, after induced reactivation, viral gene expression occurred 12–18-hour post reactivation (78), while in explant-induced reactivation, viral protein expression (ICP0, ICP4, and VP16) was detected within 8 hours after explant using IHC (79). Although it is unlikely that two phases of reactivation exist in vivo, it seems to occur in several in vitro models of latency (80, 81). Thus, detecting LAT in different immune cells from human TG is further proof that the in vitro model of latency in the presence of acyclovir or similar antiviral drug classes and in the absence of immune cells, is not consistent with the in vivo model of latency. Thus, use of these “neuronal models” are limited and do not reflect the complexity of in vivo situations, even in mouse models of infection. Of note, latency-like conditions can be established in any non-neuronal cultured cell lines when cultures are incubated with acyclovir (57). Although many articles have been published on different latency models, a detailed review of latency models was recently published by Dr. David Leib’s group (82). The authors concluded that different in vitro latency models are not consistent with, or relevant to, in vivo latency models. While in vivo models of latency may not be perfect, they are more biologically relevant than in vitro cultures of HSV-1 latency.
Conclusion: The above perspective summarizes 40 years of work related to LR-RNA/LAT with many of the ups and downs, controversies, backstabbing, and manipulation by the old and new guard. Thus, we should expect more of the same in the next 40 years or until the complexity of LAT functions are solved. After attending an NIH study section, a prominent viral immunologist commented that ‘herpes-virologists eat each other’ if you are not part of their club. We might also add that when it comes to herpes latency, some in the crowd smother competition with significant efforts to stop research funding. One example that I heard directly, along with another investigator, occurred during the 9th Annual Symposium of the Colorado α-herpesvirus Latency Symposium (CALS) held on May 8–11, 2019, in Vail, CO, where during a conference dinner, a permanent member of one of the two main virology study sections promised two individuals that they would help them with their grants. Both of these investigators’ grants were reviewed in the permanent member’s study section and were funded. I and another virologist looked at each other and could not believe what was publicly stated. The same individual aggressively approached an ad hoc reviewer in the presence of other reviewers asking why the ad hoc reviewer was so tough on the grant of one of the individual’s friends and if there was any room for negotiation. Even out of the spotlight, scientists do not discuss this type of issue so openly. We all thought this individual felt they were above the rules and did not need to comply with the ethics of science! Thus, the behavior of some herpes virologists recalls what Rosalind Franklin endured when she was denied recognition for her role in unraveling the structure of DNA. As with athletics, competition in science needs to be on a level playing field. It is often difficult to determine who was the first to discover critical issues in science and in the end, for humankind if not for history, it is not important. However, it is unfair and offensive when a small group succeeds in suppressing the input of outsiders, particularly in an environment where staying active requires robust support. Thus, when it comes to novel findings about LAT, a diligent literature review will establish, even to the most novice investigator, the timeline of discovery and reproducibly (or not) of findings to the relevant scientific community.

Acknowledgements

I like to thank many individuals that worked on different aspect of these studies from its inception and contributed to NIH-NEI fundings in the past 38 years. Special thanks to Dr. Dan L Rock as the father of LR-RNA/LAT and the late Steven L Wechsler and Oscar Perng for their significant contribution to the field of latency-reactivation, their friendship, and support.

References

  1. Phelan D, Barrozo ER, Bloom DC. 2017. HSV1 latent transcription and non-coding RNA: A critical retrospective. J Neuroimmunol 308:65-101. [CrossRef]
  2. Cook ML, Bastone VB, Stevens JG. 1974. Evidence that neurons harbor latent herpes simplex virus. Infect Immun 9:946-951. [CrossRef]
  3. Cook ML, Thompson RL, Stevens JG. 1986. A herpes simplex virus mutant is temperature sensitive for reactivation from the latent state: evidence for selective restriction in neuronal cells. Virology 155:293-296. [CrossRef]
  4. Gerdes JC, Marsden HS, Cook ML, Stevens JG. 1979. Acute infection of differentiated neuroblastoma cells by latency- positive and latency-negative herpes simplex virus ts mutants. Virology 94:430-441.
