ABSTRACT
Bovine herpesvirus 1 (BoHV-1), an important pathogen of cattle, establishes lifelong latency in sensory neurons within trigeminal ganglia (TG) after acute infection. The BoHV-1 latency-reactivation cycle, like other alphaherpesvirinae subfamily members, is essential for viral persistence and transmission. Notably, cells within pharyngeal tonsil (PT) also support a quiescent or latent BoHV-1 infection. The synthetic corticosteroid dexamethasone, which mimics the effects of stress, consistently induces BoHV-1 reactivation from latency allowing early stages of viral reactivation to be examined in the natural host. Based on previous studies, we hypothesized that stress-induced cellular factors trigger expression of key viral transcriptional regulatory genes. To explore this hypothesis, RNA-sequencing studies compared viral gene expression in PT during early stages of dexamethasone-induced reactivation from latency. Strikingly, RNA encoding infected cell protein 4 (bICP4), which is translated into an essential viral transcriptional regulatory protein, was detected 30 min after dexamethasone treatment. Ninety minutes after dexamethasone treatment bICP4 and, to a lesser extent, bICP0 RNA were detected in PT. All lytic cycle viral transcripts were detected within 3 h after dexamethasone treatment. Surprisingly, the latency related (LR) gene, the only viral gene abundantly expressed in latently infected TG neurons, was not detected in PT during latency. In TG neurons, bICP0 and the viral tegument protein VP16 are expressed before bICP4 during reactivation, suggesting distinct viral regulatory genes mediate reactivation from latency in PT versus TG neurons. Finally, these studies confirm PT is a biologically relevant site for BoHV-1 latency, reactivation from latency, and virus transmission.
IMPORTANCE BoHV-1, a neurotropic herpesvirus, establishes, maintains, and reactivates from latency in neurons. BoHV-1 DNA is also detected in pharyngeal tonsil (PT) from latently infected calves. RNA-sequencing studies revealed the viral infected cell protein 4 (bICP4) RNA was expressed in PT of latently infected calves within 30 min after dexamethasone was used to initiate reactivation. As expected, bICP4 RNA was not detected during latency. All lytic cycle viral genes were expressed within 3 h after dexamethasone treatment. Conversely, bICP0 and the viral tegument protein VP16 are expressed prior to bICP4 in trigeminal ganglionic neurons during reactivation. The viral latency related gene, which is abundantly expressed in latently infected neurons, was not abundantly expressed in PT during latency. These studies provide new evidence PT is a biologically relevant site for BoHV-1 latency and reactivation. Finally, we predict other alphaherpesvirinae subfamily members utilize PT as a site for latency and reactivation.
KEYWORDS: bovine herpesvirus 1, infected cell protein 4 (ICP4), pharyngeal tonsil, stress-induced reactivation from latency
INTRODUCTION
Bovine herpesvirus 1 (BoHV-1), an alphaherpesvirinae subfamily member, causes respiratory disorders, conjunctivitis, genital infections, encephalitis, abortions, and multi-systemic fatal disease in neonates. Acute infection immunosuppresses cattle, which predisposes cattle to secondary bacterial colonization, severe pneumonia, and even death (1). Consequently, BoHV-1 is a cofactor of bovine respiratory disease complex (BRDC) (2, 3), which costs the US cattle industry approximately $540 million in direct costs and $5 billion in indirect costs each year (4). Furthermore, BoHV-1, including commercially modified live vaccines, can initiate reproductive complications in pregnant cows, including abortion (5).
During productive infection, herpesviruses exhibit a well-regulated temporal cascade of viral gene expression that is divided into 3 stages: immediate early (IE), early (E), and late (L), reviewed in (2). Three BoHV-1 proteins are encoded by immediate early (IE) mRNAs: infected cell protein 0 (bICP0), bICP4, and bICP22 (6–9). A BoHV-1 tegument protein, VP16, specifically transactivates IE promoters during productive infection (10, 11). BoHV-1 contains a promoter, immediate early transcription unit 1 (IEtu1), that controls IE expression of IE/2.9 and IE/4.2 mRNAs, which are generated by alternative splicing (6–8). IE/2.9 mRNA is translated into the bICP0 protein, and IE/4.2 mRNA is translated into the bICP4 protein (6–8). A separate E promoter drives subsequent expression of an early transcript that also encodes the bICP0 protein (6–8, 12). The bICP0 and bICP4 proteins are crucial for productive infection because they encode proteins that stimulate viral transcription and productive infection. Early, and late genes are generally organized as other alphaherpesvirinae subfamily members and have similar functions.
Following acute infection of oral, ocular, or nasal cavities, sensory neurons within trigeminal ganglia (TG) are an important site for BoHV-1 latency. Stressful stimuli, including environmental heat, transporting cattle long distances, and/or restriction of water and food, increases the incidence of BoHV-1 reactivation from latency in cattle, reviewed in (13–15). Notably, the synthetic corticosteroid dexamethasone (DEX), which mimics the effects of stress, consistently induces reactivation from latency (16–18). The only known BoHV-1 genes abundantly expressed in latently infected neurons are the latency related (LR) gene (15) and ORF-E (19), which is upstream of the LR gene. LR gene products establish and maintain neuronal latency in calves (20), in part because these products interact with cellular transcription factors (21–25), interfere with bICP0 protein synthesis (26), promote neuronal differentiation (27, 28), and inhibit apoptosis (24, 29). In TG neurons of latently infected calves, LR gene products promote latency by directly or indirectly enhancing expression of more than 100 cellular genes that encode proteins essential for the Wnt/β-catenin and Akt signaling pathways (22, 23). In contrast to dividing cells, these cellular signaling pathways promote neuronal survival, axonal growth and maintenance, and proper synaptic connections (30–36).
In addition to TG neurons, BoHV-1 DNA is consistently detected in pharyngeal tonsil (PT) of latently infected cattle (18, 37–39). In contrast to TG neurons, LR-RNA expression is only detected in PT of latently infected calves when nested PCR is performed, indicating this gene is not abundantly expressed (18). By 6 h after latently infected calves are treated with DEX to induce reactivation from latency, in situ hybridization revealed lytic cycle viral transcripts are detected (18), suggesting reactivation occurs in PT and not indirectly via TG neurons. The intensity of in situ hybridization and higher number of PT cells expressing viral transcripts increase as a function of time after DEX treatment, which correlates with detection of infectious virus from tonsil swabs (40). In addition to PT, BoHV-1 DNA is consistently detected in peripheral blood mononuclear cells (41), lymph nodes, and spleen where infectious virus is not readily detected (42). Viral DNA from other alphaherpesvirinae subfamily members, including Pseudorabies virus (43, 44), equine herpesvirus 4 (45), and canine herpesvirus 1 (46), is also detected in PT and other lymphoid tissue during latency. Furthermore, a report concluded HSV-1, but not HSV-2 or human cytomegalovirus DNA, is detected in a subset of people who underwent tonsillectomy or adenectomy because of chronic lymphoid hyperplasia without evidence of acute HSV-1 infections (47). As expected, this report also detected Epstein-Barr Virus in PT of these patients.
The objectives of this study were to identify viral and cellular genes expressed in PT during early stages of DEX-induced reactivation from latency. Using RNA-sequencing (RNA-seq), our studies revealed the bICP4 transcript was readily detected in PT within 30 min after DEX treatment but not prior to DEX treatment. These findings support the premise that PT is a biologically relevant site for BoHV-1 reactivation from latency and virus transmission. We predict PT is an important site for reactivation for other “neurotropic” herpesviruses.
RESULTS
Identification of viral transcripts expressed in PT during early stages of reactivation from latency.
RNA-seq studies were performed to identify viral genes expressed in PT during early stages of DEX-induced reactivation from latency. For these studies, male Holstein calves were infected with a wild type (wt) virulent BoHV-1 strain (Cooper) as described previously (20, 22); see Fig. 1A for schematic summarizing the calf studies. As expected, viral transcripts were not detected in uninfected calves: however, low levels of viral transcripts were detected during latency (calves not treated with DEX) (Fig. 1B). Notably, a region localized to the left terminus of the viral genome yielded low levels of transcripts during latency (Fig. 2A). All viral reads from this region during latency contained the repeat GCTCCTCCTCCCTC. Further inspection revealed these reads span the terminus but were not derived from a single transcript. The BoHV-1 HindIII K fragment spans the left hand terminus of the viral genome and exhibits size heterogeneity in viral strains because the GCTCCTCCTCCCTC repeat can be present at different copy numbers (48). Consistent with previous studies (18), the LR transcript was not readily detected in PT of latently infected calves. In stark contrast, the LR transcript is abundantly expressed during latency in TG neurons (16, 49).
FIG 1.
Schematic of infection and frequency of viral transcription identified in PT. (A) Summary of calf studies, including important milestones during these experiments. Abbreviations: minutes (‘), hours (H), and day (D). (B) Identification of viral reads and % of total viral reads during latency and the time points during DEX-induced reactivation from latency in PT of calves latently infected calves.
FIG 2.
Viral transcripts expressed in PT during latency and DEX-induced reactivation from latency. (A) Schematic of BoHV-1 genome and position of bICP0, bICP4, and LR gene. Frequency of viral genes during latency and reactivation from latency. During latency, a family of transcripts containing the denoted repeat were expressed. (B) Distribution of RNA reads within the bICP4 transcript across the bICP4 gene. (C) Distribution of RNA reads within the bICP0 transcript across the bICP0 gene.
