Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters











Publication year range
1.
J Virol ; 98(6): e0042324, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38771044

ABSTRACT

Bovine alphaherpesvirus 1 (BoHV-1) infection causes respiratory tract disorders and immune suppression and may induce bacterial pneumonia. BoHV-1 establishes lifelong latency in sensory neurons after acute infection. Reactivation from latency consistently occurs following stress or intravenous injection of the synthetic corticosteroid dexamethasone (DEX), which mimics stress. The immediate early transcription unit 1 (IEtu1) promoter drives expression of infected cell protein 0 (bICP0) and bICP4, two viral transcriptional regulators necessary for productive infection and reactivation from latency. The IEtu1 promoter contains two glucocorticoid receptor (GR) responsive elements (GREs) that are transactivated by activated GR. GC-rich motifs, including consensus binding sites for specificity protein 1 (Sp1), are in the IEtu1 promoter sequences. E2F family members bind a consensus sequence (TTTCCCGC) and certain specificity protein 1 (Sp1) sites. Consequently, we hypothesized that certain E2F family members activate IEtu1 promoter activity. DEX treatment of latently infected calves increased the number of E2F2+ TG neurons. GR and E2F2, but not E2F1, E2F3a, or E2F3b, cooperatively transactivate a 436-bp cis-regulatory module in the IEtu1 promoter that contains both GREs. A luciferase reporter construct containing a 222-bp fragment downstream of the GREs was transactivated by E2F2 unless two adjacent Sp1 binding sites were mutated. Chromatin immunoprecipitation studies revealed that E2F2 occupied IEtu1 promoter sequences when the BoHV-1 genome was transfected into mouse neuroblastoma (Neuro-2A) or monkey kidney (CV-1) cells. In summary, these findings revealed that GR and E2F2 cooperatively transactivate IEtu1 promoter activity, which is predicted to influence the early stages of BoHV-1 reactivation from latency. IMPORTANCE: Bovine alpha-herpesvirus 1 (BoHV-1) acute infection in cattle leads to establishment of latency in sensory neurons in the trigeminal ganglia (TG). A synthetic corticosteroid dexamethasone consistently initiates BoHV-1 reactivation in latently infected calves. The BoHV-1 immediate early transcription unit 1 (IEtu1) promoter regulates expression of infected cell protein 0 (bICP0) and bICP4, two viral transcriptional regulators. Hence, the IEtu1 promoter must be activated for the reactivation to occur. The number of TG neurons expressing E2F2, a transcription factor and cell cycle regulator, increased during early stages of reactivation from latency. The glucocorticoid receptor (GR) and E2F2, but not E2F1, E2F3a, or E2F3b, cooperatively transactivated a 436-bp cis-regulatory module (CRM) in the IEtu1 promoter that contains two GR responsive elements. Chromatin immunoprecipitation studies revealed that E2F2 occupies IEtu1 promoter sequences in cultured cells. GR and E2F2 mediate cooperative transactivation of IEtu1 promoter activity, which is predicted to stimulate viral replication following stressful stimuli.


Subject(s)
Cell Cycle , E2F2 Transcription Factor , Gene Expression Regulation, Viral , Herpesvirus 1, Bovine , Immediate-Early Proteins , Promoter Regions, Genetic , Receptors, Glucocorticoid , Transcriptional Activation , Animals , Cattle , Mice , Binding Sites , Cell Line , Dexamethasone/pharmacology , E2F2 Transcription Factor/metabolism , Gene Expression Regulation, Viral/drug effects , Gene Expression Regulation, Viral/genetics , Herpesviridae Infections/virology , Herpesviridae Infections/metabolism , Herpesviridae Infections/veterinary , Herpesviridae Infections/genetics , Herpesvirus 1, Bovine/genetics , Herpesvirus 1, Bovine/physiology , Immediate-Early Proteins/genetics , Neurons/virology , Receptors, Glucocorticoid/metabolism , Response Elements/genetics , Sp1 Transcription Factor/metabolism , Trans-Activators/metabolism , Trigeminal Ganglion/cytology , Trigeminal Ganglion/virology , Virus Activation , Virus Latency
2.
J Virol ; 97(10): e0130523, 2023 10 31.
Article in English | MEDLINE | ID: mdl-37823644