  5. Javier RT, Sedarati F, Stevens JG. 1986. Two avirulent herpes simplex viruses generate lethal recombinants in vivo. Science 234:746-748. [CrossRef]
  6. Rock DL, Fraser NW. 1983. Detection of HSV-1 genome in central nervous system of latently infected mice. Nature 302:523-525. [CrossRef]
  7. Rock DL, Fraser NW. 1985. Latent herpes simplex virus type 1 DNA contains two copies of the virion DNA joint region. J Virol 55:849-852. [CrossRef]
  8. Stroop WG, Rock DL, Fraser NW. 1984. Localization of herpes simplex virus in the trigeminal and olfactory systems of the mouse central nervous system during acute and latent infections by in situ hybridization. Lab Invest 51:27-38.
  9. Rock DL, Nesburn AB, Ghiasi H, Ong J, Lewis TL, Lokensgard JR, Wechsler SL. 1987. Detection of latency-related viral RNAs in trigeminal ganglia of rabbits latently infected with herpes simplex virus type 1. J Virol 61:3820-6. [CrossRef]
  10. Stevens JG, Wagner EK, Devi-Rao GB, Cook ML, Feldman LT. 1987. RNA complementary to a herpesvirus alpha gene mRNA is prominent in latently infected neurons. Science 235:1056-1059. [CrossRef]
  11. Perng GC, Jones C, Ciacci-Zanella J, Stone M, Henderson G, Yukht A, Slanina SM, Hofman FM, Ghiasi H, Nesburn AB, Wechsler SL. 2000. Virus-induced neuronal apoptosis blocked by the herpes simplex virus latency-associated transcript. Science 287:1500-3.
  12. Thompson RL, Sawtell NM. 2000. HSV latency-associated transcript and neuronal apoptosis. Science 289:1651. [CrossRef]
  13. Inman M, Perng GC, Henderson G, Ghiasi H, Nesburn AB, Wechsler SL, Jones C. 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 75:3636-46. [CrossRef]
  14. Thompson RL, Sawtell NM. 2001. Herpes simplex virus type 1 latency-associated transcript gene promotes neuronal survival. J Virol 75:6660-75. [CrossRef]
  15. Ahmed M, Lock M, Miller CG, Fraser NW. 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 76:717-29. [CrossRef]
  16. Hamza MA, Higgins DM, Feldman LT, Ruyechan WT. 2007. The latency-associated transcript of herpes simplex virus type 1 promotes survival and stimulates axonal regeneration in sympathetic and trigeminal neurons. J Neurovirol 13:56-66. [CrossRef]
  17. Branco FJ, Fraser NW. 2005. Herpes simplex virus type 1 latency-associated transcript expression protects trigeminal ganglion neurons from apoptosis. J Virol 79:9019-25. [CrossRef]
  18. Oh JJ, Jaggi U, Tormanen K, Wang S, Hirose S, Ghiasi H. 2024. The anti-apoptotic function of HSV-1 LAT in neuronal cell cultures but not its function during reactivation correlates with expression of two small non-coding RNAs, sncRNA1&2. PLoS Pathog 20:e1012307. [CrossRef]
  19. Jin L, Carpenter D, Moerdyk-Schauwecker M, Vanarsdall AL, Osorio N, Hsiang C, Jones C, Wechsler SL. 2008. Cellular FLIP can substitute for the herpes simplex virus type 1 latency-associated transcript gene to support a wild-type virus reactivation phenotype in mice. J Neurovirol 14:389-400. [CrossRef]
  20. Jin L, Perng GC, Carpenter D, Mott KR, Osorio N, Naito J, Brick DJ, Jones C, Wechsler SL. 2007. Reactivation phenotype in rabbits of a herpes simplex virus type 1 mutant containing an unrelated antiapoptosis gene in place of latency-associated transcript. J Neurovirol 13:78-84. [CrossRef]
  21. Ciacci-Zanella J, Stone M, Henderson G, Jones C. 1999. The latency-related gene of bovine herpesvirus 1 inhibits programmed cell death. J Virol 73:9734-40. [CrossRef]