Latently infected calves (60 days after infection but not treated with DEX) were given a single IV injection of water-soluble DEX to initiate reactivation from latency (Fig. 1A). Within 30 min after DEX treatment, the number of viral transcripts detected in PT were 300-fold higher relative to latency (Fig. 1B). Strikingly, nearly all induced viral transcripts mapped to the bICP4 gene at 30 min after DEX treatment (Fig. 2A; 30 m DEX) and 90 min after DEX treatment (90 m DEX). By 90 min after DEX treatment, bICP0 RNA was also detected at low levels. These proteins are predicted to be expressed because RNA reads span the bICP4 (Fig. 2B) and bICP0 (Fig. 2C) ORFs. At 30 and 90 min after DEX treatment, transcripts within the HindIII K fragment also increased. By 3 h after DEX treatment, there was more than a 1,000-fold increase in viral transcript abundance compared to latency (Fig. 1B), and these viral transcripts spanned the entire genome. Simultaneously bICP4 reads were reduced relative to the rest of the viral genome. The bICP4 protein stimulates early and late promoters but autoregulates its expression by impairing IEtu1 promoter activity (7, 50). These activities partially explain why bICP4 transcripts were reduced relative to other lytic cycle viral genes during early stages or DEX-induced reactivation. Collectively, these studies revealed DEX treatment of latently infected calves rapidly induced expression of bICP4 RNA in PT culminating in expression of all lytic cycle viral genes within 3 h after DEX treatment.
The bICP4 protein is expressed in PT during early stages of reactivation.
Immunohistochemistry (IHC) studies were performed to confirm the bICP4 protein was expressed during early stages of reactivation from latency. The bICP4 antibody used for this study is directed against two bICP4 peptides, and bICP4 specific antibodies were affinity purified. This antibody specifically recognizes the bICP4 protein in a Western Blot and TG neurons by IHC (51). An infected calf treated with DEX for 90 min was examined for bICP4 antibody staining and to test whether bICP4 expression was localized to specific PT regions. PT contain numerous lymphoid follicles (Fig. 3A: denoted by F). The epithelium directly surrounding follicles is referred to as reticular epithelium (RE) (52), which contains epithelial cells, Micro-fold cells, lymphocytes, macrophages, dendritic cells, intraepithelial vasculature, and a discontinuous basement membrane. Non-reticular epithelium (N-RE) is present between follicles, and is comprised of pseudostratified columnar or squamous epithelium, occasional non-epithelial cells, and vascular structures. Studies focused on the boxed area of PT in Fig. 3A revealed weak bICP4 staining in follicles and RE 90 min after DEX treatment (Fig. 3B). Similarly, in situ hybridization study detected BoHV-1 transcripts in follicles and reticular epithelium (18). A couple of cell clusters exhibited higher levels of bICP4+ cells (denoted by yellow arrows). While weak background staining of PT cells from latently infected cows was present, there were no clusters of cells in latently infected calves that were bICP4+.
FIG 3.
Detection of bICP4 protein expression in PT. IHC was performed using the bICP4 peptide specific IgG as described in the materials and methods. (A) 90’ DEX is PT from a calf treated with DEX for 90 min. F denotes a follicle, N-RE is the non-reticular epithelium, and RE denotes reticulum epithelium. (B) is a higher magnification of the boxed area in (A). (C) PT from a calf latently infected with BoHV-1 (L) and incubated with the bICP4 antibody. Black arrows denote cells weakly stained by the bICP4 antibody. Yellow arrows denote a cluster of PT cells stained by the bICP4 antibody.
Additional studies were performed to confirm bICP4 was expressed during early stages of reactivation. Three hours after DEX treatment of latently infected calves, immunofluorescence (IF) was used to detect bICP4 expression. Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) and then IF was performed using the bICP4 antibody (Fig. 4). The rational for initially performing TUNEL is lymphoid cells are prone to apoptosis following corticosteroid treatment, reviewed in (53). Furthermore, published studies correlated induction of apoptosis and reactivation of certain herpesviruses, including HSV-1 latently infected TG (54) and cell lines latently infected with Kaposi’s sarcoma-associated herpesvirus (HHV-8), HHV-6A, HHV-6B, or Epstein-Barr virus (55, 56). TUNEL+ cells were detected in 2 different latently infected calves treated with DEX for 3 h (Fig. 4A and C; black arrows). Furthermore, bICP4+ cells were readily detected by 3 h after DEX treatment (Fig. 4B and D; white arrows). Notably, most of the bright staining bICP4+ cells appeared to be adjacent to TUNEL+ PT cells. PT sections from latently infected calves did not contain discernible TUNEL+ cells (Fig. 4E) or bICP4+ cells (Fig. 4F). In summary, these studies revealed the bICP4 protein was readily detected by 3 h after DEX treatment and there was a correlation between proximity of cells undergoing programmed cell death and cells expressing bICP4.
FIG 4.
Detection of bICP4 protein expression and TUNEL+ cells in PT 3 h after DEX treatment. TUNEL and IF using the bICP4 antibody were performed as described in Materials and Methods. TUNEL studies are shown in (A), (C), and (E). IF is shown (B), (D), and (F). PT from 2 different calves were treated with DEX for 3 h (A) and (B), and (C) and (D). PT sections from a latently infected calf are shown in panels E and F. For all panels, magnification was 40X.
Detection of bICP0 and VP16 protein 6 h after DEX treatment.
bICP0 and VP16 protein expression was weakly detected by IHC at 3 h after reactivation (data not shown). Additional studies examined bICP0 and VP16 expression in PT 6 h after calves were treated with DEX. PT from latently infected calves treated with DEX for 6 h were used for these studies because in situ hybridization studies detected BoHV-1 transcripts in follicles and reticular epithelium (18). bICP0 (Fig. 5A) and VP16 (Fig. 5B) positive PT cells were readily detected 6 h after DEX treatment (Fig. 5). As expected, staining of bICP0 (Fig. 5C) and VP16 (Fig. 5D) was not readily detected in PT sections from calves latently infected with BoHV-1. In summary, these studies revealed that numerous PT cells expressed bICP0 and VP16 by 6 h after DEX, which supports RNA-seq studies indicating the entire lytic cycle program of viral gene expression was activated by DEX treatment.
FIG 5.
Expression of bICP0 and VP16 protein expression in PT during reactivation from latency. IHC was performed using the bICP0 or VP16 peptide specific IgG as described in the materials and methods. IHC was performed using PT sections from latently infected calves 6 h (H) after DEX treatment using the bICP0 peptide specific antibody (A) or VP16 peptide specific antibody (B). IHC was also performed with calves latently infected with BoHV-1 (no DEX treatment) using the bICP0 antibody (C) or VP16 antibody (D).
DEX treatment progressively induces cell death in PT during reactivation from latency.
To further explore programmed cell death during reactivation, TUNEL assays were performed using additional time points during DEX-induced reactivation and latency (Fig. 6). Low levels of TUNEL+ cells were detected in tonsil sections prepared from latently infected calves. Within 30 min after latently infected calves were treated with DEX (30’ DEX panel), less than a 2-fold increase in the number of TUNEL+ cells were detected. However, 3 h after DEX treatment (3 H DEX panel) an 11-fold increase in TUNEL+ cells was consistently observed in several sections from different calves. Staining of PT sections with a cleaved caspase 3 antibody detected weak staining at 3H after DEX treatment (data not shown) suggesting a subset of cells underwent apoptosis. This observation was supported by differential gene expression analyses, which demonstrated that apoptotic processes (P-value = 0.0010) and positive apoptotic processes (P-value 0.0037) in the Reactome pathways/processes (57, 58) (data not shown). In summary, these studies revealed DEX-induced higher levels of programmed cell death in PT within 3 h after, latently infected calves were treated with DEX, which correlated with expression of all viral genes in PT.
FIG 6.
Programmed cell death increases in PT following DEX treatment. TUNEL was performed on PT sections as described in the materials and methods. Samples were prepared from latently infected calves (latency), latently infected calves treated with DEX for 30 min (30’), or 3 h (3H). Black arrowheads denote TUNEL+ cells.
Identification of transcription factors stimulated in PT during early stages of reactivation from latency.
Since bICP4 was the first viral transcript detected during DEX-induced reactivation from latency, we hypothesized that cellular transcription factors stimulate bICP4 expression. Consequently, we identified differentially expressed genes (DEGs) at 30 min after DEX treatment compared to latency and specifically looked for DEGs that encode transcription factors. The rational for focusing on 30 min after DEX treatment is lytic cycle viral gene expression (bICP4) was readily detected suggesting differentially expressed cellular transcription factors at this time mediated bICP4 expression. The pituitary homeobox 1 (PITX1), the #1 most highly induced transcription factor 30 min after DEX treatment (Table 1), cooperates with GR to transactivate a simple promoter containing nuclear hormone receptor binding sites (59). Furthermore, this study demonstrated PITX1 did not cooperatively transactivate androgen, estrogen-α, estrogen-β, progesterone, or retinoic acid receptors suggesting PITX1 is specific for GR-mediated transactivation. The ovo-like zinc finger 1 (OVOL1) regulates epithelial differentiation during embryonic development in numerous tissues and also regulates stemness of cancer cells (60). Krüppel like factor 15 (KLF15) and Zinc finger and BTB domain containing 16 (ZBTB16), also known as the promyelocytic leukemia zinc finger protein (PLZF), are members of the KLF family of transcription factors and are DEGs in TG neurons of calves during stress-induced reactivation (61). Notably, GR and KLF15 form a feed-forward transcription loop (62, 63) and cooperatively transactivate the IEtu1 promoter (64) and increase bICP4 expression (7, 50). GR and KLF15 also cooperatively transactivate key HSV-1 promoters (ICP0, ICP4, and ICP27) and silencing KLF15 expression impairs productive infection (65–68). ZBTB16 stimulates BoHV-1 productive infection better than other stress-induced transcription factors identified in TG (69): however, the only viral promoter we have identified to be transactivated by ZBTB16 alone or with GR is the HSV-1 ICP4 promoter (68). The other induced transcription factors may mediate certain aspects of stress-induced reactivation from latency: however, their roles are not as clear as PITX1, ZBTB16, or KLF15. For example, Homeobox Protein Distal-Less 3 (DLX3), specifically binds DNA and is a member of the distal-less family of non-Antennapedia homeobox genes, which contributes to mammalian trophoblast functions (70, 71). Dual specificity phosphatase 26 (DUSP26) regulates the mitogen activated protein kinase (MAPK) signaling pathway, reviewed in (72), suggesting it mediates DEX-induced transcription. We predict a subset of cellular transcription factors upregulated by DEX directly or indirectly stimulate lytic cycle viral gene expression during early stages of reactivation in PT.