ABSTRACT

IMPORTANCE: A correlation exists between stress and increased episodes of human alpha-herpes virus 1 reactivation from latency. Stress increases corticosteroid levels; consequently, the glucocorticoid receptor (GR) is activated. Recent studies concluded that a GR agonist, but not an antagonist, accelerates productive infection and reactivation from latency. Furthermore, GR and certain stress-induced transcription factors cooperatively transactivate promoters that drive the expression of infected cell protein 0 (ICP0), ICP4, and VP16. This study revealed female mice expressing a GR containing a serine to alanine mutation at position 229 (GRS229A) shed significantly lower levels of infectious virus during explant-induced reactivation compared to male GRS229A or wild-type parental C57BL/6 mice. Furthermore, female GRS229A mice contained fewer VP16 + TG neurons compared to male GRS229A mice or wild-type mice during the early stages of explant-induced reactivation from latency. Collectively, these studies revealed that GR transcriptional activity has female-specific effects, whereas male mice can compensate for the loss of GR transcriptional activation.


Subject(s)
Herpes Simplex , Herpesvirus 1, Human , Receptors, Glucocorticoid , Virus Activation , Animals , Female , Male , Mice , Herpes Simplex/genetics , Herpes Simplex/virology , Herpesvirus 1, Human/physiology , Immediate-Early Proteins/metabolism , Mice, Inbred C57BL , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Trigeminal Ganglion , Ubiquitin-Protein Ligases/metabolism , Virus Activation/genetics , Virus Latency/genetics
3.
Curr Clin Microbiol Rep ; 10(4): 236-245, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38173564

ABSTRACT

Purpose of Review: Numerous studies concluded stress (acute, episodic acute, or chronic) increases the incidence of human alpha-herpes virus 1 (HSV-1) reactivation from latency in neurons. This review will summarize how stress stimulates viral gene expression, replication, and reactivation from latency. Recent Findings: Stress (capital S) stress-mediated activation of the glucocorticoid receptor (GR) accelerates reactivation from latency, whereas a corticosteroid-specific antagonist impairs viral replication and reactivation from latency. GR and specific stress-induced cellular transcription factors also stimulate viral promoters that drive expression of key viral transcriptional regulators: infected cell protein 0 (ICP0), ICP4, ICP27 and viral tegument protein (VP16). Hence, GR is predicted to initially stimulate viral gene expression. GR-mediated immune-inhibitory functions are also predicted to enhance viral replication and viral spread. Summary: Identifying cellular factors and viral regulatory proteins that trigger reactivation from latency in neurons may provide new therapeutic strategies designed to reduce the incidence of reactivation from latency.

4.
J Virol ; 96(23): e0101022, 2022 12 14.
Article in English | MEDLINE | ID: mdl-36416585

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.


Subject(s)
Adenoids , Herpesviridae Infections , Herpesvirus 1, Bovine , Viral Envelope Proteins , Virus Activation , Animals , Cattle , Adenoids/virology , Dexamethasone/pharmacology , Etoposide/pharmacology , Herpesvirus 1, Bovine/physiology , RNA/metabolism , Trigeminal Ganglion , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Latency , Viral Envelope Proteins/metabolism
5.
J Virol ; 96(5): e0213021, 2022 03 09.
Article in English | MEDLINE | ID: mdl-35019726