  22. Shen W, Jones C. 2008. Open reading frame 2, encoded by the latency-related gene of bovine herpesvirus 1, has antiapoptotic activity in transiently transfected neuroblastoma cells. J Virol 82:10940-5. [CrossRef]
  23. Inman M, Lovato L, Doster A, Jones C. 2002. A mutation in the latency-related gene of bovine herpesvirus 1 disrupts the latency reactivation cycle in calves. J Virol 76:6771-9. [CrossRef]
  24. Lovato L, Inman M, Henderson G, Doster A, Jones C. 2003. Infection of cattle with a bovine herpesvirus 1 strain that contains a mutation in the latency-related gene leads to increased apoptosis in trigeminal ganglia during the transition from acute infection to latency. J Virol 77:4848-57. [CrossRef]
  25. Perng GC, Maguen B, Jin L, Mott KR, Osorio N, Slanina SM, Yukht A, Ghiasi H, Nesburn AB, Inman M, Henderson G, Jones C, Wechsler SL. 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 76:1224-35. [CrossRef]
  26. Mott KR, Osorio N, Jin L, Brick DJ, Naito J, Cooper J, Henderson G, Inman M, Jones C, Wechsler SL, Perng GC. 2003. The bovine herpesvirus-1 LR ORF2 is critical for this gene’s ability to restore the high wild-type reactivation phenotype to a herpes simplex virus-1 LAT null mutant. J Gen Virol 84:2975-85. [CrossRef]
  27. Barber DL, Wherry EJ, Masopust D, Zhu B, Allison JP, Sharpe AH, Freeman GJ, Ahmed R. 2006. Restoring function in exhausted CD8 T cells during chronic viral infection. Nature 439:682-7. 8. [CrossRef]
  28. Barber DL, Wherry EJ, Masopust D, Zhu B, Allison JP, Sharpe AH, Freeman GJ, Ahmed R. 2006. Restoring function in exhausted CD8 T cells during chronic viral infection. Nature 439:682-7. [CrossRef]
  29. Day CL, Kaufmann DE, Kiepiela P, Brown JA, Moodley ES, Reddy S, Mackey EW, Miller JD, Leslie AJ, DePierres C, Mncube Z, Duraiswamy J, Zhu B, Eichbaum Q, Altfeld M, Wherry EJ, Coovadia HM, Goulder PJ, Klenerman P, Ahmed R, Freeman GJ, Walker BD. 2006. PD-1 expression on HIV-specific T cells is associated with T-cell exhaustion and disease progression. Nature 443:350-4. [CrossRef]
  30. Mueller SN, Ahmed R. 2009. High antigen levels are the cause of T cell exhaustion during chronic viral infection. Proc Natl Acad Sci U S A 106:8623-8. [CrossRef]
  31. Liu T, Khanna KM, Chen X, Fink DJ, Hendricks RL. 2000. CD8(+) T cells can block herpes simplex virus type 1 (HSV-1) reactivation from latency in sensory neurons. J Exp Med 191:1459-66. [CrossRef]
  32. Khanna KM, Bonneau RH, Kinchington PR, Hendricks RL. 2003. Herpes simplex virus-specific memory CD8+ T cells are selectively activated and retained in latently infected sensory ganglia. Immunity 18:593-603. [CrossRef]
  33. Allen SJ, Hamrah P, Gate DM, Mott KR, Mantopoulos D, Zheng L, Town T, Jones C, von Andrian UH, Freeman GJ, Sharpe AH, Benmohamed L, Ahmed R, Wechsler SL, Ghiasi H. 2011. The role of LAT in increased CD8+ T cell exhaustion in trigeminal ganglia of mice latently infected with herpes simplex virus type 1. J Virol 85:4184-4197.
  34. Chentoufi AA, Kritzer E, Tran MV, Dasgupta G, Lim CH, Yu DC, Afifi RE, Jiang X, Carpenter D, Osorio N, Hsiang C, Nesburn AB, Wechsler SL, BenMohamed L. 2011. The herpes simplex virus 1 latency-associated transcript promotes functional exhaustion of virus-specific CD8+ T cells in latently infected trigeminal ganglia: a novel immune evasion mechanism. J Virol 85:9127-38. [CrossRef]
  35. St Leger AJ, Jeon S, Hendricks RL. 2013. Broadening the Repertoire of Functional Herpes Simplex Virus Type 1-Specific CD8+ T Cells Reduces Viral Reactivation from Latency in Sensory Ganglia. J Immunol 191:2258-65.