TABLE 1.
Top 20 list of transcription factors increased within 30 min after latently infected calves were treated with DEX. For details, see text
| Gene | Fold-Change (log2) | p-value |
|---|---|---|
| PITX1 | 2.95522 | 5e-05 |
| OVOL1 | 2.57038 | 5e-05 |
| DUSP26 | 2.23067 | 5e-05 |
| YBX2 | 1.80612 | 0.0001 |
| ARSE | 1.66924 | 5e-05 |
| ZBTB16 | 1.62876 | 0.0007 |
| TFCP2L1 | 1.48281 | 0.0003 |
| NFE2 | 1.38612 | 0.00085 |
| DLX3 | 1.36805 | 0.00455 |
| KLF15 | 1.33602 | 5e-05 |
| RARG | 1.30147 | 5e-05 |
| MYBPH | 1.28599 | 0.0006 |
| MYBPC1 | 1.22954 | 5e-05 |
| PLEK2 | 1.2231 | 0.00105 |
| ARHGEF19 | 1.21883 | 5e-05 |
| HOPX | 1.21118 | 5e-05 |
| CARD14 | 1.17831 | 0.0005 |
| IRX3 | 1.17204 | 0.0002 |
| TUNAR | 1.09796 | 0.00295 |
| ELF3 | 1.05685 | 5e-05 |
Identification of transcription factors with reduced expression in PT during early stages of reactivation from latency.
The ligand dependent nuclear receptor corepressor like (LCoRL) gene was the #1 downregulated cellular transcription factor at 30 min after DEX treatment (Table 2). LCoRL is a nuclear protein reported to impair agonist-activated GR (69). LCoRL interacts with certain histone deacetylases and C-terminal binding protein (CtBP) corepressors and recruits this complex to GREs or GR bound DNA. LCoRL expresses 5 to 7 isoforms, varying from 319 to 1904 amino acids and is strongly associated with body weight in livestock via interactions with NCAPG (non-SMC condensin I complex subunit G), reviewed in (73). NCAPG is a subunit of chromatin condensin1 and critical for mitotic chromatin condensation. Interestingly, 6 zinc finger proteins (ZC2HCA1, ZNF713, ZNF711, ZNF367, ZNF148, ZNF148) were downregulated in PT at 30 min after DEX treatment. Zinc finger proteins interact with RNA, DNA, poly-ADP-ribose, and/or proteins. Certain zinc finger transcription factors are linked with DEX-induced cell differentiation, reviewed in (74), which correlates with DEX-induced activation of keratinocyte differentiation pathways in PT (data not shown). We suggest that developmental changes in PT induced by DEX treatment (directly or indirectly) promotes viral reactivation from latency.
TABLE 2.
Top 20 list of transcription factors decreased within 30 min after latently infected calves were treated with DEX. For details, see text
| Gene | Fold-Change (log2) | p-value |
|---|---|---|
| LCORL | −1.96098 | 5e-05 |
| SPESP1 | −1.77104 | 0.00335 |
| LARP4 | −1.65137 | 5e-05 |
| ZC2HC1A | −1.59249 | 5e-05 |
| ZNF713 | −1.58335 | 5e-05 |
| MARCH7 | −1.45943 | 5e-05 |
| MIER1 | −1.44925 | 5e-05 |
| ZNF711 | −1.43753 | 5e-05 |
| AP1AR | −1.40514 | 5e-05 |
| PLEKHF2 | −1.35821 | 5e-05 |
| ZNF367 | −1.35634 | 5e-05 |
| ZNF148 | −1.32711 | 5e-05 |
| ARL6 | −1.31382 | 5e-05 |
| RERGL | −1.31376 | 0.00495 |
| HLTF | −1.30188 | 5e-05 |
| ZNF644 | −1.27672 | 5e-05 |
| KLF10 | −1.26841 | 5e-05 |
| MIS18BP1 | −1.26382 | 5e-05 |
| AEBP2 | −1.24726 | 5e-05 |
| IKZF5 | −1.19533 | 5e-05 |
La-related protein-4 (LARP4) interacts with poly(A)-binding protein (PABP) and mediates T cell activation by stabilizing NFκB1 mRNA levels, which culminates in stimulating expression of interleukin-2 and interferon gamma (75). Interestingly, BoHV-1 was previously shown to infect activated CD4+ T cells in culture (76) and in acutely infected calves (38). The membrane-associated RING-CH-type finger (MARCHF7) is an E3 ubiquitin ligase, reviewed in (77). Up-regulated ubiquitination genes are enriched in samples from 30 and 90 min after DEX treatment supporting MARCHF7 mediated ubiquitination of NLRP3 (NOD LRR- and pyrin domain containing protein 3), a critical mediator of an inflammasome activated by numerous pathogens, reviewed in (78). Mesoderm induction early response 1 (MIERI) expresses 2 major isoforms that recruit histone deacetylase 1 (HDAC1) to repress transcription (79). Furthermore, MIERI interacts with Creb-binding protein (CBP) and inhibits the histone acetyltransferase activity of CREB. KLF10 overexpression is sufficient to induce apoptosis in epithelial cells and lymphoid cells and can impair growth of tumors, reviewed in (80). In general, transcription factors downregulated by DEX during early stages of reactivation in PT repress transcription of genes that stimulate innate immune responses, inflammation, and cell death.
Summary of differentially expressed cellular signaling pathways during early stages of reactivation from latency in PT.
To better understand changes that occur in PT within 30 min after DEX treatment, we identified differentially expressed genes that regulate key cellular signaling pathways. At 30 min after DEX treatment, neutrophil degranulation was strongly induced in PT (Fig. 7). Neutrophils are present on the surface of PT (81), and are present in mucosal secretions of PT (82). Interestingly, corticosteroids prevent apoptosis in neutrophils and exhibit pro-inflammatory and anti-inflammatory effects, reviewed in (83, 84). While neutrophil degranulation may not have a direct effect on viral gene expression during early stages of reactivation from latency, it is predicted to impact the PT environment after DEX treatment.
FIG 7.
Reactome of immune system and neutrophil degranulation pathway. Overrepresentation chart of differentially expressed genes in PT at 30 min after latently infected calves were treated with DEX. Neutrophil degranulation exhibited a highly signficant increase compared to PT in latently infected calves. Activation of RAS in B-cells was also overrepresented. The color legend in the top right of the figure reflects P-values.
RhoV, a member of the Rho family of GTPases, is a family of small G proteins that belongs to the Ras superfamily of GTPases, reviewed in (85). Although it is not clear whether RhoV exchanges GDP or GTP, it is primarily bound to GTP. Rho signaling pathways are significantly overrepresented in PT 30 min after DEX treatment, which supports its role in stimulating cell death (Fig. 8). The exact role that Rho GTPases is currently being examined in the context of BoHV-1 viral replication and gene expression.
FIG 8.
Reactome of Rho GTPase signaling pathways. Overrepresentation chart of differentially expressed genes in PT at 30 min after latently infected calves were treated with DEX. Color legend reflecting P-values is shown in top right.
DISCUSSION
Stress, as mimicked by DEX treatment, rapidly induced bICP4 RNA expression in PT of latently infected calves. The HSV-1 ICP4 protein is essential for productive infection (86) because it activates early (E) and late (L) gene expression (87). ICP4 specifically binds many sites on the viral genome (88) and interacts with the TATA-binding protein and RNA polymerase II coactivators (89, 90) to stimulate E and L viral gene expression (87, 90, 91). There is also a correlation between the ability of ICP4 to stimulate viral transcription and increase histone dynamics (92). Since bICP4 is the functional analogue of HSV-1 ICP4, we predict bICP4 has similar functions and efficiently triggers BoHV-1 lytic cycle gene expression resulting in virus production and successful reactivation from latency. This prediction is supported by the finding that all viral genes were detected in PT at 3 h after DEX treatment and infectious virus is readily detected in PT swabs during DEX-induced reactivation (40).
The initial events that promote DEX-induced reactivation from latency (host and virus transcripts) in PT appear to be unique relative to TG (Fig. 9A). For example, bICP4 was readily detected in PT at 30 min after DEX treatment, which was prior to bICP0 and VP16 detection. Conversely, bICP0 and VP16 expression are readily detected in TG neurons within 1 h after latently infected calves are treated with DEX (93, 94). Furthermore, bICP4 and the other IE protein (bICP22) are detected after bICP0 and VP16 in TG neurons (51). Late viral gene expression was readily detected in PT by 3 h after DEX treatment whereas in TG 2 late viral proteins, glycoprotein C (gC) and gD, are not readily detected until 6 h after DEX treatment (93). Studies focused on identifying viral genes expressed in TG relied on IHC studies and viral specific antibodies because RNA-seq studies of whole TG did not readily detect lytic cycle viral gene expression. When compared to PT, we suggest only a subset of latently infected neurons express low levels of viral genes following DEX treatment.