ABSTRACT

Acute infection of the ocular, oral, or nasal cavity by bovine herpesvirus 1 (BoHV-1) culminates in lifelong latency in sensory neurons within trigeminal ganglia. The BoHV-1 latency reactivation cycle, including calves latently infected with commercially available modified live vaccines, can lead to reproductive complications, including abortions. Recent studies demonstrated progesterone stimulated BoHV-1 productive infection and sporadically induced reactivation from latency in male rabbits. The progesterone receptor (PR) and progesterone transactivate the immediate early transcription unit 1 (IEtu1) promoter and the infected cell protein 0 (bICP0) early promoter. These viral promoters drive expression of two viral transcriptional regulatory proteins (bICP0 and bICP4) that are crucial for productive infection. Based on these observations, we hypothesize that progesterone induces reactivation in a subset of calves latently infected with BoHV-1. These studies demonstrated progesterone was less efficient than dexamethasone at initiating reactivation from latency in female calves. Notably, heat stress correlated with enhancing the ability of progesterone to induce reactivation from latency. Previous studies demonstrated that heat stress activates the glucocorticoid receptor (GR), which suggested GR activation augments progesterone-mediated reactivation from latency. Additional studies revealed GR and PR cooperatively stimulated productive infection and synergistically transactivated the IEtu1 promoter when cultures were treated with dexamethasone. Mutating one or both GR binding sites in the IEtu1 promoter blocked transactivation. Collectively, these studies indicated that progesterone intermittently triggered reactivation from latency, and heat stress augmented reactivation from reactivation. Finally, these studies suggest progesterone enhances virus spread in tissues and cells where PR is abundantly expressed. IMPORTANCE Steroid hormone fluctuations are predicted to enhance or initiate bovine herpesvirus 1 (BoHV-1) replication and virus spread in cattle. For example, stress increases the incidence of BoHV-1 reactivation from latency in cattle, and the synthetic corticosteroid dexamethasone consistently induces reactivation from latency. The glucocorticoid receptor (GR) and dexamethasone stimulate key viral regulatory promoters and productive infection, in part because the viral genome contains numerous consensus GR-responsive elements (GREs). The progesterone receptor (PR) and GR belong to the type I nuclear hormone receptor family. PR and progesterone specifically bind to and transactivate viral promoters that contain GREs and stimulate BoHV-1 productive infection. Although progesterone did not induce reactivation from latency in female calves as efficiently as dexamethasone, heat stress enhanced progesterone-mediated reactivation from latency. Consequently, we predict that low levels of stressful stimuli can cooperate with progesterone to induce reactivation from latency or promote virus spread.


Subject(s)
Herpesviridae Infections , Herpesvirus 1, Bovine , Progesterone , Animals , Cattle , Dexamethasone/pharmacology , Female , Heat-Shock Response , Herpesviridae Infections/virology , Herpesvirus 1, Bovine/physiology , Male , Progesterone/pharmacology , Rabbits , Receptors, Glucocorticoid/metabolism , Receptors, Progesterone/genetics , Receptors, Progesterone/metabolism , Virus Activation/drug effects , Virus Latency/drug effects
6.
Vitam Horm ; 117: 101-132, 2021.
Article in English | MEDLINE | ID: mdl-34420577

ABSTRACT

Neurotropic α-herpesvirinae subfamily members, herpes simplex virus type 1 (HSV-1) and bovine herpesvirus 1 (BoHV-1), are important viral pathogens in their respective hosts. Following acute infection on mucosal surfaces, these viruses establish life-long latency in neurons within trigeminal ganglia (TG) and central nervous system. Chronic or acute stress (physiological or psychological) increases the frequency of reactivation from latency, which leads to virus shedding, virus transmission, and recurrent disease. While stress impairs immune responses and inflammatory signaling cascades, we predict stressful stimuli directly stimulate viral gene expression and productive infection during early stages of reactivation from latency. For example, BoHV-1 and HSV-1 productive infection is impaired by glucocorticoid receptor (GR) antagonists but is stimulated by the synthetic corticosteroid dexamethasone. Promoters that drive expression of key viral transcriptional regulatory proteins are cooperatively stimulated by GR and specific Krüppel like transcription factors (KLF) induced during stress induced reactivation from latency. The BoHV-1 immediate early transcription unit 1 promoter and contains two GR response elements (GRE) that are essential for cooperative transactivation by GR and KLF15. Conversely, the HSV-1 infected cell protein 0 (ICP0) and ICP4 promoter as well as the BoHV-1 ICP0 early promoter lack consensus GREs: however, these promoters are cooperatively transactivated by GR and KLF4 or KLF15. Hence, growing evidence suggests GR and stress-induced transcription factors directly stimulate viral gene expression and productive infection during early stages of reactivation from latency. We predict the immune inhibitory effects of stress enhance virus spread at late stages during reactivation from latency.