  36. Mott KR, Allen SJ, Zandian M, Konda B, Sharifi BG, Jones C, Wechsler SL, Town T, Ghiasi H. 2014. CD8a dendritic cells drive establishment of HSV-1 latency. PLoS One 9:e93444. [CrossRef]
  37. Mott KR, Gate D, Matundan HH, Ghiasi YN, Town T, Ghiasi H. 2016. CD8+ T Cells Play a Bystander Role in Mice Latently Infected with Herpes Simplex Virus 1. J Virol 90:5059-67. [CrossRef]
  38. Liu T, Khanna KM, Carriere BN, Hendricks RL. 2001. Gamma interferon can prevent herpes simplex virus type 1 reactivation from latency in sensory neurons. J Virol 75:11178-84. [CrossRef]
  39. Ghiasi H, Cai S, Perng GC, Nesburn AB, Wechsler SL. 2000. The role of natural killer cells in protection of mice against death and corneal scarring following ocular HSV-1 infection. Antiviral Res 45:33-45. [CrossRef]
  40. Ghiasi H, Osorio Y, Hedvat Y, Perng GC, Nesburn AB, Wechsler SL. 2002. Infection of BALB/c mice with a herpes simplex virus type 1 recombinant virus expressing IFN-g driven by the LAT promoter. Virology 302:144-154.
  41. Lee DH, Ghiasi H. 2018. An M2 Rather than a TH2 Response Contributes to Better Protection against Latency Reactivation following Ocular Infection of Naive Mice with a Recombinant Herpes Simplex Virus 1 Expressing Murine Interleukin-4. J Virol 92:e00051-18. [CrossRef]
  42. Wechsler SL, Nesburn AB, Watson R, Slanina S, Ghiasi H. 1988. Fine mapping of the major latency-related RNA of herpes simplex virus type 1 in humans. J Gen Virol 69:3101-6. [CrossRef]
  43. Verjans GM, Hintzen RQ, van Dun JM, Poot A, Milikan JC, Laman JD, Langerak AW, Kinchington PR, Osterhaus AD. 2007. Selective retention of herpes simplex virus-specific T cells in latently infected human trigeminal ganglia. Proc Natl Acad Sci U S A 104:3496-501. [CrossRef]
  44. Hill JM, Sedarati F, Javier RT, Wagner EK, Stevens JG. 1990. Herpes simplex virus latent phase transcription facilitates in vivo reactivation. Virology 174:117-125. [CrossRef]
  45. Stevens, JG. 1989. Human herpesviruses: a consideration of the latent state. Microbiol Rev 53:318-332. [CrossRef]
  46. Wechsler SL, Nesburn AB, Watson R, Slanina SM, Ghiasi H. 1988. Fine mapping of the latency-related gene of herpes simplex virus type 1: alternative splicing produces distinct latency-related RNAs containing open reading frames. J Virol 62:4051-4058. [CrossRef]
  47. Feldman LT, Ellison AR, Voytek CC, Yang L, Krause P, Margolis TP. 2002. Spontaneous molecular reactivation of herpes simplex virus type 1 latency in mice. Proc Natl Acad Sci U S A 99:978-83. [CrossRef]
  48. Kramer MF, Coen DM. 1995. Quantification of transcripts from the ICP4 and thymidine kinase genes in mouse ganglia latently infected with herpes simplex virus. J Virol 69:1389-1399. [CrossRef]
  49. Margolis TP, Elfman FL, Leib D, Pakpour N, Apakupakul K, Imai Y, Voytek C. 2007. Spontaneous reactivation of herpes simplex virus type 1 in latently infected murine sensory ganglia. J Virol 81:11069-74. [CrossRef]
  50. Wang S, Song X, Rajewski A, Santiskulvong C, Ghiasi H. 2023. Stacking the odds: Multiple sites for HSV-1 latency. Sci Adv 9:eadf4904. [CrossRef]
  51. Yan L, Yang M, Guo H, Yang L, Wu J, Li R, Liu P, Lian Y, Zheng X, Yan J, Huang J, Li M, Wu X, Wen L, Lao K, Li R, Qiao J, Tang F. 2013. Single-cell RNA-Seq profiling of human preimplantation embryos and embryonic stem cells. Nat Struct Mol Biol 20:1131-9. [CrossRef]
  52. Tirosh I, Izar B, Prakadan SM, Wadsworth MH, 2nd, Treacy D, Trombetta JJ, Rotem A, Rodman C, Lian C, Murphy G, Fallahi-Sichani M, Dutton-Regester K, Lin JR, Cohen O, Shah P, Lu D, Genshaft AS, Hughes TK, Ziegler CG, Kazer SW, Gaillard A, Kolb KE, Villani AC, Johannessen CM, Andreev AY, Van Allen EM, Bertagnolli M, Sorger PK, Sullivan RJ, Flaherty KT, Frederick DT, Jane-Valbuena J, Yoon CH, Rozenblatt-Rosen O, Shalek AK, Regev A, Garraway LA. 2016. Dissecting the multicellular ecosystem of metastatic melanoma by single-cell RNA-seq. Science 352:189-96.
  53. Trapnell C, Cacchiarelli D, Grimsby J, Pokharel P, Li S, Morse M, Lennon NJ, Livak KJ, Mikkelsen TS, Rinn JL. 2014. The dynamics and regulators of cell fate decisions are revealed by pseudotemporal ordering of single cells. Nat Biotechnol 32:381-386. [CrossRef]
  54. Gupta I, Collier PG, Haase B, Mahfouz A, Joglekar A, Floyd T, Koopmans F, Barres B, Smit AB, Sloan SA, Luo W, Fedrigo O, Ross ME, Tilgner HU. 2018. Single-cell isoform RNA sequencing characterizes isoforms in thousands of cerebellar cells. Nat Biotechnol. [CrossRef]
  55. Collier JL, Pauken KE, Lee CAA, Patterson DG, Markson SC, Conway TS, Fung ME, France JA, Mucciarone KN, Lian CG, Murphy GF, Sharpe AH. 2023. Single-cell profiling reveals unique features of diabetogenic T cells in anti-PD-1-induced type 1 diabetes mice. J Exp Med 220:e20221920. [CrossRef]
  56. Yang L, Xu M, Bhuiyan SA, Li J, Zhao J, Cohrs RJ, Susterich JT, Signorelli S, Green U, Stone JR, Levy D, Lennerz JK, Renthal W. 2022. Human and mouse trigeminal ganglia cell atlas implicates multiple cell types in migraine. Neuron 110:1806-1821 e8. [CrossRef]
  57. Cohen EM, Avital N, Shamay M, Kobiler O. 2020. Abortive herpes simplex virus infection of nonneuronal cells results in quiescent viral genomes that can reactivate. Proc Natl Acad Sci U S A 117:635-640. [CrossRef]
  58. Allen SJ, Rhode-Kurnow A, Mott KR, Jiang X, Carpenter D, Rodriguez-Barbosa JI, Jones C, Wechsler SL, Ware CF, Ghiasi H. 2014. Regulatory Interactions between Herpesvirus Entry Mediator (TNFRSF14) and Latency Associated Transcript (LAT) during HSV-1 Latency. J Virol 88:1961-1971.
  59. Leib DA, Bogard CL, Kosz-Vnenchak M, Hicks KA, Coen DM, Knipe DM, Schaffer PA. 1989. A deletion mutant of the latency-associated transcript of herpes simplex virus type 1 reactivates from the latent state with reduced frequency. J Virol 63:2893-2900. [CrossRef]
  60. Sawtell NM, Thompson RL. 1992. Herpes simplex virus type 1 latency-associated transcription unit promotes anatomical site-dependent establishment and reactivation from latency. J Virol 66:2157-2169. [CrossRef]
  61. Perng GC, Ghiasi H, Slanina SM, Nesburn AB, Wechsler SL. 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 70:976-84. [CrossRef]
  62. Willey DE, Trousdale MD, Nesburn AB. 1984. Reactivation of murine latent HSV infection by epinephrine iontophoresis. Invest Ophthalmol Vis Sci 25:945-950.
  63. Perng GC, Dunkel EC, Geary PA, Slanina SM, Ghiasi H, Kaiwar R, Nesburn AB, Wechsler SL. 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 68:8045-55. [CrossRef]
  64. Mott KR, Ghiasi H. 2008. Role of dendritic cells in enhancement of herpes simplex virus type 1 latency and reactivation in vaccinated mice. Clin Vaccine Immunol 15:1859-67. Epub 2008 Oct 29. [CrossRef]
  65. Mott KR, UnderHill D, Wechsler SL, Ghiasi H. 2008. Lymphoid-related CD11c+CD8a+ dendritic cells are involved in enhancing HSV-1 latency. J Virol 82:9870-9879.
  66. Mott K, Brick DJ, van Rooijen N, Ghiasi H. 2007. Macrophages Are Important Determinants of Acute Ocular HSV-1 Infection in Immunized Mice. Invest Ophthalmol Vis Sci 48:5605-15. [CrossRef]
  67. Mott KR, Osorio Y, Brown DJ, Morishige N, Wahlert A, Jester JV, Ghiasi H. 2007. The corneas of naive mice contain both CD4+ and CD8+ T cells. Mol Vis 13:1802-12.
  68. Mott KR, Osorio Y, Maguen E, Nesburn AB, Wittek AE, Cai S, Chattopadhyay S, Ghiasi H. 2007. Role of anti-glycoproteins D (anti-gD) and K (anti-gK) IgGs in pathology of herpes stromal keratitis in humans. Invest Ophthalmol Vis Sci 48:2185-93.
  69. Mott KR, Perng GC, Osorio Y, Kousoulas KG, Ghiasi H. 2007. A Recombinant Herpes Simplex Virus Type 1 Expressing Two Additional Copies of gK Is More Pathogenic than Wild-Type Virus in Two Different Strains of Mice. J Virol 81:12962-12972. [CrossRef]
  70. Osorio Y, Mott KR, Jabbar AM, Moreno A, Foster TP, Kousoulas KG, Ghiasi H. 2007. Epitope mapping of HSV-1 glycoprotein K (gK) reveals a T cell epitope located within the signal domain of gK. Virus Res 128:71-80. [CrossRef]
  71. Mott KR, Bresee CJ, Allen SJ, BenMohamed L, Wechsler SL, Ghiasi H. 2009. Level of herpes simplex virus type 1 latency correlates with severity of corneal scarring and exhaustion of CD8+ T cells in trigeminal ganglia of latently infected mice. J Virol 83:2246-54. [CrossRef]
  72. Mott KR, Chentoufi AA, Carpenter D, Benmohamed L, Wechsler SL, Ghiasi H. 2009. The role of a glycoprotein K (gK) CD8+ T-cell epitope of herpes simplex virus on virus replication and pathogenicity. Invest Ophthalmol Vis Sci 50:2903-12. [CrossRef]
  73. Mott KR, Underhill D, Wechsler SL, Town T, Ghiasi H. 2009. A role for the JAK-STAT1 pathway in blocking replication of HSV-1 in dendritic cells and macrophages. Virol J 6:56. [CrossRef]
  74. Walker J, Laycock KA, Pepose JS, Leib DA. 1998. Postexposure vaccination with a virion host shutoff defective mutant reduces UV-B radiation-induced ocular herpes simplex virus shedding in mice. Vaccine 16:6-8. [CrossRef]
  75. Sawtell NM, Thompson RL. 1992. Rapid in vivo reactivation of herpes simplex virus in latently infected murine ganglionic neurons after transient hyperthermia. J Virol 66:2150-2156. [CrossRef]
  76. Wilcox CL, Johnson EM, Jr. 1987. Nerve growth factor deprivation results in the reactivation of latent herpes simplex virus in vitro. J Virol 61:2311-2315. [CrossRef]
  77. Edwards TG, Bloom DC. 2019. Lund Human Mesencephalic (LUHMES) Neuronal Cell Line Supports Herpes Simplex Virus 1 Latency In Vitro. J Virol 93:e02210-18. [CrossRef]
  78. Cliffe AR, Wilson AC. 2017. Restarting Lytic Gene Transcription at the Onset of Herpes Simplex Virus Reactivation. J Virol 91:e01419-16. [CrossRef]
  79. Harrison KS, Zhu L, Thunuguntla P, Jones C. 2019. Antagonizing the Glucocorticoid Receptor Impairs Explant-Induced Reactivation in Mice Latently Infected with Herpes Simplex Virus 1. J Virol 93:e00418-19. [CrossRef]
  80. Kobayashi M, Kim JY, Camarena V, Roehm PC, Chao MV, Wilson AC, Mohr I. 2012. A primary neuron culture system for the study of herpes simplex virus latency and reactivation. J Vis Exp. [CrossRef]
  81. Hu HL, Srinivas KP, Mohr I, Huang TT, Wilson AC. 2020. Using Primary SCG Neuron Cultures to Study Molecular Determinants of HSV-1 Latency and Reactivation. Methods Mol Biol 2060:263-277.
  82. Canova PN, Charron AJ, Leib DA. 2024. Models of Herpes Simplex Virus Latency. Viruses 16:747. [CrossRef]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.
Copyright: This open access article is published under a Creative Commons CC BY 4.0 license, which permit the free download, distribution, and reuse, provided that the author and preprint are cited in any reuse.
Prerpints.org logo

Preprints.org is a free preprint server supported by MDPI in Basel, Switzerland.

Subscribe

© 2024 MDPI (Basel, Switzerland) unless otherwise stated