FIG 9.
Schematic of reactivation from latency in PT versus TG and schematic of BoHV-1 IEtu1. (A) Summarizes the differences observed during reactivation from latency in TG versus PT. (B) BoHV-1 genome and location of unique long (UL) region, direct repeats (open rectangles), and unique short region (US). IE/4.2 mRNA encodes the bICP4 protein and IE/2.9 mRNA encodes the bICP0 protein. An IE promoter activates expression of IE/4.2 and IE/2.9 and is designated IEtu1 (black rectangle) (7, 8). E/2.6 is the early bICP0 mRNA and its expression is driven by the bICP0 early promoter (E pro; gray rectangle). bICP0 protein coding sequences are in Exon 2 (e2). An origin of replication (ORI) separates IEtu1 from IEtu2. IEtu2 promoter (IEtu2 pro) drives IE1.7 mRNA expression, which is translated into the bICP22 protein. Solid lines in IE/2.9, IE/4.2, and IE/1.7 are exons (e1, e2, or e3) and dashed lines introns. (C) Full length IEtu1 promoter showing direction of transcription (arrow) start site of IEtu1 transcription (black triangle), TATA box, binding site for VP16/Oct1 complex (TAATGARAT), and location of GRE#1 plus GRE#2. Additional transcription factor binding sites in the IEtu1 promoter are denoted. ETs-1 belongs to the ETS (erythroblast transformation-specific) transcription factor family. The alternative bICP4 (blue triangle) and bICP0 (green triangle) mRNA start sites are upstream of the initiating methionine (96, 97).
Based on studies in this report and previously published studies (51, 61, 93, 94), we predict different anatomic specific cellular factors quickly activate bICP4 expression in PT relative to TG. For example, PITX1 and LCoR were differentially expressed in PT but were not identified in TG during early stages of reactivation from latency (61) (Fig. 9A). Downregulation of LCoR, a GR transcriptional repressor, during early stages of reactivation in PT implies this is an important step during reactivation from latency in PT but not TG. LCoR is also predicted to directly participate in maintaining latency by repressing IEtu1 promoter activity. Two stress-induced transcription factors, KLF15 and ZBTB16, were differentially expressed in TG and PT suggesting these common factors mediate reactivation in TG and PT. However, KLF4 and Slug expression were induced in TG (61), but not PT, during DEX-induced reactivation from latency. Unlike reactivation from latency in PT, VP16 is readily detected in TG neurons during early stage of reactivation from latency (93, 94), suggesting VP16 plays a prominent role with respect to driving IE gene expression in TG, but not PT.
IEtu1 is a crucial BoHV-1 regulatory locus because it contains coding sequences for bICP0, bICP4 (6–8), and the LR gene (15). The finding that bICP4 was the most abundant viral gene expressed at 30 and 90 min after DEX treatment was unexpected because the IEtu1 promoter was initially reported to drive bICP4 and bICP0 expression from a common transcriptional start site under immediate early conditions during productive infection (6–8) (Fig. 9B; denoted by arrow). This initial study concluded the start site for the IETu1 transcript is 24–31 nucleotides downstream of the TATA box (black diamond). In contrast, a recent study concluded bICP0 and bICP4 contain distinct transcriptional start sites (Fig. 9C; bICP4 and bICP0 start sites are denoted by blue and green diamonds) (95). With respect to this recent study, the bICP0 RNA start site is 726 bases downstream from the TATA box in IETu1 and the bICP4 RNA start site is 826 bp downstream of the TATA box. Both RNA start sites are upstream of the initiating methionine residue reported for these proteins (denoted by ATG in Fig. 9C). Although there is not a consensus TATA box in close proximity to the bICP0 or bICP4 start sites, promoters lacking a TATA box have been described and the cellular transcription factor specificity protein 1 (Sp1) is linked to regulating transcription (96, 97). Three consensus Sp1 binding sites (CCCGCC) are downstream of the TATA box, and prior to the initiating methionine for bICP0 and bICP4. The only TAATGARAT motif (Oct-1 binding site) in the IEtu1 promoter region is upstream of the TATA box. Regardless of how bICP4 is selectively expressed during early stages of reactivation in PT, 2 functional glucocorticoid response elements (GREs) in the IEtu1 promoter/regulatory region (65, 98) are predicted to mediate stress-induced promoter activation of bICP4 RNA expression during early stages of reactivation from latency in PT. Although GRE#1 is 1,006 bases upstream of the TATA box (98), certain GREs can stimulate transcription when 5–19 kb from the start site of transcription (99).
The studies in this report and a previous study (18), revealed cells in PT follicles and reticular epithelium contain viral genomes and support viral gene expression during early stages of reactivation from latency. Several distinct cell types are present in PT follicles and reticular epithelium. For example, the follicle is generally comprised of lymphoid cells whereas reticular epithelium contains epithelial cells, Micro-fold cells, lymphocytes, macrophages, and dendritic cells. Micro-fold cells transport antigens to follicles; thus, initiating immune responses, reviewed in (100). Previous studies indicated BoHV-1 infects circulating CD4+ T cells during acute infection of calves suggesting CD4+ T cells in PT may be a site for BoHV-1 latency (38). However, a preliminary study suggests CD4+ and CD8+ T cells in PT of latently infected calves contained no viral DNA when compared to total PT suggesting infected T cells do not survive and are not the reservoir for latency (Harrison and Jones, unpublished data). Since acutely infected CD4+ T cells frequently undergo apoptosis (38), this finding was somewhat expected. Antibodies that specifically identify single populations of other bovine lymphocytes, epithelial cells, and Micro-fold cells will be used to identify specific cell population(s) that harbor viral DNA.
The LR gene is abundantly expressed in TG neurons during latency (49, 101, 102): however, this was not the case in PT as judged by RNA-seq and RT-PCR studies (18). This was not surprising because the LR promoter is regulated by neuronal specific factors (103–105) that are apparently absent or not abundantly expressed in PT. LR gene products have multiple functions designed to maintain latency: for example, impairing apoptosis, productive infection and bICP0 expression, Notch-mediated transcription, and stress-induced transactivation of IEtu1 promoter activity (24, 26, 29, 106–110). Hence, the absence of LR gene products in PT will not influence viral gene expression and the number of cells that successfully reactivate from latency following DEX treatment. The family of transcripts with the repetitive element expressed in PT of latently infected calves are not likely to express a protein and their putative role in the latency-reactivation cycle is currently unknown.
In conclusion, these studies provide compelling evidence that reactivation from latency occurs rapidly in PT following DEX treatment. Furthermore, reactivation from latency in PT may be more efficient than TG with respect to virus transmission following stressful stimuli. This statement is supported by the knowledge that reactivation from latency via TG neurons or other neurons requires viral particles to travel anterogradely from neurons toward nerve terminals and then reinfect dermis or other innervated cell types prior to virus transmission to another host. However, virus transmission during reactivation from latency in PT can occur directly because PT is at the base of the oral cavity.
MATERIALS AND METHODS
Infection of calves and DEX treatment to induce reactivation from latency.
For these studies, weaned male Holstein calves (~440 lbs.) were acclimated for 7 days prior to infection with BoHV-1 (Cooper strain, GenBank Z48053.1) as previously described (18, 38, 40, 111, 112). In brief, BoHV-1-free calves were randomly assigned and housed in isolation rooms (2-3 calves in each room) in a BSL2 facility for large animals. During the acclimation period, calves were treated with the antibiotic Liquamycin (oxytetracycline; Zoetis) using 4.5 mL for each 100 lbs. of the calf every 4 days. Calves were anesthetized with Xylazine (approximately 50 mg/50 kg body weight; Bayer Corp.). Calves were inoculated with 1 mL of a solution containing 1x107 PFU/mL of BoHV-1 in each nostril and eye, without scarification, for a total of 4x107 PFU per animal. Instillation of the virus in the ocular cavity was performed by adding 0.1 to 0.2 mL of virus and then closing the eyelid and rubbing gently to facilitate infection. Infectious virus in ocular and nasal swabs were monitored after infection to ensure acute infection occurred as previously observed (40, 111). Calves were treated with Liquamycin until 8 days after infection to prevent bacterial pneumonia. TG and PT were collected during latency (≥ 60 days after infection). Latently infected calves were injected intravenously (jugular vein) with 100 mg water-soluble DEX (Sigma) suspended in Minimal Essential Media (MEM) that did not contain antibiotics or fetal bovine serum to initiate reactivation from latency (18). Calves were then loaded onto a cattle trailer and transported to the University of Nebraska-Lincoln Veterinary Diagnostic Laboratory (approximately ¼ of a mile from the BSL-2 large animal facility to the Diagnostic Laboratory). Loading calves onto the trailer, transporting the calves, and unloading them took approximately 10 min. For the 30 min treatment with DEX calves, they were transported to the Diagnostic Lab, placed in a pen, and then injected with DEX. Prior to euthanasia by electrocution, calves were heavily sedated with Xylazine. It is unlikely that the stress of transporting calves ¼ of a mile to the diagnostic center initiated lytic cycle viral gene expression. This observation is supported by the finding that latently infected calves are not expressing detectable levels of lytic cycle viral genes in PT (Fig. 1, 4, and 5) using the same methods to transport these latently infected calves (no DEX treatment) from the large animal facility to the Diagnostic Lab. Other studies also revealed that lytic cycle viral genes are not detected in TG of latently infected calves unless DEX is given to these calves and transported as described above (22, 51, 61, 93, 94, 113). After decapitation, TG and PT were collected, and samples from each were formalin fixed and then paraffin embedded. The remainder of TG or pharyngeal tonsil were minced into small pieces, placed into 50 mL conical tubes, tubes placed in a dry ice ethanol bath, and stored at −80°C. Following decapitation, it took approximately 10 min to process tissues and submerge tubes containing TG or pharyngeal tonsil in a dry ice ethanol bath. All protocols were approved by the Institutional Animal Care and Use Committee of the University of Nebraska.