Subject(s)
Herpesviridae Infections , Receptors, Glucocorticoid , Gene Expression Regulation, Viral , Herpesviridae Infections/genetics , Herpesviridae Infections/metabolism , Humans , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Transcription Factors/genetics , Virus Activation/genetics
8.
J Virol ; 95(20): e0076821, 2021 09 27.
Article in English | MEDLINE | ID: mdl-34319779

ABSTRACT

Following bovine herpesvirus 1 (BoHV-1) acute infection of ocular, oral, or nasal cavities, sensory neurons within trigeminal ganglia are an important site for latency. Stress, as mimicked by the synthetic corticosteroid dexamethasone, consistently induces reactivation from latency. Expression of two key viral transcriptional regulatory proteins, BoHV-1 infected cell protein 0 (bICP0) and bICP4, are regulated by sequences within the immediate early promoter (IEtu1). A separate early promoter also drives bICP0 expression, presumably to ensure sufficient levels of this important transcriptional regulatory protein. Productive infection and bICP0 early promoter activity are cooperatively transactivated by Krüppel-like factor 4 (KLF4) and a type I nuclear hormone receptor (NHR), androgen receptor, glucocorticoid receptor, or progesterone receptor. The bICP0 early promoter contains three separate transcriptional enhancers that mediate cooperative transactivation. In contrast to the IEtu1 promoter, the bICP0 early promoter lacks consensus type I NHR binding sites. Consequently, we hypothesized that KLF4 and Sp1 binding sites are essential for type I NHR and KLF4 to transactivate the bICP0 promoter. Mutating KLF4 and Sp1 binding sites in each enhancer domain significantly reduced transactivation by KLF4 and a type I NHR. Chromatin immunoprecipitation (ChIP) studies demonstrated that occupancy of bICP0 early promoter sequences by KLF4 and type I NHR is significantly reduced when KLF4 and/or Sp1 binding sites are mutated. These studies suggest that cooperative transactivation of the bICP0 E promoter by type I NHRs and a stress-induced pioneer transcription factor (KLF4) promote viral replication and spread in neurons or nonneural cells in reproductive tissue. IMPORTANCE Understanding how stressful stimuli and changes in the cellular milieu mediate viral replication and gene expression in the natural host is important for developing therapeutic strategies that impair virus transmission and disease. For example, bovine herpesvirus 1 (BoHV-1) reactivation from latency is consistently induced by the synthetic corticosteroid dexamethasone, which mimics the effects of stress. Furthermore, BoHV-1 infection increases the incidence of abortion in pregnant cows, suggesting that sex hormones stimulate viral growth in certain tissues. Previous studies revealed that type I nuclear hormone receptors (NHRs) (androgen, glucocorticoid, or progesterone) and a pioneer transcription factor, Krüppel-like factor 4 (KLF4), cooperatively transactivate the BoHV-1 infected cell protein 0 (bICP0) early promoter. Transactivation was mediated by Sp1 and/or KLF4 consensus binding sites within the three transcriptional enhancers. These studies underscore the complexity by which BoHV-1 exploits type I NHR fluctuations to enhance viral gene expression, replication, and transmission in the natural host.


Subject(s)
Herpesvirus 1, Bovine/metabolism , Trans-Activators/genetics , Ubiquitin-Protein Ligases/genetics , A549 Cells , Animals , Binding Sites , Gene Expression Regulation, Viral/genetics , Herpesviridae Infections/virology , Herpesvirus 1, Bovine/genetics , Humans , Immediate-Early Proteins/metabolism , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/metabolism , Mice , Promoter Regions, Genetic/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Trans-Activators/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/metabolism , Viral Proteins/metabolism , Virus Activation/genetics , Virus Latency/genetics , Virus Replication
9.
Front Microbiol ; 11: 1910, 2020.
Article in English | MEDLINE | ID: mdl-33013729

ABSTRACT

The protein encoded by the UL48 gene of alphaherpesviruses is named VP16 or alpha-gene-transactivating factor (α-TIF). In the early stage of viral replication, VP16 is an important transactivator that can activate the transcription of viral immediate-early genes, and in the late stage of viral replication, VP16, as a tegument, is involved in viral assembly. This review will explain the mechanism of VP16 acting as α-TIF to activate the transcription of viral immediate-early genes, its role in the transition from viral latency to reactivation, and its effects on viral assembly and maturation. In addition, this review also provides new insights for further research on the life cycle of alphaherpesviruses and the role of VP16 in the viral life cycle.