Preparation of total RNA from PT and RNA-seq.
PT were collected from 3 calves per time point in the study: latency, 30 min, 90 min, or 3 h after DEX treatment. Total RNA was prepared from approximately 2 g of tissue using TRIzol (Life Technologies) according to the manufacturer's instructions and as previously described (22). RNA integrity and concentration were quantified with a 5200 Fragment Analyzer System (Agilent Technologies). An Illumina TruSeq stranded mRNA library preparation kit was used to construct cDNA libraries, which were sequenced as 75-bp paired-end reads on an Illumina NextSeq 500 (Illumina) to the depth of approximately 100 million reads per library.
Read alignment of the RNA-seq data was carried out as previously described (22). In brief, raw paired-end sequence reads in individual fastq files were trimmed to remove adapter sequences and low-quality bases using Trimmomatic software (version 0.35) (114). The remaining reads were mapped to the UMD 3.1 genome assembly with Tophat2 (version 2.1.1) (115) using the National Center for Biotechnology Information (NCBI) Bos taurus and BoHV-1 reference annotation to guide the alignment. The default parameters for Tophat2 were used.
Cuffdiff software (version 2.2.1) (116) was employed to process the aligned sequence read files and test for differential gene expression. Transcript abundance was estimated for each transcript in the NCBI Bos taurus reference annotation (UMD3.1) or BoHV-1 reference genome for each sample. A gene was defined as being expressed provided it had an average fragment per kilobase of transcript per million mapped reads (FPKM) of 1.0 in at least 1 of the 2 groups in the comparison. Unexpressed genes were removed from further downstream analysis. Differential gene expression was detected by comparing the log (base 2) ratios of the FPKM values for every gene and transcript. The resulting P-values were corrected for multiple testing using Benjamini-Hochberg correction (117). P values were considered statistically significant at a Benjamini-Hochberg adjusted P value of ≤ 0.05.
Immunohistochemistry and immunofluorescence.
PT from latently infected or DEX treated calves were harvested and directly placed in formalin. Slide preparation and IHC was performed as previously described (22, 23, 118). Briefly, 4-5μm sections of paraffin embedded PT were cut and mounted onto glass slides. Slides were incubated at 65°C for ~20 min before deparaffinization in Xylene and serial rehydration using decreasing concentrations of Ethanol. Endogenous peroxidases were blocked by incubating slides in 0.045% H2O2 for 20 min. Antigen retrieval was performed using Proteinase K (Agilent Dako) and slides were blocked using Animal Free Blocking Solution (AFBS; Cell Signaling Technologies) for 45 min at room temperature (RT). Avidin and Biotin were blocked using Vector Labs Avidin/Biotin blocking kit as per manufacturer’s instructions (Vector Labs). Slides were incubated overnight at 4°C with the bICP0, VP16, or bICP4 peptide specific antibody in AFBS. These are peptide specific antibodies that specifically recognize these proteins and they were previously described (51). The following day, Vectastain ABC HRP Kit was used according to manufacturer’s instructions (Vector Labs). Biotinylated secondary antibody was prepared in AFBS, and slides were incubated for 30 min at RT. Slides were then developed with NovaRed substrate (Vector Labs) and lightly counterstained with Mayer’s hematoxylin (Sigma-Aldrich). Slides were imaged using an Olympus BX43 microscope and captured using CellSense software. The bICP0 or bICP4+ PT cells were examined in a blinded fashion.
For IF studies, slides were dual stained with bICP4 overnight at 4°C using a 1:200 dilution after completion of TUNEL assay as described below. The following day slides were washed and stained with a secondary Alexa 488 (Invitrogen, Catalog # A32731) diluted 5 μg/mL for 1 h at room temperature. All images were captured as described above using the 40 X objective. Brightfield images were obtained with autoexposure times ~0.01 s; fluorescent images were obtained with equal manual exposure of 0.1 s.
Detection of apoptosis in fixed tissue.
The TUNEL assay was performed to detect cells undergoing programmed cell death in PT tissue sections as described previously (17, 38). For these studies, the DeadEnd Colorimetric TUNEL System was used (Promega, catalogue # G7360). Positive control slides were included by treating with OPTIZYME DNase I (Fisher) at 5U/mL for 10 min at RT prior to TdT staining (data not shown).
Data availability.
The RNA-seq data was deposited into the NCBI GEO repository as series record GSE216685.
ACKNOWLEDGMENTS
All authors thank Sue Hauver for technical support. This research was supported by grants from the U.S. Department of Agriculture (USDA)-National Institute of Food and Agriculture (NIFA) Competitive Grants Program (2018-06668 and 2021-67015), Oklahoma Center for Respiratory and Infectious Diseases (National Institutes of Health Centers for Biomedical Research Excellence Grant # P20GM103648), National Institute of Neurological Disorders and Stroke of the National Institutes of Health under Award Number R01NS111167, and funds from the Sitlington Endowment.
Mention of trade names or commercial products in this publication is solely for the purpose of providing specific information and does not imply recommendation or endorsement by USDA or NIH. The USDA and NIH are an equal opportunity provider and employer.
Contributor Information
Clinton Jones, Email: clint.jones10@okstate.edu.
Felicia Goodrum, University of Arizona.
REFERENCES
- 1.Tikoo SK, Campos M, Babiuk L. 1995. Bovine herpesvirus 1 (BHV-1): biology, pathogenesis, and control. Adv Virus Res 45:191–223. 10.1016/S0065-3527(08)60061-5. [DOI] [PubMed] [Google Scholar]
- 2.Jones C, Chowdhury S. 2007. A review of the biology of bovine herpesvirus type 1 (BHV-1), its role as a cofactor in the bovine respiratory disease complex, and development of improved vaccines. Anim Health Res Rev 8:187–205. 10.1017/S146625230700134X. [DOI] [PubMed] [Google Scholar]
- 3.Hodgson PD, Aich P, Manuja A, Hokamp K, Roche FM, Brinkman FSL, Potter A, Babiuk LA, Griebel PJ. 2005. Effect of stress on viral-bacterial synergy in bovine respiratoryt disease: novel mechanisms to regulate inflammation. Comp Funct Genomics 6:244–250. 10.1002/cfg.474. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Johnson K, Pendell DL. 2017. Market impacts of reducing the prevalence of bovine respiratory disease in United States beef cattle feedlots. Front Vet Sci 4:Article 189. 10.3389/fvets.2017.00189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Chase C, Fulton RW, O'Toole D, Gillette B, Daly RF, Perry G, Clement T. 2017. Bovine herpesvirus 1 modified live vaccines for cattle reproduction: balancing protection with undesired effects. Vet Microbiol 206:69–77. 10.1016/j.vetmic.2017.03.016. [DOI] [PubMed] [Google Scholar]
- 6.Wirth UV, Gunkel K, Engels M, Schwyzer M. 1989. Spatial and temporal distribution of bovine herpesvirus 1 transcripts. J Virol 63:4882–4889. 10.1128/JVI.63.11.4882-4889.1989. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Wirth UV, Vogt B, Schwyzer M. 1991. The three major immediate-early transcripts of bovine herpesvirus 1 arise from two divergent and spliced transcription units. J Virol 65:195–205. 10.1128/JVI.65.1.195-205.1991. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Wirth UV, Fraefel C, Vogt B, Vlcek C, Paces V, Schwyzer M. 1992. Immediate-early RNA 2.9 and early RNA 2.6 of bovine herpesvirus 1 are 3' coterminal and encode a putative zinc finger transactivator protein. J Virol 66:2763–2772. 10.1128/JVI.66.5.2763-2772.1992. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Schwyzer M, Wirth UV, Vogt B, Fraefel C. 1994. BICP22 of bovine herpesvirus 1 is encoded by a spliced 1.7 kb RNA which exhibits immediate early and late transcription kinetics. J Gen Virol 75:1703–1711. 10.1099/0022-1317-75-7-1703. [DOI] [PubMed] [Google Scholar]
- 10.Misra V, Bratanich AC, Carpenter D, O'Hare P. 1994. Protein and DNA elements involved in transactivation of the promoter of the bovine herpesvirus (BHV) 1 IE-1 transcription unit by the BHV alpha gene trans-inducing factor. J Virol 68:4898–4909. 10.1128/JVI.68.8.4898-4909.1994. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Misra V, Walker S, Hayes S, O'Hare P. 1995. The bovine herpesvirus alpha gene trans-inducing factor activates transcription by mechanisms different from those of its herpes simplex virus type 1 counterpart VP16. J Virol 69:5209–5216. 10.1128/JVI.69.9.5209-5216.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Fraefel C, Zeng J, Choffat Y, Engels M, Schwyzer M, Ackermann M. 1994. Identification and zinc dependence of the bovine herpesvirus 1 transactivator protein BICP0. J Virol 68:3154–3162. 10.1128/JVI.68.5.3154-3162.1994. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Jones C, da Silva LF, Sinani D. 2011. Regulation of the latency-reactivation cycle by products encoded by the bovine herpesvirus 1 (BHV-1) latency-related gene. J Neurovirol 17:535–545. 10.1007/s13365-011-0060-3. [DOI] [PubMed] [Google Scholar]
- 14.Jones C. 2014. Reactivation from latency by alpha-herpesvirinae submfamily members: a stressful situation. Current Topics in Virology 12:99–118. 10.1128/JVI.00901-20. [DOI] [Google Scholar]
- 15.Jones C. 2016. Latency of bovine herpesvirus 1 (BoHV-1) in sensory neurons. In Omgradi J (ed), Herpesviridae pp. 237–261. In Tech, London England. [Google Scholar]
- 16.Rock D, Lokensgard J, Lewis T, Kutish G. 1992. Characterization of dexamethasone-induced reactivation of latent bovine herpesvirus 1. J Virol 66:2484–2490. 10.1128/JVI.66.4.2484-2490.1992. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Winkler MT, Doster A, Sur JH, Jones C. 2002. Analysis of bovine trigeminal ganglia following infection with bovine herpesvirus 1. Vet Microbiol 86:139–155. 10.1016/S0378-1135(01)00498-9. [DOI] [PubMed] [Google Scholar]
- 18.Winkler MT, Doster A, Jones C. 2000. Persistence and reactivation of bovine herpesvirus-1 (BHV-1) in tonsils of latently infected cattle. J Virol 74:5337–5346. 10.1128/jvi.74.11.5337-5346.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Inman M, Zhou J, Webb H, Jones C. 2004. Identification of a novel bovine herpesvirus 1 transcript containing a small open reading frame that is expressed in trigeminal ganglia of latently infected cattle. J Virol 78:5438–5447. 10.1128/jvi.78.10.5438-5447.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.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 76:6771–6779. 10.1128/jvi.76.13.6771-6779.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Liu Y, Hancock M, Workman A, Doster A, Jones C. 2016. Beta-catenin, a transcription factor activated by canonical Wnt signaling, is expressed in sensory neurons of calves latently infected with bovine herpesvirus 1. J Virol 90:3148–3159. 10.1128/JVI.02971-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Workman A, Zhu L, Keel BN, Smith TPL, Jones C. 2018. The Wnt signaling pathway is differentially expressed during the bovine herpesvirus 1 latency-reactivation cycle: evidence that two proteinkinases associated with neuronal survival, Akt3 and BMPR2, are expressed at higher levels during latency. J Virol 92:e01937-17. 10.1128/JVI.01937-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Zhu L, Workman A, Jones C. 2017. A potential role for a beta-catenin coactivator (high mobility group AT-hook 1 protein) during the latency-reactivation cycle of bovine herpesvirus 1. J Virol 91:e02132-16. 10.1128/JVI.02132-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Sinani D, Jones C. 2011. Localization of sequences in a protein encoded by the latency related gene of bovine herpesvirus 1 (ORF2) that inhibits apoptosis and interferes with Notch1 mediated trans-activation of the bICP0 promoter. J Virol 85:12124–12133. 10.1128/JVI.05478-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Meyer F, Jones C. 2009. The cellular transcription factor, CCAAT enhancer-binding protein alpa (C/EBP-a) has the potential to activate the bovine herpesvirus 1 immediate early transcription unit 1 promoter. J Neurovirol 15:123–130. 10.1080/13550280802534771. [DOI] [PubMed] [Google Scholar]
- 26.Jaber T, Workman A, Jones C. 2010. Small noncoding RNAs encoded within the bovine herpesvirus 1 latency-related gene can reduce steady-state levels of infected cell protein 0 (bICP0). J Virol 84:6297–6307. 10.1128/JVI.02639-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Sinani D, Frizzo da Silva L, Jones C. 2013. A bovine herpesvirus 1 protein expressed in latently infected neurons (ORF2) promotes neurite sprouting in the presence of activated Notch1 or Notch3. J Virol 87:1183–1192. 10.1128/JVI.02783-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Perez S, Meyer F, Henderson G, Jiang Y, Sherman S, Doster A, Inman M, Jones C. 2007. A protein encoded by the bovine herpesvirus 1 ORF E gene induces neurite-like morphological changes in mouse neuroblastoma cells and is expressed in trigeminal ganglionic neurons. J Neurovirol 13:139–149. 10.1080/13550280701191459. [DOI] [PubMed] [Google Scholar]
- 29.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 82:10940–10945. 10.1128/JVI.01289-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Berwick DC, Harvey K. and2012. The importance of Wnt signaling for neurodegeneration in Parkinson's disease. Biochem Soc Trans 40:1123–1128. 10.1042/BST20120122. [DOI] [PubMed] [Google Scholar]
- 31.Fukumoto S, Hsieh C-M, Maemura K, Layne MD, Yet S-F, Lee K-H, Matsui T, Rosenzweig A, Taylor WG, Rubin JS, Perrella MA, Lee M-E. 2001. Akt participation in the Wnt signaling pathway through dishevelled. J Biol Chem 276:17479–17483. 10.1074/jbc.C000880200. [DOI] [PubMed] [Google Scholar]
- 32.Inestrosa NC, Varela-Nallar L. 2014. Wnt signaling in the nervous system and in Alzheimer’s disease. J Mol Cell Biol 6:64–74. 10.1093/jmcb/mjt051. [DOI] [PubMed] [Google Scholar]
- 33.Kondo T, Matsuoka AJ, Shimomura A, Koehler KR, Chan RJ, Miller JM, Srour EF, Hashino E. 2011. Wnt signaling promotes neuronal differentiation from mesencymal stem cells through activation of Tlx3. Stem Cells 29:836–846. 10.1002/stem.624. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Lambert C, Cisternas P, Inestrosa NC. 2016. Role of Wnt signaling in central nervous system injury. Mol Neurobiol 53:2297–2311. 10.1007/s12035-015-9138-x. [DOI] [PubMed] [Google Scholar]
- 35.Rosso SB, Inestrosa NC. 2013. Wnt signaling in neuronal maturation and synaptogenesis. Cellular Neuroscience 7:1–11. 10.3389/fncel.2013.00103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Salinas PC. 2012. Wnt signaling in the vertebrate central nervous system: from axon guidance to synaptic function. Cold Spring Harb Perpect Biol 4:a008003. 10.1101/cshperspect.a008003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Carrasco-Serrano C, Criado M. 2004. Glucocorticoid activation of the neuronal nicotinic acetylcholine receptor alpha7 subunit gene: involvement of transcription factor Egr-1. FEBS Lett 566:247–250. 10.1016/j.febslet.2004.04.049. [DOI] [PubMed] [Google Scholar]
- 38.Winkler MT, Doster A, Jones C. 1999. Bovine herpesvirus 1 can infect CD4(+) T lymphocytes and induce programmed cell death during acute infection of cattle. J Virol 73:8657–8668. 10.1128/JVI.73.10.8657-8668.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Perez S, Inman M, Doster A, Jones C. 2005. Latency-related gene encoded by bovine herpesvirus 1 promotes virus growth and reactivation from latency in tonsils of infected calves. J Clin Microbiol 43:393–401. 10.1128/JCM.43.1.393-401.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Jones C, Newby TJ, Holt T, Doster A, Stone M, Ciacci-Zanella J, Webster CJ, Jackwood MW. 2000. Analysis of latency in cattle after inoculation with a temperature sensitive mutant of bovine herpesvirus 1 (RLB106). Vaccine 18:3185–3195. 10.1016/S0264-410X(00)00106-7. [DOI] [PubMed] [Google Scholar]
- 41.Fuchs M, Hubert P, Detterer J, Rziha HJ. 1999. Detection of bovine herpesvirus type 1 in blood from naturally infected cattle by using a sensitive PCR that discriminates between wild-type virus and virus lacking glycoprotein E. J Clin Microbiol 37:2498–2507. 10.1128/JCM.37.8.2498-2507.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Mweene AS, Okazaki K, Kida H. 1996. Detection of viral genome in non-neural tissues of cattle experimentally infected with bovine herpesvirus 1. Jpn J Vet Res 44:165–174. [PubMed] [Google Scholar]
- 43.Cheung AK. 1995. Investigation of pseudorabies virus DNA and RNA in trigeminal ganglia and tonsil tissues of latently infected swine. Am J Vet Res 56:45–50. [PubMed] [Google Scholar]
- 44.Sabo A, Rajanci J. 1976. Latent pseudorabies virus infection in pigs. Acta Virologica 20:208–214. [PubMed] [Google Scholar]
- 45.Borchers K, Wolfinger U, Ludwig H. 1999. Latency-associated transcript of equine herpesvirus 4 in trigeminal ganglia of naturally infected horses. J General Virology 80:2165–2171. 10.1099/0022-1317-80-8-2165. [DOI] [PubMed] [Google Scholar]
- 46.Miyoshi M, Ishii Y, Takiguchi M, Takada A, Yasuda J, Hashimoto A, Okazaki K, Kida H. 1999. Detection of canine herpesvirus DNA in ganglionic neurons and the lymph node lymphocytes of latently infected dogs. J Vet Med Sci 61:375–379. 10.1292/jvms.61.375. [DOI] [PubMed] [Google Scholar]
- 47.Sahin F, Gerceker D, Karasartova D, Ozsan TM. 2007. Detection of herpes simplex virus type 1 in addition to Epstein–Bar virus in tonsils using a new multiplex polymerase chain reaction assay. Diagn Microbiol Infect Dis 57:47–51. 10.1016/j.diagmicrobio.2006.09.013. [DOI] [PubMed] [Google Scholar]
- 48.Hammerschmidt W, Ludwig H, Buhk H-J. 1986. Short repeats cause heterogeneity at genomic terminus of bovine herpesvirus 1. J Virol 58:43–49. 10.1128/JVI.58.1.43-49.1986. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Rock DL, Beam SL, Mayfield JE. 1987. Mapping bovine herpesvirus type 1 latency-related RNA in trigeminal ganglia of latently infected rabbits. J Virol 61:3827–3831. 10.1128/JVI.61.12.3827-3831.1987. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Schwyzer M, Vlcek C, Menekse O, Fraefel C, Paces V. 1993. Promoter, spliced leader, and coding sequence for BICP4, the largest of the immediate-early proteins of bovine herpesvirus 1. Virology 197:349–357. 10.1006/viro.1993.1596. [DOI] [PubMed] [Google Scholar]
- 51.Guo J, Li Q, Jones C. 2019. The bovine herpesvirus 1 regulatory proteins, bICP4 and bICP22, are expressed during the escape from latency. J Neurovirol 25:42–49. 10.1007/s13365-018-0684-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Casteleyn C, Breugelmans S, Simoens P, Van den Broeck W. 2011. The Tonsils revisited: review of the anatomical localization and histological characteristics of the tonsils of domestic and laboratory animals. Clin Dev Immunol 2011:349–357. 10.1155/2011/472460. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Rathmell JC, Thompson CB. 2002. Pathways of apoptosis in lymphocyte development, homeostasis, and disease. Cell 109:S97–S107. 10.1016/S0092-8674(02)00704-3. [DOI] [PubMed] [Google Scholar]
- 54.Du T, Zhou G, Roizman B. 2012. Induction of apoptosis accelerates reactivation from latent HSV-1 in ganglionic organ cultures and replication in cell cultures. Proc Natl Acad Sci USA 109:14616–14621. 10.1073/pnas.1212661109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Prasad A, Remick J, Zeichner SL. 2013. Activation of human herpesvirus replication by apoptosis. J Virol 87:10641–10650. 10.1128/JVI.01178-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Prasad A, Lu M, Lukac DM, Zeichner SL. 2012. An alternative Kaposi’s sarcoma-associated herpesvirus replication program triggered by host cell apoptosis. J Virol 86:4404–4419. 10.1128/JVI.06617-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Fabregat A, Sidiropoulos K, Garapati P, Gillespie M, Hausmann K, Haw R, Jassal B, Jupe S, Korninger F, McKay S, Matthews L, May B, Milacic M, Rothfels K, Shamovsky V, Webber M, Weiser J, Williams M, Wu G, Stein L, Hermjakob H, D'Eustachio P. 2016. The reactome pathway knowledgebase. Nucleic Acids Res 44:D481–D487. 10.1093/nar/gkv1351. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Fabregat A, Sidiropoulos K, Viteri G, Forner O, Marin-Garcia P, Arnau V, D'Eustachio P, Stein L, Hermjakob H. 2017. Reactome pathway analysis: a high performance in-memory approach. BMC Bioinformatics 18:4404–4419. 10.1186/s12859-017-1559-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Stender JD, Stossi F, Funk CC, Charn TH, Barnett DH, Katzenellenbogen BS. 2011. The estrogen-regulated transcription factor PITX1 coordinates gene-specific regulation by estrogen receptor-alpha in breast cancer cells. Mol Endocrinol 25:1699–1709. 10.1210/me.2011-0102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Saxena K, Srikrishnan S, Celia-Terrassa T, Jolly MK. 2022. OVOL1/2: drivers of epithelial differentiation in development, disease, and reprogramming. Cells Tissues Organs 211:183–192. 10.1159/000511383. [DOI] [PubMed] [Google Scholar]
- 61.Workman A, Eudy J, Smith L, Frizzo da Silva L, Sinani D, Bricker H, Cook E, Doster A, Jones C. 2012. Cellular transcription factors induced in trigeminal ganglia during dexamethasone-induced reactivation from latency stimulate bovine herpesvirus 1 productive infection and certain viral promoters. J Virol 86:2459–2473. 10.1128/JVI.06143-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Mangan S, Alon U. 2003. Structure and function of the feed-forward loop network motif. Proc Natl Acad Sci USA 100:11980–11985. 10.1073/pnas.2133841100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Sasse SK, Mailloux CM, Barczak AJ, Wang Q, Altonsy MO, Jain MK, Haldar SM, Gerber AN. 2013. The glucocorticoid receptor and KLF15 regulate gene expression dynamics and integrate signals through feed-forward circuitry. Mol Cell Biol 33:2104–2115. 10.1128/MCB.01474-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.El-Mayet FS, El-Habbaa AS, D’Offay J, Jones C. 2019. Synergistic activation of bovine herpesvirus 1 productive infection and viral regulatory promoters by the progesterone receptor and Krüppel-like transcription factor 15. J Virol 93:e01519-18. 10.1128/JVI.01519-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.El-Mayet FS, Sawant L, Thunuguntla P, Jones C. 2017. Combinatorial effects of the glucocorticoid receptor and Krüppel-like transcription factor 15 on bovine herpesvirus 1 transcription and productive infection. J Virol 91:e00904-17. 10.1128/JVI.00904-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.El-Mayet FS, Harrison KS, Jones C. 2021. Regulation of Krüppel-like factor 15 expression by herpes simplex virus type 1 or bovine herpesvirus 1 productive infection. Viruses 13:13. 10.3390/v13061148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Ostler J, Jones C. 2021. Stress induced transcription factors transactivate the herpes simplex virus 1 infected cell protein 27 (ICP27) transcriptional enhancer. Viruses 13:2296. 10.3390/v13112296. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Ostler JB, Thunuguntla P, Hendrickson BY, Jones C. 2021. Transactivation of HSV-1 infected cell protein 4 (ICP4) enhancer by glucocorticoid receptor and stress-induced transcription factors requires overlapping Krüppel like transcription factor 4/Sp1 binding sites. J Virol 95:e01776-20. 10.1128/JVI.01776-20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Fernandes I, Bastien Y, Wai T, Nygard K, Lin R, Cormier O, Lee HS, Eng F, Bertos NR, Pelletier N, Mader S, Han VKM, Yang X-J, White JH. 2003. Ligand-dependent nuclear receptor corepressor LCoR functions by histone deacetylase-dependent and -independent mechanisms. Mol Cell 11:139–150. 10.1016/S1097-2765(03)00014-5. [DOI] [PubMed] [Google Scholar]
- 70.Morasso MI, Grinberg A, Robinson G, Sargent TD, Mahon KA. 1999. Placental failure in mice lacking the homeobox gene Dlx3. Proc Natl Acad Sci USA 96:162–167. 10.1073/pnas.96.1.162. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Peng L, Payne AH. 2002. AP-2 gamma and the homeodomain protein distal-less 3 are required for placental-specific expression of the murine 3b-hydroxysteroid dehydrogenase VI gene, Hsd3b6. J Biol Chem 277:7945–7954. 10.1074/jbc.M106765200. [DOI] [PubMed] [Google Scholar]
- 72.Thompson EM, Stoker AW. 2021. Review of DUSP26: Structure, Regulation and Relevance in Human Disease. Int J Mol Sci 22:776. 10.3390/ijms22020776. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Takasugra A. 2016. PLAG1 and NCAPG-LCORL in livestock. Anim Sci J 87:159–167. 10.1111/asj.12417. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Cassandri M, Smirnov A, Novelli F, Pitolli C, Agostini M, Malewicz M, Melino G, Raschell G. 2017. Zinc-finger proteins in health and disease. Cell Death Discov 3:17071. 10.1038/cddiscovery.2017.71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Tian Y, Zeng Z, Li X, Wang Y, Chen R, Mattijssen S, Gaidamakov S, Wu Y, Maraia RJ, Peng W, Zhu J. 2020. Transcriptome-wide stability analysis uncovers LARP4-mediated NFκB1 mRNA stabilization during T cell activation. Nucleic Acids Res 48:8724–8739. 10.1093/nar/gkaa643. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Eskra L, Splitter GA. 1997. Bovine herpesvirus-1 infects activated CD4+ lymphocytes. J Gen Virol 78:2159–2166. 10.1099/0022-1317-78-9-2159. [DOI] [PubMed] [Google Scholar]
- 77.Heng L, Li S, Shu H-B. 2019. The membrane-associated MARCH E3 ligase family: emerging roles in immune regulation. Front Immunol 24:1751. 10.1093/nar/gkaa643. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Swanson K, Deng M, Ting J-Y. 2019. The NLRP3 inflammasome: molecular activation and regulation to therapeutics. Nat Rev Immunol 19:477–489. 10.1038/s41577-019-0165-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Blackmore TM, Mercer CF, Paterno GD, Gillespie LL. 2008. The transcriptional cofactor MIER1-beta negatively regulates histone acetyltransferase activity of the CREB-binding protein. BMC Res Notes 1:68. 10.1186/1756-0500-1-68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Subramaniam M, Hawse JR, Rajamannan NM, Ingle JN, Spelsberg TC. 2010. Functional role of KLF10 in multiple disease processes. Biofactors 36:8–18. 10.1002/biof.67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Ebenfelt A, Ivarsson M. 2001. Neutrophil migration in tonsils. J Anat 198:497–500. 10.1046/j.1469-7580.2001.19840497.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Lundqvist A, Lundqvist H, Dahlgren C, Lundberg C. 1996. Neutrophils in mucosal secretion are functionally active. Clin Exp Immunol 106:404–409. 10.1046/j.1365-2249.1996.d01-846.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Kobayashi SD, Malachowa N, DeLeo FR. 2017. Influences of microbes and neutrophil life and death. Front in Cellular and Infection Microbiology 7:Article 159. 10.3389/fcimb.2017.00159. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Wright HL, Moots RJ, Bucknall RC, Edwards SW. 2010. Neutrophil function in inflammation and inflammatory diseases. Rheumatology (Oxford) 49:1618–1631. 10.1093/rheumatology/keq045. [DOI] [PubMed] [Google Scholar]
- 85.Hodge RG, Ridley AJ. 2020. Regulation and functions of RhoU and RhoV. Small GTPases 11:8–15. 10.1080/21541248.2017.1362495. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.DeLuca NA, McCarthy AM, Schaffer PA. 1985. Isolation and characterization of deletion mutants of herpes simplex virus type 1 in the gene encoding immediate-early regulatory protein ICP4. J Virol 56:558–570. 10.1128/JVI.56.2.558-570.1985. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.DeLuc NA. 2011. Functions and mechanism of action of the herpes simplex virus regulatory protein, ICP4, p 17–38. In Weller SK (ed), Alphaherpesviruses: Molecular Virology. Caister Academic Press, Norfolk, UK. [Google Scholar]
- 88.Kristie TM, Roizman B. 1986. Alpha 4, the major regulatory protein of herpes simplex virus type 1, is stably and specifically associated with promoter-regulatory domains of alpha genes and of selected other viral genes. Proc Natl Acad Sci USA 83:3218–3222. 10.1073/pnas.83.10.3218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Carrozza MJ, DeLuca NA. 1996. Interaction of the viral activator protein ICP4 with TFIID through TAF250. Mol Cell Biol 16:3085–3093. 10.1128/MCB.16.6.3085. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Smith CA, Bates P, Rivera-Gonzalez R, Gu B, DeLuca NA. 1993. ICP4, the major transcriptional regulatory protein of herpes simplex virus type 1, forms a tripartite complex with TATA-binding protein and TFIIB. J Virol 67:4676–4687. 10.1128/JVI.67.8.4676-4687.1993. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.DeLuca NA, Schaffer PA. 1985. Activation of immediate-early, early, and late promoters by temperature- sensitive and wild-type forms of herpes simplex virus type 1 protein ICP4. Mol Cell Biol 5:1997–2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Gibeault R, Conn KL, Bildersheim MD, Schang LM. 2016. An essential viral transcription activator modulates chromatin dynamics. PLoS Pathog 12:e1005842. 10.1371/journal.ppat.1005842. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Frizzo da Silva L, Kook I, Doster A, Jones C. 2013. Bovine herpesvirus 1 regulatory proteins, bICP0 and VP16, are readily detected in trigeminal ganglionic neurons expressing the glucocorticoid receptor during the early stages of reactivation from latency. J Virol 87:11214–11222. 10.1128/JVI.01737-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Kook I, Doster A, Jones C. 2015. Bovine herpesvirus 1 regulatory proteins are detected in trigeminal ganglionic neurons during the early stages of stress-induced escape from latency. J Neurovirol 21:585–591. 10.1007/s13365-015-0339-x. [DOI] [PubMed] [Google Scholar]
- 95.Pokhriyal M, Verma OP, Ratta B, Kumar A, Saxena M, Sharma B. 2016. Bovine herpes virus 1 major immediate early transcription unit 1 (IETU-1) uses alternative promoters to transcribe BICP0 and BICP4 transcripts. Curr Microbiol 72:420–425. 10.1007/s00284-015-0961-0. [DOI] [PubMed] [Google Scholar]
- 96.Pugh B, Tjian R. 1990. Mechanism of transcriptional activation by Sp 1: evidence for coactivators. Cell 61:1187–1197. 10.1016/0092-8674(90)90683-6. [DOI] [PubMed] [Google Scholar]
- 97.Pugh B, Tjian R. 1991. Transcription from a TATA-Iess promoter requires a multisubunit TFIID complex. Genes Dev 5:1935–1945. 10.1101/gad.5.11.1935. [DOI] [PubMed] [Google Scholar]
- 98.Kook I, Henley C, Meyer F, Hoffmann F, Jones C. 2015. Bovine herpesvirus 1 productive infection and the immediate early transcription unit 1 are stimulated by the synthetic corticosteroid dexamethasone. Virology 484:377–385. 10.1016/j.virol.2015.06.010. [DOI] [PubMed] [Google Scholar]
- 99.Polman JA, Welten JE, Bosch DS, de Jonge RT, Balog J, van der Maarel SM, de Kloet ER, Datson NA. 2012. A genome-wide signature of glucocorticoid receptor binding in neuronal PC12 cells. BMC Neurosci 13:118–125. 10.1186/1471-2202-13-118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Kobayashi N, Takahashi D, Takano S, Kimura S, Hase K. 2019. The roles of peyer’s patches and microfold cells in the gut immune system: relevance to autoimmune diseases. Front Immunol 10:2345. 10.3389/fimmu.2019.02345. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Devireddy LR, Jones C. 1998. Alternative splicing of the latency-related transcript of bovine herpesvirus 1 yields RNAs containing unique open reading frames. J Virol 72:7294–7301. 10.1128/JVI.72.9.7294-7301.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Schang LM, Hossain A A, Jones C. 1996. The latency-related gene of bovine herpesvirus 1 encodes a product which inhibits cell cycle progression. J Virol 70:3807–3814. 10.1128/JVI.70.6.3807-3814.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Jones C, Delhon G, Bratanich A, Kutish G, Rock D. 1990. Analysis of the transcriptional promoter which regulates the latency- related transcript of bovine herpesvirus 1. J Virol 64:1164–1170. 10.1128/JVI.64.3.1164-1170.1990. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Bratanich AC, Jones C. 1992. Localization of cis-acting sequences in the latency-related promoter of bovine herpesvirus 1 which are regulated by neuronal cell type factors and immediate-early genes. J Virol 66:6099–6106. 10.1128/JVI.66.10.6099-6106.1992. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Delhon G, Jones C. 1997. Identification of DNA sequences in the latency related promoter of bovine herpes virus type 1 which are bound by neuronal specific factors. Virus Res 51:93–103. 10.1016/S0168-1702(97)00085-3. [DOI] [PubMed] [Google Scholar]
- 106.Bratanich AC, Hanson ND, Jones C. 1992. The latency-related gene of bovine herpesvirus 1 inhibits the activity of immediate-early transcription unit 1. Virology 191:988–991. 10.1016/0042-6822(92)90278-W. [DOI] [PubMed] [Google Scholar]
- 107.Liu Y, Jones C. 2016. Regulation of Notch-mediated transcription by a bovine herpesvirus 1 encoded protein (ORF2) that is expressed in latently infected sensory neurons. J Neurovirol 22:518–528. 10.1007/s13365-015-0394-3. [DOI] [PubMed] [Google Scholar]
- 108.Pittayakhajonwut D, Sinani D, Jones C. 2013. A protein (ORF2) encoded by the latency related gene of bovine herpesvirus 1 interacts with DNA. J Virol 87:5493–5501. 10.1128/JVI.00193-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Workman A, Sinani D, Pittayakhajonwut D, Jones C. 2011. A protein (ORF2) encoded by the latency related gene of bovine herpesvirus 1 interacts with notch1 and notch3. J Virol 85:2536–2546. 10.1128/JVI.01937-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Zhao J, Wijesekera N, Jones C. 2021. Inhibition of stress-induced viral promoters by a bovine herpesvirus 1 non-coding RNA and the cellular transcription factor, beta-Catenin. Int J Mol Sci 22:519. 10.3390/ijms22020519. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Inman M, Lovato L, Doster A, Jones C. 2001. A mutation in the latency-related gene of bovine herpesvirus 1 leads to impaired ocular shedding in acutely infected calves. J Virol 75:8507–8515. 10.1128/jvi.75.18.8507-8515.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Schang L, Jones C. 1997. Analysis of bovine herpesvirus 1 transcripts during a primary infection of trigeminal ganglia of cattle. J Virol 71:6786–6795. 10.1128/JVI.71.9.6786-6795.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Sinani D, Cordes E, Workman A, Thunuguntia P, Jones C. 2013. Stress induced cellular transcription factors expressed in trigeminal ganglionic neurons stimulate the herpes simplex virus type 1 (HSV-1) infected cell protein 0 (ICP0) promoter. J Virol 87:1183–1192. 10.1128/JVI.02476-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Bolger AM, Lohse M, Usadel B. 2014. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics 30:2114–2120. 10.1093/bioinformatics/btu170. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Trapnell C, Pachter L, Salzberg SL. 2009. TopHat: discovering splice junctions with RNA-Seq. Bioinformatics 25:1105–1111. 10.1093/bioinformatics/btp120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Trapnell C, Williams BA, Pertea G, Mortazavi A, Kwan G, van Baren MJ, Salzberg SL, Wold B, Pachter L. 2010. Transcript assembly and abundance estimation from RNA-Seq reveals thousands of new transcripts and switching among isoforms. Nat Biotechnol 28:511–515. 10.1038/nbt.1621. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Benjamini Y, Hochberg Y. 1995. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J R Stat Soc Series B 57:289–300. 10.1111/j.2517-6161.1995.tb02031.x. [DOI] [Google Scholar]
- 118.Abendorth A, Arvin AM. 2000. Host responses to primary infection, p 142–156. In Arvin AM, Gershwin AA (ed), Varicella-Zoster virus. Varicella Viruses, vol 1. Cambridge University Press, Cambridge, England. [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
The RNA-seq data was deposited into the NCBI GEO repository as series record GSE216685.