10.
Viruses ; 12(9)2020 09 14.
Article in English | MEDLINE | ID: mdl-32937797

ABSTRACT

Bovine herpesvirus 1 (BHV-1) causes respiratory infection and abortion in cattle. Following a primary infection, BHV-1 establishes lifelong latency in the trigeminal ganglia (TG). Periodic reactivation of the latent virus in TG neurons results in anterograde virus transport to nerve endings in the nasal mucosa and nasal virus shedding. The BHV-1 glycoprotein E cytoplasmic tail (gE-CT) is necessary for virus cell-to-cell spread in epithelial cells and neuronal anterograde transport. Recently, we identified two tyrosine residues, Y467 and Y563, within the tyrosine-based motifs 467YTSL470 and 563YTVV566, which, together, account for the gE CT-mediated efficient cell-to-cell spread of BHV-1 in epithelial cells. Here, we determined that in primary neuron cultures in vitro, the individual alanine exchange Y467A or Y563A mutants had significantly diminished anterograde axonal spread. Remarkably, the double-alanine-exchanged Y467A/Y563A mutant virus was not transported anterogradely. Following intranasal infection of rabbits, both wild-type (wt) and the Y467A/Y563A mutant viruses established latency in the TG. Upon dexamethasone-induced reactivation, both wt and the mutant viruses reactivated and replicated equally efficiently in the TG. However, upon reactivation, only the wt, not the mutant, was isolated from nasal swabs. Therefore, the gE-CT tyrosine residues Y467 and Y563 together are required for gE CT-mediated anterograde neuronal transport.


Subject(s)
Biological Transport/physiology , Glycoproteins/metabolism , Herpesvirus 1, Bovine/physiology , Neurons/virology , Tyrosine/metabolism , Animals , Cattle , Cattle Diseases/virology , Cell Line , Herpesviridae Infections/virology , Herpesvirus 1, Bovine/genetics , Lab-On-A-Chip Devices , Rabbits , Trigeminal Ganglion/virology , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism , Virus Activation , Virus Latency , Virus Shedding
11.
Virus Res ; 276: 197803, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31697987

ABSTRACT

Bovine herpesvirus 1 (BoHV-1), including modified live vaccines, can cause abortions in pregnant cows. Progesterone maintains pregnancy and promotes spermiogenesis and testosterone biosynthesis in males: furthermore, progesterone is a neuro-steroid. Recent published studies demonstrated progesterone stimulated the BoHV-1 immediate early transcription unit 1 (IEtu1) promoter, and two glucocorticoid receptor response elements within the promoter were required for progesterone mediated transactivation. In this study, we tested whether progesterone induces reactivation from latency in rabbits. As expected, the synthetic corticosteroid dexamethasone consistently induced reactivation from latency in males and females. While progesterone induced reactivation from latency in approximately one-half of male rabbits, virus shedding was sporadic compared to dexamethasone and less efficient in female rabbits. Progesterone significantly increased productive infection in rabbit skin cells, which correlated with stimulating reactivation. These studies suggest progesterone promotes BoHV-1 spread in cattle, in part, by increasing the frequency of reactivation from latency.


Subject(s)
Cattle Diseases/metabolism , Cattle Diseases/virology , Herpesviridae Infections/veterinary , Herpesvirus 1, Bovine/physiology , Progesterone/metabolism , Virus Activation , Virus Latency , Animals , Antibodies, Viral , Cattle , Cattle Diseases/immunology , Female , Male , Progesterone/pharmacology , Rabbits , Sex Factors , Virus Activation/drug effects , Virus Activation/immunology , Virus Latency/drug effects , Virus Latency/immunology , Virus Replication/drug effects , Virus Replication/immunology , Virus Shedding
12.
J Neurovirol ; 25(1): 42-49, 2019 02.
Article in English | MEDLINE | ID: mdl-30402823

ABSTRACT

Following acute infection of mucosal surfaces by bovine herpesvirus 1 (BoHV-1), sensory neurons are a primary site for lifelong latency. Stress, as mimicked by the synthetic corticosteroid dexamethasone, consistently induces reactivation from latency. Two viral regulatory proteins (VP16 and bICP0) are expressed within 1 h after calves latently infected with BoHV-1 are treated with dexamethasone. Since the immediate early transcription unit 1 (IEtu1) promoter regulates both BoHV-1 infected cell protein 0 (bICP0) and bICP4 expressions, we hypothesized that the bICP4 protein is also expressed during early stages of reactivation from latency. In this study, we tested whether bICP4 and bICP22, the only other BoHV-1 protein known to be encoded by an immediate early gene, were expressed during reactivation from latency by generating peptide-specific antiserum to each protein. bICP4 and bICP22 protein expression were detected in trigeminal ganglionic (TG) neurons during early phases of dexamethasone-induced reactivation from latency, operationally defined as the escape from latency. Conversely, bICP4 and bICP22 were not readily detected in TG neurons of latently infected calves. In summary, it seems clear that all proteins encoded by known BoHV-1 IE genes (bICP4, bICP22, and bICP0) were expressed during early stages of dexamethasone-induced reactivation from latency.


Subject(s)
Gene Expression Regulation, Viral , Herpesvirus 1, Bovine/genetics , Immediate-Early Proteins/genetics , Infectious Bovine Rhinotracheitis/virology , Sensory Receptor Cells/virology , Trigeminal Ganglion/virology , Viral Proteins/genetics , Animals , Antibodies, Viral/chemistry , Cattle , Cell Line , Dexamethasone/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/pathology , Epithelial Cells/virology , Herpesvirus 1, Bovine/growth & development , Herpesvirus 1, Bovine/metabolism , Immediate-Early Proteins/metabolism , Infectious Bovine Rhinotracheitis/pathology , Kidney/drug effects , Kidney/pathology , Kidney/virology , Male , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/pathology , Trigeminal Ganglion/drug effects , Trigeminal Ganglion/metabolism , Trigeminal Ganglion/pathology , Viral Proteins/metabolism , Virus Activation/drug effects , Virus Latency/drug effects
13.
J Virol ; 92(2)2018 01 15.
Article in English | MEDLINE | ID: mdl-29118123

ABSTRACT

Human immunodeficiency virus (HIV) viremia rebounds rapidly after treatment interruption, and a variety of strategies are being explored to reduce or control viral reactivation posttreatment. This viral rebound arises from reactivation of individual latently infected cells, which spread during ongoing rounds of productive infection. The level of virus produced by the initial individual reactivating cells is not known, although it may have major implications for the ability of different immune interventions to control viral rebound. Here we use data from both HIV and simian immunodeficiency virus (SIV) treatment interruption studies to estimate the initial viral load postinterruption and thereby the initial individual reactivation event. Using a barcoded virus (SIVmac239M) to track reactivation from individual latent cells, we use the observed viral growth rates and frequency of reactivation to model the dynamics of reactivation to estimate that a single reactivated latent cell can produce an average viral load equivalent to ∼0.1 to 0.5 viral RNA (vRNA) copies/ml. Modeling of treatment interruption in HIV suggests an initial viral load equivalent of ∼0.6 to 1 vRNA copies/ml. These low viral loads immediately following latent cell reactivation provide a window of opportunity for viral control by host immunity, before further replication allows viral spread. This work shows the initial levels of viral production that must be controlled in order to successfully suppress HIV reactivation following treatment interruption.IMPORTANCE Current treatment for HIV is able to suppress viral replication and prevent disease progression. However, treatment cannot eradicate infection, because the virus lies silent within latently infected cells. If treatment is stopped, the virus usually rebounds above the level of detection within a few weeks. There are a number of approaches being tested aimed at either eradicating latently infected cells or controlling the virus if it returns. Studying both the small pool of latently infected cells and the early events during viral reactivation is difficult, because these involve very small levels of virus that are difficult to measure directly. Here, we combine experimental data and mathematical modeling to understand the very early events during viral reactivation from latency in both HIV infection of humans and SIV infection of monkeys. We find that the initial levels of virus are low, which may help in designing therapies to control early viral reactivation.


Subject(s)
HIV Infections/virology , HIV/physiology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/physiology , Viral Load , Virus Activation , Virus Latency , Algorithms , Animals , Antiretroviral Therapy, Highly Active , HIV Infections/drug therapy , Humans , Models, Biological , Time Factors
14.
Cell Host Microbe ; 20(6): 785-797, 2016 Dec 14.
Article in English | MEDLINE | ID: mdl-27978436

ABSTRACT

A population of CD4 T lymphocytes harboring latent HIV genomes can persist in patients on antiretroviral therapy, posing a barrier to HIV eradication. To examine cellular complexes controlling HIV latency, we conducted a genome-wide screen with a pooled ultracomplex shRNA library and in vitro system modeling HIV latency and identified the mTOR complex as a modulator of HIV latency. Knockdown of mTOR complex subunits or pharmacological inhibition of mTOR activity suppresses reversal of latency in various HIV-1 latency models and HIV-infected patient cells. mTOR inhibitors suppress HIV transcription both through the viral transactivator Tat and via Tat-independent mechanisms. This inhibition occurs at least in part via blocking the phosphorylation of CDK9, a p-TEFb complex member that serves as a cofactor for Tat-mediated transcription. The control of HIV latency by mTOR signaling identifies a pathway that may have significant therapeutic opportunities.


Subject(s)
HIV Infections/virology , HIV-1/drug effects , HIV-1/genetics , TOR Serine-Threonine Kinases/pharmacology , Virus Latency/drug effects , Adaptor Proteins, Signal Transducing/genetics , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/virology , Cell Line , Clustered Regularly Interspaced Short Palindromic Repeats , Cyclin-Dependent Kinase 9/metabolism , Gene Expression Regulation, Viral , Gene Knockdown Techniques , Genes, Viral , HIV-1/physiology , Humans , K562 Cells , Phosphorylation , Positive Transcriptional Elongation Factor B/metabolism , RNA, Small Interfering/genetics , Signal Transduction , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Transcription, Genetic/drug effects , mTOR Associated Protein, LST8 Homolog , tat Gene Products, Human Immunodeficiency Virus
15.
J Neurovirol ; 21(5): 585-91, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25860382

ABSTRACT

Bovine herpesvirus 1 (BHV-1) establishes latency in sensory neurons. The synthetic corticosteroid dexamethasone consistently induces reactivation from latency. Within 90 min after latently infected calves are treated with dexamethasone, two BHV-1 regulatory proteins, BHV-1-infected cell protein 0 (bICP0) and viral protein 16 (VP16), are expressed in the same neuron. In this study, we demonstrate that VP16 and bICP0 can be detected at 22 and 33 min after dexamethasone (DEX) treatment of latently infected calves. However, we were unable to discern whether VP16 or bICP0 was expressed at early times after reactivation. VP16+ neurons consistently express the glucocorticoid receptor suggesting corticosteroid-mediated activation of its receptor rapidly stimulates reactivation from latency.


Subject(s)
Herpes Simplex Virus Protein Vmw65/biosynthesis , Herpesviridae Infections/virology , Herpesvirus 1, Bovine/physiology , Trans-Activators/biosynthesis , Ubiquitin-Protein Ligases/biosynthesis , Virus Activation/physiology , Virus Latency/physiology , Animals , Apoptosis/physiology , Apoptosis/radiation effects , Cattle , Cattle Diseases/virology , Dexamethasone/pharmacology , Glucocorticoids/pharmacology , Immunohistochemistry , In Situ Nick-End Labeling , Sensory Receptor Cells/virology , Trigeminal Ganglion/virology , Virus Activation/drug effects , Virus Latency/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL