1
|
Krakowiak PA, Flores ME, Cuddy SR, Whitford AL, Dochnal SA, Babnis A, Miyake T, Tigano M, Engel DA, Cliffe AR. Co-option of mitochondrial nucleic acid-sensing pathways by HSV-1 UL12.5 for reactivation from latent infection. Proc Natl Acad Sci U S A 2025; 122:e2413965122. [PMID: 39854226 PMCID: PMC11789124 DOI: 10.1073/pnas.2413965122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 12/05/2024] [Indexed: 01/26/2025] Open
Abstract
Although viruses subvert innate immune pathways for their replication, there is evidence they can also co-opt antiviral responses for their benefit. The ubiquitous human pathogen, Herpes simplex virus-1 (HSV-1), encodes a protein (UL12.5) that induces the release of mitochondrial nucleic acid into the cytosol, which activates immune-sensing pathways and reduces productive replication in nonneuronal cells. HSV-1 establishes latency in neurons and can reactivate to cause disease. We found that UL12.5 is required for HSV-1 reactivation in neurons and acts to directly promote viral lytic gene expression during initial exit from latency. Further, the direct activation of innate immune-sensing pathways triggered HSV-1 reactivation and compensated for a lack of UL12.5. Finally, we found that the induction of HSV-1 lytic genes during reactivation required intact RNA- and DNA-sensing pathways, demonstrating that HSV-1 can respond to and active antiviral nucleic acid-sensing pathways to reactivate from a latent infection.
Collapse
Affiliation(s)
- Patryk A. Krakowiak
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA22908
| | - Matthew E. Flores
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA22908
| | - Sean R. Cuddy
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA22908
- Neuroscience Graduate Program, University of Virginia, Charlottesville, VA22908
| | - Abigail L. Whitford
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA22908
| | - Sara A. Dochnal
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA22908
| | - Aleksandra Babnis
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA22908
| | - Tsuyoshi Miyake
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA22908
| | - Marco Tigano
- Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA19107
| | - Daniel A. Engel
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA22908
| | - Anna R. Cliffe
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA22908
| |
Collapse
|
2
|
Cuddy SR, Flores ME, Krakowiak PA, Whitford AL, Dochnal SA, Babnis A, Miyake T, Tigano M, Engel DA, Cliffe AR. Co-option of mitochondrial nucleic acid sensing pathways by HSV-1 UL12.5 for reactivation from latent Infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.06.601241. [PMID: 39005440 PMCID: PMC11245091 DOI: 10.1101/2024.07.06.601241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Although viruses subvert innate immune pathways for their replication, there is evidence they can also co-opt anti-viral responses for their benefit. The ubiquitous human pathogen, Herpes Simplex Virus-1 (HSV-1), encodes a protein (UL12.5) that induces the release of mitochondrial nucleic acid into the cytosol, which activates immune sensing pathways and reduces productive replication in non-neuronal cells. HSV-1 establishes latency in neurons and can reactivate to cause disease. We found that UL12.5 is required for HSV-1 reactivation in neurons and acts to directly promote viral lytic gene expression during initial exit from latency. Further, the direct activation of innate immune sensing pathways triggered HSV reactivation and compensated for a lack of UL12.5. Finally, we found that the induction of HSV-1 lytic genes during reactivation required intact RNA and DNA sensing pathways, demonstrating that HSV-1 can both respond to and active antiviral nucleic acid sensing pathways to reactivate from a latent infection.
Collapse
Affiliation(s)
- Sean R. Cuddy
- Neuroscience Graduate Program, University of Virginia, Charlottesville, VA, 22908
| | - Matthew E. Flores
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, 22908
| | - Patryk A. Krakowiak
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, 22908
| | - Abigail L. Whitford
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, 22908
| | - Sara A. Dochnal
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, 22908
| | - Aleksandra Babnis
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, 22908
| | - Tsuyoshi Miyake
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, 22908
| | - Marco Tigano
- Department of Pathology and Genomic Medicine, Thomas Jefferson University, 1020 Locust Street, Philadelphia 19107
| | - Daniel A. Engel
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, 22908
| | - Anna. R Cliffe
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, 22908
| |
Collapse
|
3
|
Moshirfar M, Kelkar N, Peterson T, Bradshaw J, Parker L, Ronquillo YC, Hoopes PC. The Impact of Antiviral Resistance on Herpetic Keratitis. Eye Contact Lens 2023; 49:127-134. [PMID: 36374154 DOI: 10.1097/icl.0000000000000952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/11/2022] [Indexed: 11/16/2022]
Abstract
ABSTRACT Herpes simplex keratitis resistance to antiviral treatment presents a growing concern. The herpes simplex virus has many different mechanisms of resistance to antiviral treatment, which have been well described. Resistance to acyclovir occurs because of mutations in the viral thymidylate kinase and DNA polymerase that decrease this enzyme's affinity for its substrate. This article discusses factors that explain the prevalence of this resistance, the ability for recurrences in immunocompromised populations, current treatments for acyclovir-resistant herpes simplex keratitis, and novel therapies for this growing concern.
Collapse
Affiliation(s)
- Majid Moshirfar
- Hoopes Vision Research Center (M.M., Y.C.R., P.C.H.), Hoopes Vision, Draper, UT; John A. Moran Eye Center (M.M.), Department of Ophthalmology and Visual Sciences, University of Utah School of Medicine, Salt Lake City, UT; Utah Lions Eye Bank (M.M.), Murray, UT; University of Arizona College of Medicine-Phoenix (N.K.), Phoenix, AZ; and Rocky Vista University College of Osteopathic Medicine (T.P., J.B., L.P.), Ivins, UT
| | | | | | | | | | | | | |
Collapse
|
4
|
Dochnal S, Merchant HY, Schinlever AR, Babnis A, Depledge DP, Wilson AC, Cliffe AR. DLK-Dependent Biphasic Reactivation of Herpes Simplex Virus Latency Established in the Absence of Antivirals. J Virol 2022; 96:e0050822. [PMID: 35608347 PMCID: PMC9215246 DOI: 10.1128/jvi.00508-22] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 04/30/2022] [Indexed: 01/07/2023] Open
Abstract
Understanding the molecular mechanisms of herpes simplex virus 1 (HSV-1) latent infection and reactivation in neurons requires the use of in vitro model systems. Establishing a quiescent infection in cultured neurons is problematic, as any infectious virus released can superinfect the cultures. Previous studies have used the viral DNA replication inhibitor acyclovir to prevent superinfection and promote latency establishment. Data from these previous models have shown that reactivation is biphasic, with an initial phase I expression of all classes of lytic genes, which occurs independently of histone demethylase activity and viral DNA replication but is dependent on the cell stress protein DLK. Here, we describe a new model system using HSV-1 Stayput-GFP, a reporter virus that is defective for cell-to-cell spread and establishes latent infections without the need for acyclovir. The establishment of a latent state requires a longer time frame than previous models using DNA replication inhibitors. This results in a decreased ability of the virus to reactivate using established inducers, and as such, a combination of reactivation triggers is required. Using this system, we demonstrate that biphasic reactivation occurs even when latency is established in the absence of acyclovir. Importantly, phase I lytic gene expression still occurs in a histone demethylase and viral DNA replication-independent manner and requires DLK activity. These data demonstrate that the two waves of viral gene expression following HSV-1 reactivation are independent of secondary infection and not unique to systems that require acyclovir to promote latency establishment. IMPORTANCE Herpes simplex virus-1 (HSV-1) enters a latent infection in neurons and periodically reactivates. Reactivation manifests as a variety of clinical symptoms. Studying latency and reactivation in vitro is invaluable, allowing the molecular mechanisms behind both processes to be targeted by therapeutics that reduce the clinical consequences. Here, we describe a novel in vitro model system using a cell-to-cell spread-defective HSV-1, known as Stayput-GFP, which allows for the study of latency and reactivation at the single neuron level. We anticipate this new model system will be an incredibly valuable tool for studying the establishment and reactivation of HSV-1 latent infection in vitro. Using this model, we find that initial reactivation events are dependent on cellular stress kinase DLK but independent of histone demethylase activity and viral DNA replication. Our data therefore further validate the essential role of DLK in mediating a wave of lytic gene expression unique to reactivation.
Collapse
Affiliation(s)
- Sara Dochnal
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia, USA
| | - Husain Y. Merchant
- Department of Microbiology, New York University School of Medicine, New York, New York, USA
| | - Austin R. Schinlever
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia, USA
| | - Aleksandra Babnis
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia, USA
| | - Daniel P. Depledge
- Department of Microbiology, New York University School of Medicine, New York, New York, USA
| | - Angus C. Wilson
- Department of Microbiology, New York University School of Medicine, New York, New York, USA
| | - Anna R. Cliffe
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
5
|
Hu H, Srinivas KP, Wang S, Chao MV, Lionnet T, Mohr I, Wilson AC, Depledge DP, Huang TT. Single-cell transcriptomics identifies Gadd45b as a regulator of herpesvirus-reactivating neurons. EMBO Rep 2022; 23:e53543. [PMID: 34842321 PMCID: PMC8811635 DOI: 10.15252/embr.202153543] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 11/10/2021] [Accepted: 11/11/2021] [Indexed: 02/05/2023] Open
Abstract
Single-cell RNA sequencing (scRNA-seq) is a powerful technique for dissecting the complexity of normal and diseased tissues, enabling characterization of cell diversity and heterogeneous phenotypic states in unprecedented detail. However, this technology has been underutilized for exploring the interactions between the host cell and viral pathogens in latently infected cells. Herein, we use scRNA-seq and single-molecule sensitivity fluorescent in situ hybridization (smFISH) technologies to investigate host single-cell transcriptome changes upon the reactivation of a human neurotropic virus, herpes simplex virus-1 (HSV-1). We identify the stress sensor growth arrest and DNA damage-inducible 45 beta (Gadd45b) as a critical antiviral host factor that regulates HSV-1 reactivation events in a subpopulation of latently infected primary neurons. We show that distinct subcellular localization of Gadd45b correlates with the viral late gene expression program, as well as the expression of the viral transcription factor, ICP4. We propose that a hallmark of a "successful" or "aborted" HSV-1 reactivation state in primary neurons is determined by a unique subcellular localization signature of the stress sensor Gadd45b.
Collapse
Affiliation(s)
- Hui‐Lan Hu
- Department of Biochemistry & Molecular PharmacologyNew York University School of MedicineNew YorkNYUSA
| | | | - Shuoshuo Wang
- Department of Cell BiologyInstitute for Systems GeneticsNew York University School of MedicineNew YorkNYUSA
| | - Moses V Chao
- Departments of Cell Biology, Physiology & Neuroscience, and PsychiatrySkirball Institute of Biomolecular MedicineNew York University School of MedicineNew YorkNYUSA
| | - Timothee Lionnet
- Department of Cell BiologyInstitute for Systems GeneticsNew York University School of MedicineNew YorkNYUSA
| | - Ian Mohr
- Department of MicrobiologyNew York University School of MedicineNew YorkNYUSA
| | - Angus C Wilson
- Department of MicrobiologyNew York University School of MedicineNew YorkNYUSA
| | - Daniel P Depledge
- Department of MedicineNew York University School of MedicineNew YorkNYUSA
- Present address:
Institute of VirologyHannover Medical SchoolHannoverGermany
| | - Tony T Huang
- Department of Biochemistry & Molecular PharmacologyNew York University School of MedicineNew YorkNYUSA
| |
Collapse
|
6
|
Kondo H, Koshizuka T, Majima R, Takahashi K, Ishioka K, Suzutani T, Inoue N. Characterization of a thiourea derivative that targets viral transactivators of cytomegalovirus and herpes simplex virus type 1. Antiviral Res 2021; 196:105207. [PMID: 34774602 DOI: 10.1016/j.antiviral.2021.105207] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 11/04/2021] [Accepted: 11/09/2021] [Indexed: 11/16/2022]
Abstract
Although currently available antivirals against certain herpesviruses are effective, the development of resistance during long-term use has necessitated the search for seed compounds that work against novel target molecules. In this report, we identified a thiourea derivative compound, 147B3, that inhibits the infection of human cytomegalovirus (HCMV) in fibroblasts and herpes simplex virus type 1 (HSV-1) in Vero cells at a 50% effective concentration of 0.5 μM and 1.9 μM, respectively. Characterization of the compound provided the following clues regarding its mode of action. 1) Time-of-addition and block-release assays showed that 147B3 behaved similarly to ganciclovir. 2) 147B3 reduced the expression of early and late but not immediate-early gene products and the accumulation of viral genomic DNA in both HCMV-infected and HSV-1-infected cells. 3) 147B3 inhibited the HCMV IE2-dependent activation of viral early gene promoters. 4) Four HSV-1 clones resistant to 147B3 were isolated and next-generation sequencing analysis of their genome DNA revealed that all of them had a mutation(s) in the infected cell protein 4 (ICP4) gene, which encodes a viral transcriptional factor. 5) Although 147B3 did not reduce the amount of ICP4 in an immunoblotting analysis, it changed the localization of the ICP4 from the speckles in the nuclei to diffused dots in the cytoplasm. 6) 147B3 did not affect the localization of promyelocytic leukemia (PML) bodies. Our findings suggest that 147B3 targets viral transactivators, potentially through their interaction with factors required for the viral gene expression system.
Collapse
Affiliation(s)
- Hiroki Kondo
- Microbiology and Immunology, Gifu Pharmaceutical University, 1-25-4 Daigaku-Nishi, Gifu-Shi, Gifu, 501-1196, Japan
| | - Tetsuo Koshizuka
- Microbiology and Immunology, Gifu Pharmaceutical University, 1-25-4 Daigaku-Nishi, Gifu-Shi, Gifu, 501-1196, Japan; Center for Highly Advanced Integration of Nano and Life Sciences (G-CHAIN), Gifu University, Japan.
| | - Ryuichi Majima
- Microbiology and Immunology, Gifu Pharmaceutical University, 1-25-4 Daigaku-Nishi, Gifu-Shi, Gifu, 501-1196, Japan
| | - Keita Takahashi
- Microbiology and Immunology, Gifu Pharmaceutical University, 1-25-4 Daigaku-Nishi, Gifu-Shi, Gifu, 501-1196, Japan
| | - Ken Ishioka
- Department of Microbiology, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima, 960-1295, Japan
| | - Tatsuo Suzutani
- Department of Microbiology, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima, 960-1295, Japan
| | - Naoki Inoue
- Microbiology and Immunology, Gifu Pharmaceutical University, 1-25-4 Daigaku-Nishi, Gifu-Shi, Gifu, 501-1196, Japan.
| |
Collapse
|
7
|
Suzich JB, Cuddy SR, Baidas H, Dochnal S, Ke E, Schinlever AR, Babnis A, Boutell C, Cliffe AR. PML-NB-dependent type I interferon memory results in a restricted form of HSV latency. EMBO Rep 2021; 22:e52547. [PMID: 34197022 PMCID: PMC8419685 DOI: 10.15252/embr.202152547] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 06/02/2021] [Accepted: 06/08/2021] [Indexed: 01/23/2023] Open
Abstract
Herpes simplex virus (HSV) establishes latent infection in long-lived neurons. During initial infection, neurons are exposed to multiple inflammatory cytokines but the effects of immune signaling on the nature of HSV latency are unknown. We show that initial infection of primary murine neurons in the presence of type I interferon (IFN) results in a form of latency that is restricted for reactivation. We also find that the subnuclear condensates, promyelocytic leukemia nuclear bodies (PML-NBs), are absent from primary sympathetic and sensory neurons but form with type I IFN treatment and persist even when IFN signaling resolves. HSV-1 genomes colocalize with PML-NBs throughout a latent infection of neurons only when type I IFN is present during initial infection. Depletion of PML prior to or following infection does not impact the establishment latency; however, it does rescue the ability of HSV to reactivate from IFN-treated neurons. This study demonstrates that viral genomes possess a memory of the IFN response during de novo infection, which results in differential subnuclear positioning and ultimately restricts the ability of genomes to reactivate.
Collapse
Affiliation(s)
- Jon B Suzich
- Department of Microbiology, Immunology and Cancer BiologyUniversity of VirginiaCharlottesvilleVAUSA
| | - Sean R Cuddy
- Neuroscience Graduate ProgramUniversity of VirginiaCharlottesvilleVAUSA
| | - Hiam Baidas
- Department of Microbiology, Immunology and Cancer BiologyUniversity of VirginiaCharlottesvilleVAUSA
| | - Sara Dochnal
- Department of Microbiology, Immunology and Cancer BiologyUniversity of VirginiaCharlottesvilleVAUSA
| | - Eugene Ke
- Department of Microbiology, Immunology and Cancer BiologyUniversity of VirginiaCharlottesvilleVAUSA
| | - Austin R Schinlever
- Department of Microbiology, Immunology and Cancer BiologyUniversity of VirginiaCharlottesvilleVAUSA
| | - Aleksandra Babnis
- Department of Microbiology, Immunology and Cancer BiologyUniversity of VirginiaCharlottesvilleVAUSA
| | - Chris Boutell
- MRC‐University of Glasgow Centre for Virus Research (CVR)GlasgowUK
| | - Anna R Cliffe
- Department of Microbiology, Immunology and Cancer BiologyUniversity of VirginiaCharlottesvilleVAUSA
| |
Collapse
|
8
|
Role of the Herpes Simplex Virus CVSC Proteins at the Capsid Portal Vertex. J Virol 2020; 94:JVI.01534-20. [PMID: 32967953 DOI: 10.1128/jvi.01534-20] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 09/16/2020] [Indexed: 12/31/2022] Open
Abstract
The packaging of DNA into preformed capsids is a critical step during herpesvirus infection. For herpes simplex virus, this process requires the products of seven viral genes: the terminase proteins pUL15, pUL28, and pUL33; the capsid vertex-specific component (CVSC) proteins pUL17 and pUL25; and the portal proteins pUL6 and pUL32. The pUL6 portal dodecamer is anchored at one vertex of the capsid by interactions with the adjacent triplexes as well as helical density attributed to the pUL17 and pUL25 subunits of the CVSC. To define the roles and structures of the CVSC proteins in virus assembly and DNA packaging, we isolated a number of recombinant viruses expressing pUL25, pUL17, and pUL36 fused with green or red fluorescent proteins as well as viruses with specific deletions in the CVSC genes. Biochemical and structural studies of these mutants demonstrated that (i) four of the helices in the CVSC helix bundle can be attributed to two copies each of pUL36 and pUL25, (ii) pUL17 and pUL6 are required for capsid binding of the terminase complex in the nucleus, (iii) pUL17 is important for determining the site of the first cleavage reaction generating replicated genomes with termini derived from the long-arm component of the herpes simplex virus 1 (HSV-1) genome, (iv) pUL36 serves no direct role in cleavage/packaging, (v) cleavage and stable packaging of the viral genome involve an ordered interaction of the terminase complex and pUL25 with pUL17 at the portal vertex, and (vi) packaging of the viral genome results in a dramatic displacement of the portal.IMPORTANCE Herpes simplex virus 1 (HSV-1) is the causative agent of several pathologies ranging in severity from the common cold sore to life-threatening encephalitic infection. A critical step during productive HSV-1 infection is the cleavage and packaging of replicated, concatemeric viral DNA into preformed capsids. A key knowledge gap is how the capsid engages the replicated viral genome and the subsequent packaging of a unit-length HSV genome. Here, biochemical and structural studies focused on the unique portal vertex of wild-type HSV and packaging mutants provide insights into the mechanism of HSV genome packaging. The significance of our research is in identifying the portal proteins pUL6 and pUL17 as key viral factors for engaging the terminase complex with the capsid and the subsequent cleavage, packaging, and stable incorporation of the viral genome in the HSV-1 capsid.
Collapse
|
9
|
Keil T, Liu D, Lloyd M, Coombs W, Moffat J, Visalli R. DNA Encapsidation and Capsid Assembly Are Underexploited Antiviral Targets for the Treatment of Herpesviruses. Front Microbiol 2020; 11:1862. [PMID: 32903425 PMCID: PMC7434925 DOI: 10.3389/fmicb.2020.01862] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 07/15/2020] [Indexed: 12/21/2022] Open
Abstract
Although there are effective nucleoside analogs to treat HSV, VZV, and HCMV disease, herpesvirus infections continue to contribute to significant morbidity and mortality. Acyclovir is the drug of choice for HSV encephalopathy, yet there is an estimated 6-19% mortality rate with half of the survivors experiencing moderate to severe chronic neurological deficits. For VZV, current treatments are inadequate to prevent acute and persistent pain due to zoster. Treatment of HCMV with GCV requires close monitoring particularly in patients with impaired renal function and there are no approved treatments for congenital HCMV infections. New therapeutic options to control cytomegalovirus reactivation in bone marrow and stem cell transplant patients are needed to improve patient outcome. No successful chemotherapeutic options are available for EBV, HHV-6, 7, and 8. Drug resistance is a concern for HCMV, HSV, and VZV since approved drugs share common mechanisms of action. Targeting DNA encapsidation or capsid assembly provide additional options for the development of non-nucleoside, small molecule anti-herpesviral drugs.
Collapse
Affiliation(s)
- Tara Keil
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, GA, United States
| | - Dongmei Liu
- Department of Microbiology and Immunology, Upstate Medical University, Syracuse, NY, United States
| | - Megan Lloyd
- Department of Microbiology and Immunology, Upstate Medical University, Syracuse, NY, United States
| | - Wanda Coombs
- Department of Microbiology and Immunology, Upstate Medical University, Syracuse, NY, United States
| | - Jennifer Moffat
- Department of Microbiology and Immunology, Upstate Medical University, Syracuse, NY, United States
| | - Robert Visalli
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, GA, United States
| |
Collapse
|
10
|
Translation of the long-term fundamental studies on viral DNA packaging motors into nanotechnology and nanomedicine. SCIENCE CHINA-LIFE SCIENCES 2020; 63:1103-1129. [DOI: 10.1007/s11427-020-1752-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 06/04/2020] [Indexed: 02/07/2023]
|
11
|
Molecular anatomy of the subcellular localization and nuclear import mechanism of herpes simplex virus 1 UL6. Aging (Albany NY) 2020; 12:5751-5763. [PMID: 32235005 PMCID: PMC7185102 DOI: 10.18632/aging.102965] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 02/08/2020] [Indexed: 12/24/2022]
Abstract
As an indispensable structure protein, the herpes simplex virus 1 (HSV-1) UL6 has been described to exert numerous roles in viral proliferation. However, its exact subcellular localization and subcellular transport mechanism is not well known. In the present study, by utilizing confocal fluorescent microscopy, UL6 was shown to mainly locate in the nucleus in enhanced yellow fluorescent protein or Flag tag fused expression plasmid-transfected cells or HSV-1-infected cells, whereas its predicted nuclear localization signal was nonfunctional. In addition, by exploiting dominant negative mutant and inhibitor of different nuclear import receptors, as well as co-immunoprecipitation and RNA interference assays, UL6 was established to interact with importin α1, importin α7 and transportin-1 to mediate its nuclear translocation under the help of Ran-mediated GTP hydrolysis. Accordingly, these results will advance the knowledge of UL6-mediated biological significances in HSV-1 infection cycle.
Collapse
|
12
|
Dedeo CL, Cingolani G, Teschke CM. Portal Protein: The Orchestrator of Capsid Assembly for the dsDNA Tailed Bacteriophages and Herpesviruses. Annu Rev Virol 2019; 6:141-160. [PMID: 31337287 PMCID: PMC6947915 DOI: 10.1146/annurev-virology-092818-015819] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Tailed, double-stranded DNA bacteriophages provide a well-characterized model system for the study of viral assembly, especially for herpesviruses and adenoviruses. A wealth of genetic, structural, and biochemical work has allowed for the development of assembly models and an understanding of the DNA packaging process. The portal complex is an essential player in all aspects of bacteriophage and herpesvirus assembly. Despite having low sequence similarity, portal structures across bacteriophages share the portal fold and maintain a conserved function. Due to their dynamic role, portal proteins are surprisingly plastic, and their conformations change for each stage of assembly. Because the maturation process is dependent on the portal protein, researchers have been working to validate this protein as a potential antiviral drug target. Here we review recent work on the role of portal complexes in capsid assembly, including DNA packaging, as well as portal ring assembly and incorporation and analysis of portal structures.
Collapse
Affiliation(s)
- Corynne L Dedeo
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, Connecticut 06269, USA;
| | - Gino Cingolani
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, USA
| | - Carolyn M Teschke
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, Connecticut 06269, USA;
- Department of Chemistry, University of Connecticut, Storrs, Connecticut 06269, USA
| |
Collapse
|
13
|
Dünn-Kittenplon DD, Kalt I, Lellouche JPM, Sarid R. The KSHV portal protein ORF43 is essential for the production of infectious viral particles. Virology 2019; 529:205-215. [PMID: 30735904 DOI: 10.1016/j.virol.2019.01.028] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 01/13/2019] [Accepted: 01/21/2019] [Indexed: 02/09/2023]
Abstract
Herpesvirus capsid assembly involves cleavage and packaging of the viral genome. The Kaposi's sarcoma-associated herpesvirus (KSHV) open reading frame 43 (orf43) encodes a putative portal protein. The portal complex functions as a gate through which DNA is packaged into the preformed procapsids, and is injected into the cell nucleus upon infection. The amino acid sequence of the portal proteins is conserved among herpesviruses. Here, we generated an antiserum to ORF43 and determined late expression kinetics of ORF43 along with its nuclear localization. We generated a recombinant KSHV mutant, which fails to express ORF43 (BAC16-ORF43-null). Assembled capsids were observed upon lytic induction of this virus; however, the released virions lacked viral DNA and thus could not establish infection. Ectopic expression of ORF43 rescued the ability to produce infectious particles. ORF43 antiserum and the recombinant ORF43-null virus can provide an experimental system for further studies of the portal functions and its interactions.
Collapse
Affiliation(s)
- Daniela Dana Dünn-Kittenplon
- The Mina and Everard Goodman Faculty of Life Sciences, Bar Ilan University, Ramat-Gan 5290002, Israel; Department of Chemistry, Bar Ilan University, Ramat-Gan 5290002, Israel; Advanced Materials and Nanotechnology Institute, Bar Ilan University, Ramat-Gan 5290002, Israel
| | - Inna Kalt
- The Mina and Everard Goodman Faculty of Life Sciences, Bar Ilan University, Ramat-Gan 5290002, Israel; Advanced Materials and Nanotechnology Institute, Bar Ilan University, Ramat-Gan 5290002, Israel
| | - Jean-Paul Moshe Lellouche
- Department of Chemistry, Bar Ilan University, Ramat-Gan 5290002, Israel; Advanced Materials and Nanotechnology Institute, Bar Ilan University, Ramat-Gan 5290002, Israel
| | - Ronit Sarid
- The Mina and Everard Goodman Faculty of Life Sciences, Bar Ilan University, Ramat-Gan 5290002, Israel; Advanced Materials and Nanotechnology Institute, Bar Ilan University, Ramat-Gan 5290002, Israel.
| |
Collapse
|
14
|
Visalli RJ, Schwartz AM, Patel S, Visalli MA. Identification of the Epstein Barr Virus portal. Virology 2019; 529:152-159. [PMID: 30710799 DOI: 10.1016/j.virol.2019.01.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 12/21/2018] [Accepted: 01/03/2019] [Indexed: 12/12/2022]
Abstract
Little is known about Epstein Barr Virus (EBV) proteins that participate in viral DNA cleavage and packaging. Genes encoding potential terminase subunit and portal protein homologs include BGRF1/BDRF1, BALF3, BFRF1A and BBRF1 respectively. EBV mutants with deletions in one or more of these genes were impaired for DNA packaging (Pavlova et al., 2013). In the current study, BBRF1 oligomers were purified from recombinant baculovirus infected insect cell extracts. Transmission electron microscopy revealed that purified EBV portals retained features typically found in other portals including a central channel with clip, stem and wing/crown domains. Although compounds have been identified that target DNA encapsidation in human cytomegalovirus, herpes simplex viruses and varicella-zoster virus, the identification of new EBV targets has lagged significantly. Characterization of the EBV portal will direct studies aimed at developing potential small molecular inhibitors of the EBV encapsidation process.
Collapse
Affiliation(s)
- Robert J Visalli
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, GA 31404, USA.
| | - Adam M Schwartz
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, GA 31404, USA
| | - Shivam Patel
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, GA 31404, USA
| | - Melissa A Visalli
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, GA 31404, USA
| |
Collapse
|
15
|
Miller JT, Zhao H, Masaoka T, Varnado B, Cornejo Castro EM, Marshall VA, Kouhestani K, Lynn AY, Aron KE, Xia A, Beutler JA, Hirsch DR, Tang L, Whitby D, Murelli RP, Le Grice SFJ. Sensitivity of the C-Terminal Nuclease Domain of Kaposi's Sarcoma-Associated Herpesvirus ORF29 to Two Classes of Active-Site Ligands. Antimicrob Agents Chemother 2018; 62:e00233-18. [PMID: 30061278 PMCID: PMC6153795 DOI: 10.1128/aac.00233-18] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 07/19/2018] [Indexed: 01/03/2023] Open
Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV), the etiological agent of Kaposi's sarcoma, belongs to the Herpesviridae family, whose members employ a multicomponent terminase to resolve nonparametric viral DNA into genome-length units prior to their packaging. Homology modeling of the ORF29 C-terminal nuclease domain (pORF29C) and bacteriophage Sf6 gp2 have suggested an active site clustered with four acidic residues, D476, E550, D661, and D662, that collectively sequester the catalytic divalent metal (Mn2+) and also provided important insight into a potential inhibitor binding mode. Using this model, we have expressed, purified, and characterized the wild-type pORF29C and variants with substitutions at the proposed active-site residues. Differential scanning calorimetry demonstrated divalent metal-induced stabilization of wild-type (WT) and D661A pORF29C, consistent with which these two enzymes exhibited Mn2+-dependent nuclease activity, although the latter mutant was significantly impaired. Thermal stability of WT and D661A pORF29C was also enhanced by binding of an α-hydroxytropolone (α-HT) inhibitor shown to replace divalent metal at the active site. For the remaining mutants, thermal stability was unaffected by divalent metal or α-HT binding, supporting their role in catalysis. pORF29C nuclease activity was also inhibited by two classes of small molecules reported to inhibit HIV RNase H and integrase, both of which belong to the superfamily of nucleotidyltransferases. Finally, α-HT inhibition of KSHV replication suggests ORF29 nuclease function as an antiviral target that could be combined with latency-activating compounds as a shock-and-kill antiviral strategy.
Collapse
Affiliation(s)
- Jennifer T Miller
- Basic Research Laboratory, National Cancer Institute, Frederick, Maryland, USA
| | - Haiyan Zhao
- Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas, USA
| | - Takashi Masaoka
- Basic Research Laboratory, National Cancer Institute, Frederick, Maryland, USA
| | - Brittany Varnado
- Department of Chemistry, Brooklyn College, City University of New York, Brooklyn, New York, USA
| | - Elena M Cornejo Castro
- Viral Oncology Section, AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute, Frederick, Maryland, USA
| | - Vickie A Marshall
- Viral Oncology Section, AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute, Frederick, Maryland, USA
| | - Kaivon Kouhestani
- Basic Research Laboratory, National Cancer Institute, Frederick, Maryland, USA
| | - Anna Y Lynn
- Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas, USA
| | - Keith E Aron
- Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas, USA
| | - Anqi Xia
- Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas, USA
| | - John A Beutler
- Molecular Targets Program, National Cancer Institute, Frederick, Maryland, USA
| | - Danielle R Hirsch
- Department of Chemistry, Brooklyn College, City University of New York, Brooklyn, New York, USA
- Molecular Targets Program, National Cancer Institute, Frederick, Maryland, USA
| | - Liang Tang
- Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas, USA
| | - Denise Whitby
- Viral Oncology Section, AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute, Frederick, Maryland, USA
| | - Ryan P Murelli
- Department of Chemistry, Brooklyn College, City University of New York, Brooklyn, New York, USA
- Ph.D. Program in Chemistry, The Graduate Center of The City University of New York, New York, New York, USA
| | - Stuart F J Le Grice
- Basic Research Laboratory, National Cancer Institute, Frederick, Maryland, USA
| |
Collapse
|
16
|
Andronova VL. MODERN ETHIOTROPIC CHEMOTHERAPY OF HERPESVIRUS INFECTIONS: ADVANCES, NEW TRENDS AND PERSPECTIVES. ALPHAHERPESVIRUSES (PART II). Vopr Virusol 2018; 63:149-159. [PMID: 36494970 DOI: 10.18821/0507-4088-2018-63-4-149-159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Indexed: 12/13/2022]
Abstract
A key role in the treatment of herpesviral infections is played by modified nucleosides and their predecessors - acyclovir, its L-valine ester (valaciclovir) and famciclovir (prodrug of penciclovir). The biological activity of compounds of this class is determined by their similarity to natural nucleosides. After phosphorylation by viral thymidine kinase and then cell enzymes to the triphosphate forms, acyclovir and penciclovir inhibit the activity of viral DNA polymerase and synthesis of viral DNA. The increasing role of herpesvirus infections in human infectious pathology, as well as the development of drug resistance in viruses, mainly in patients with immunodeficiencies of various origins, necessitate the search for new compounds possessing anti-herpesvirus activity, using as a biological target not DNA polymerase, but other viral proteins and enzymes, unique or different from cellular proteins, performing similar functions.
Collapse
Affiliation(s)
- V L Andronova
- National Research Center for Epidemiology and Microbiology named after the honorary academician N.F. Gamaleya
| |
Collapse
|
17
|
Yasui R, Yoshida C, Yamaguchi T, Inoue N. Characterization of an anti-varicella-zoster virus compound that targets the portal protein encoded by ORF54. Microbiol Immunol 2018; 61:398-402. [PMID: 28833387 DOI: 10.1111/1348-0421.12507] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2017] [Revised: 07/05/2017] [Accepted: 08/12/2017] [Indexed: 12/01/2022]
Abstract
An anti-varicella-zoster virus compound, a 5-chlorobenzo[b]thiophen derivative (45B5), was characterized. Its 50% effective concentration against the cell-free vaccine Oka strain and 50% cytotoxic concentration in human fibroblasts were 16.9 µM and more than 100 µM, respectively. Treatment with 45B5 decreased viral DNA synthesis and IE62 expression weakly but significantly. All 45B5-resistant viral clones isolated were found to have at least one mutation in ORF54 that encodes the portal protein. There were no effects on interaction between the portal and scaffold proteins. Thus, 45B5 may inhibit nuclear delivery of viral DNA.
Collapse
Affiliation(s)
- Ruka Yasui
- Department of Microbiology and Immunology, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu-Shi, Gifu 501-1196, Japan
| | - Chinatsu Yoshida
- Department of Microbiology and Immunology, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu-Shi, Gifu 501-1196, Japan
| | - Toyofumi Yamaguchi
- Department of Biosciences, Teikyo University of Science, 2-2-1 Senju-Sakuragi, Adachi-ku, Tokyo 120-0045, Japan
| | - Naoki Inoue
- Department of Microbiology and Immunology, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu-Shi, Gifu 501-1196, Japan
| |
Collapse
|
18
|
Kornfeind EM, Visalli RJ. Human herpesvirus portal proteins: Structure, function, and antiviral prospects. Rev Med Virol 2018; 28:e1972. [PMID: 29573302 DOI: 10.1002/rmv.1972] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 01/26/2018] [Accepted: 01/27/2018] [Indexed: 01/28/2023]
Abstract
Herpesviruses (Herpesvirales) and tailed bacteriophages (Caudovirales) package their dsDNA genomes through an evolutionarily conserved mechanism. Much is known about the biochemistry and structural biology of phage portal proteins and the DNA encapsidation (viral genome cleavage and packaging) process. Although not at the same level of detail, studies on HSV-1, CMV, VZV, and HHV-8 have revealed important information on the function and structure of herpesvirus portal proteins. During dsDNA phage and herpesviral genome replication, concatamers of viral dsDNA are cleaved into single length units by a virus-encoded terminase and packaged into preformed procapsids through a channel located at a single capsid vertex (portal). Oligomeric portals are formed by the interaction of identical portal protein monomers. Comparing portal protein primary aa sequences between phage and herpesviruses reveals little to no sequence similarity. In contrast, the secondary and tertiary structures of known portals are remarkable. In all cases, function is highly conserved in that portals are essential for DNA packaging and also play a role in releasing viral genomic DNA during infection. Preclinical studies have described small molecules that target the HSV-1 and VZV portals and prevent viral replication by inhibiting encapsidation. This review summarizes what is known concerning the structure and function of herpesvirus portal proteins primarily based on their conserved bacteriophage counterparts and the potential to develop novel portal-specific DNA encapsidation inhibitors.
Collapse
Affiliation(s)
- Ellyn M Kornfeind
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, GA, USA
| | - Robert J Visalli
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, GA, USA
| |
Collapse
|
19
|
Cliffe AR, Arbuckle JH, Vogel JL, Geden MJ, Rothbart SB, Cusack CL, Strahl BD, Kristie TM, Deshmukh M. Neuronal Stress Pathway Mediating a Histone Methyl/Phospho Switch Is Required for Herpes Simplex Virus Reactivation. Cell Host Microbe 2016; 18:649-58. [PMID: 26651941 DOI: 10.1016/j.chom.2015.11.007] [Citation(s) in RCA: 112] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Revised: 10/13/2015] [Accepted: 11/19/2015] [Indexed: 01/27/2023]
Abstract
Herpes simplex virus (HSV) reactivation from latent neuronal infection requires stimulation of lytic gene expression from promoters associated with repressive heterochromatin. Various neuronal stresses trigger reactivation, but how these stimuli activate silenced promoters remains unknown. We show that a neuronal pathway involving activation of c-Jun N-terminal kinase (JNK), common to many stress responses, is essential for initial HSV gene expression during reactivation. This JNK activation in neurons is mediated by dual leucine zipper kinase (DLK) and JNK-interacting protein 3 (JIP3), which direct JNK toward stress responses instead of other cellular functions. Surprisingly, JNK-mediated viral gene induction occurs independently of histone demethylases that remove repressive lysine modifications. Rather, JNK signaling results in a histone methyl/phospho switch on HSV lytic promoters, a mechanism permitting gene expression in the presence of repressive lysine methylation. JNK is present on viral promoters during reactivation, thereby linking a neuronal-specific stress pathway and HSV reactivation from latency.
Collapse
Affiliation(s)
- Anna R Cliffe
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Jesse H Arbuckle
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jodi L Vogel
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Matthew J Geden
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Scott B Rothbart
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Corey L Cusack
- Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Neurobiology Curriculum, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Brian D Strahl
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Thomas M Kristie
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Mohanish Deshmukh
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
20
|
Pires de Mello CP, Bloom DC, Paixão IC. Herpes simplex virus type-1: replication, latency, reactivation and its antiviral targets. Antivir Ther 2016; 21:277-86. [PMID: 26726828 DOI: 10.3851/imp3018] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/17/2015] [Indexed: 10/22/2022]
Abstract
Infection by herpes simplex virus type-1 (HSV-1) causes several diseases, ranging from cutaneous, oral and genital infections to fatal encephalitis. Despite the availability of antiviral therapies on the market, their efficacies are incomplete, and new cases of resistant strains arise, mainly in the immunocompromised, but also recently documented in immunocompetent patients. Over the last decades a lot has been discovered about the molecular basis of infection which has been of great benefit to the investigation of new anti-HSV-1 molecules. In this review we summarize replication, latency and reactivation highlighting potential antiviral targets and new molecules described in the past several years in the literature.
Collapse
Affiliation(s)
- Camilly P Pires de Mello
- Department of Cellular and Molecular Biology, Universidade Federal Fluminense, Biology Institute, Niterói, Brazil
| | | | | |
Collapse
|
21
|
Intermolecular Complementation between Two Varicella-Zoster Virus pORF30 Terminase Domains Essential for DNA Encapsidation. J Virol 2015. [PMID: 26202238 DOI: 10.1128/jvi.01313-15] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
UNLABELLED The herpesviral terminase complex is part of the intricate machinery that delivers a single viral genome into empty preformed capsids (encapsidation). The varicella-zoster virus (VZV) terminase components (pORF25, pORF30, and pORF45/42) have not been studied as extensively as those of herpes simplex virus 1 and human cytomegalovirus (HCMV). In this study, VZV bacterial artificial chromosomes (BACs) were generated with small (Δ30S), medium (Δ30M), and large (Δ30L) ORF30 internal deletions. In addition, we isolated recombinant viruses with specific alanine substitutions in the putative zinc finger motif (30-ZF3A) or in a conserved region (region IX) with predicted structural similarity to the human topoisomerase I core subdomains I and II (30-IXAla, 30-620A, and 30-622A). Recombinant viruses replicated in an ORF30-complementing cell line (ARPE30) but failed to replicate in noncomplementing ARPE19 and MeWo cells. Transmission electron microscopy of 30-IXAla-, 30-620A-, and 30-622A-infected ARPE19 cells revealed only empty VZV capsids. Southern analysis showed that cells infected with parental VZV (VZVLUC) or a repaired virus (30R) contained DNA termini, whereas cells infected with Δ30L, 30-IXAla, 30-620A, or 30-622A contained little or no processed viral DNA. These results demonstrated that pORF30, specifically amino acids 619 to 624 (region IX), was required for DNA encapsidation. A luciferase-based assay was employed to assess potential intermolecular complementation between the zinc finger domain and conserved region IX. Complementation between 30-ZF3A and 30-IXAla provided evidence that distinct pORF30 domains can function independently. The results suggest that pORF30 may exist as a multimer or participate in higher-order assemblies during viral DNA encapsidation. IMPORTANCE Antivirals with novel mechanisms of action are sought as additional therapeutic options to treat human herpesvirus infections. Proteins involved in the viral DNA encapsidation process have become promising antiviral targets. For example, letermovir is a small-molecule drug targeting HCMV terminase that is currently in phase III clinical trials. It is important to define the structural and functional characteristics of proteins that make up viral terminase complexes to identify or design additional terminase-specific compounds. The VZV ORF30 mutants described in this study represent the first VZV terminase mutants reported to date. Targeted mutations confirmed the importance of a conserved zinc finger domain found in all herpesvirus ORF30 terminase homologs but also identified a novel, highly conserved region (region IX) essential for terminase function. Homology modeling suggested that the structure of region IX is present in all human herpesviruses and thus represents a potential structurally conserved antiviral target.
Collapse
|
22
|
Gable J, Acker TM, Craik CS. Current and potential treatments for ubiquitous but neglected herpesvirus infections. Chem Rev 2014; 114:11382-412. [PMID: 25275644 PMCID: PMC4254030 DOI: 10.1021/cr500255e] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Indexed: 02/07/2023]
Affiliation(s)
- Jonathan
E. Gable
- Department
of Pharmaceutical Chemistry, University
of California, San Francisco, 600 16th Street, San Francisco, California 94158-2280, United States
- Graduate
Group in Biophysics, University of California,
San Francisco, 600 16th
Street, San Francisco, California 94158-2280, United States
| | - Timothy M. Acker
- Department
of Pharmaceutical Chemistry, University
of California, San Francisco, 600 16th Street, San Francisco, California 94158-2280, United States
| | - Charles S. Craik
- Department
of Pharmaceutical Chemistry, University
of California, San Francisco, 600 16th Street, San Francisco, California 94158-2280, United States
| |
Collapse
|
23
|
Hornig J, McGregor A. Design and development of antivirals and intervention strategies against human herpesviruses using high-throughput approach. Expert Opin Drug Discov 2014; 9:891-915. [DOI: 10.1517/17460441.2014.922538] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
24
|
The varicella-zoster virus portal protein is essential for cleavage and packaging of viral DNA. J Virol 2014; 88:7973-86. [PMID: 24807720 DOI: 10.1128/jvi.00376-14] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The varicella-zoster virus (VZV) open reading frame 54 (ORF54) gene encodes an 87-kDa monomer that oligomerizes to form the VZV portal protein, pORF54. pORF54 was hypothesized to perform a function similar to that of a previously described herpes simplex virus 1 (HSV-1) homolog, pUL6. pUL6 and the associated viral terminase are required for processing of concatemeric viral DNA and packaging of individual viral genomes into preformed capsids. In this report, we describe two VZV bacterial artificial chromosome (BAC) constructs with ORF54 gene deletions, Δ54L (full ORF deletion) and Δ54S (partial internal deletion). The full deletion of ORF54 likely disrupted essential adjacent genes (ORF53 and ORF55) and therefore could not be complemented on an ORF54-expressing cell line (ARPE54). In contrast, Δ54S was successfully propagated in ARPE54 cells but failed to replicate in parental, noncomplementing ARPE19 cells. Transmission electron microscopy confirmed the presence of only empty VZV capsids in Δ54S-infected ARPE19 cell nuclei. Similar to the HSV-1 genome, the VZV genome is composed of a unique long region (UL) and a unique short region (US) flanked by inverted repeats. DNA from cells infected with parental VZV (VZVLUC strain) contained the predicted UL and US termini, whereas cells infected with Δ54S contained neither. This result demonstrates that Δ54S is not able to process and package viral DNA, thus making pORF54 an excellent chemotherapeutic target. In addition, the utility of BAC constructs Δ54L and Δ54S as tools for the isolation of site-directed ORF54 mutants was demonstrated by recombineering single-nucleotide changes within ORF54 that conferred resistance to VZV-specific portal protein inhibitors. Importance: Antivirals with novel mechanisms of action would provide additional therapeutic options to treat human herpesvirus infections. Proteins involved in the herpesviral DNA encapsidation process have become promising antiviral targets. Previously, we described a series of N-α-methylbenzyl-N'-aryl thiourea analogs that target the VZV portal protein (pORF54) and prevent viral replication in vitro. To better understand the mechanism of action of these compounds, it is important to define the structural and functional characteristics of the VZV portal protein. In contrast to HSV, no VZV mutants have been described for any of the seven essential DNA encapsidation genes. The VZV ORF54 deletion mutant described in this study represents the first VZV encapsidation mutant reported to date. We demonstrate that the deletion mutant can serve as a platform for the isolation of portal mutants via recombineering and provide a strategy for more in-depth studies of VZV portal structure and function.
Collapse
|
25
|
New Herpes Simplex Virus Replication Targets. Antiviral Res 2014. [DOI: 10.1128/9781555815493.ch20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
26
|
Visalli RJ, Howard AJ. Non-axial view of the varicella-zoster virus portal protein reveals conserved crown, wing and clip architecture. Intervirology 2014; 57:121-5. [PMID: 24642670 DOI: 10.1159/000360225] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Accepted: 01/31/2014] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Herpesviridae encode a family of protein homologues that function as the 'port of entry' for insertion of the viral DNA into preformed capsids during encapsidation. METHODS Transmission electron microscopy (TEM) of recombinant varicella-zoster virus pORF54 was performed. RESULTS Results suggest that pORF54 forms higher-order structures with itself. Enriched fractions analyzed by TEM revealed non-axial oriented portals with defined central channels and distinguishable crown, wing and clip regions. CONCLUSION These morphological features are consistent with those previously reported for other herpesvirus and bacteriophage portal proteins.
Collapse
Affiliation(s)
- Robert J Visalli
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, Ga., USA
| | | |
Collapse
|
27
|
Yang K, Wills E, Baines JD. A herpes simplex virus scaffold peptide that binds the portal vertex inhibits early steps in viral replication. J Virol 2013; 87:6876-87. [PMID: 23576509 PMCID: PMC3676109 DOI: 10.1128/jvi.00421-13] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Accepted: 04/03/2013] [Indexed: 12/12/2022] Open
Abstract
Previous experiments identified a 12-amino-acid (aa) peptide that was sufficient to interact with the herpes simplex virus 1 (HSV-1) portal protein and was necessary to incorporate the portal into capsids. In the present study, cells were treated at various times postinfection with peptides consisting of a portion of the Drosophila antennapedia protein, previously shown to enter cells efficiently, fused to either wild-type HSV-1 scaffold peptide (YPYYPGEARGAP) or a control peptide that contained changes at positions 4 and 5. These 4-tyrosine and 5-proline residues are highly conserved in herpesvirus scaffold proteins and were previously shown to be critical for the portal interaction. Treatment early in infection with subtoxic levels of wild-type peptide reduced viral infectivity by over 1,000-fold, while the mutant peptide had little effect on viral yields. In cells infected for 3 h in the presence of wild-type peptide, capsids were observed to transit to the nuclear rim normally, as viewed by fluorescence microscopy. However, observation by electron microscopy in thin sections revealed an aberrant and significant increase of DNA-containing capsids compared to infected cells treated with the mutant peptide. Early treatment with peptide also prevented formation of viral DNA replication compartments. These data suggest that the antiviral peptide stabilizes capsids early in infection, causing retention of DNA within them, and that this activity correlates with peptide binding to the portal protein. The data are consistent with the hypothesis that the portal vertex is the conduit through which DNA is ejected to initiate infection.
Collapse
Affiliation(s)
- Kui Yang
- Department of Microbiology and Immunology, Cornell University, Ithaca, New York, USA
| | | | | |
Collapse
|
28
|
Wang JB, Zhu Y, McVoy MA, Parris DS. Changes in subcellular localization reveal interactions between human cytomegalovirus terminase subunits. Virol J 2012; 9:315. [PMID: 23259714 PMCID: PMC3547700 DOI: 10.1186/1743-422x-9-315] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2012] [Accepted: 12/14/2012] [Indexed: 11/15/2022] Open
Abstract
Background During herpesvirus replication, terminase packages viral DNA into capsids. The subunits of herpes simplex virus terminase, UL15, UL28, and UL33, assemble in the cytoplasm prior to nuclear import of the complex. Methods To detect similar interactions between human cytomegalovirus terminase subunits, the orthologous proteins UL89, UL56, and UL51 were expressed in HEK-293 T cells (via transfection) or insect cells (via baculovirus infection) and subcellular localizations were detected by cellular fractionation and confocal microscopy. Results In both cell types, UL56 and UL89 expressed alone were exclusively cytoplasmic, whereas UL51 was ~50% nuclear. Both UL89 and UL56 became ~50% nuclear when expressed together, as did UL56 when expressed with UL51. Nuclear localization of each protein was greatest when all three proteins were co-expressed. Conclusions These results support inclusion of UL51 as an HCMV terminase subunit and suggest that nuclear import of human cytomegalovirus terminase may involve nuclear import signals that form cooperatively upon subunit associations.
Collapse
Affiliation(s)
- Jian Ben Wang
- Department of Pediatrics, Virginia Commonwealth University, Richmond, VA 23298-0163, USA
| | | | | | | |
Collapse
|
29
|
Kobayashi M, Kim JY, Camarena V, Roehm PC, Chao MV, Wilson AC, Mohr I. A primary neuron culture system for the study of herpes simplex virus latency and reactivation. J Vis Exp 2012:3823. [PMID: 22491318 DOI: 10.3791/3823] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Herpes simplex virus type-1 (HSV-1) establishes a life-long latent infection in peripheral neurons. This latent reservoir is the source of recurrent reactivation events that ensure transmission and contribute to clinical disease. Current antivirals do not impact the latent reservoir and there are no vaccines. While the molecular details of lytic replication are well-characterized, mechanisms controlling latency in neurons remain elusive. Our present understanding of latency is derived from in vivo studies using small animal models, which have been indispensable for defining viral gene requirements and the role of immune responses. However, it is impossible to distinguish specific effects on the virus-neuron relationship from more general consequences of infection mediated by immune or non-neuronal support cells in live animals. In addition, animal experimentation is costly, time-consuming, and limited in terms of available options for manipulating host processes. To overcome these limitations, a neuron-only system is desperately needed that reproduces the in vivo characteristics of latency and reactivation but offers the benefits of tissue culture in terms of homogeneity and accessibility. Here we present an in vitro model utilizing cultured primary sympathetic neurons from rat superior cervical ganglia (SCG) (Figure 1) to study HSV-1 latency and reactivation that fits most if not all of the desired criteria. After eliminating non-neuronal cells, near-homogeneous TrkA(+) neuron cultures are infected with HSV-1 in the presence of acyclovir (ACV) to suppress lytic replication. Following ACV removal, non-productive HSV-1 infections that faithfully exhibit accepted hallmarks of latency are efficiently established. Notably, lytic mRNAs, proteins, and infectious virus become undetectable, even in the absence of selection, but latency-associated transcript (LAT) expression persists in neuronal nuclei. Viral genomes are maintained at an average copy number of 25 per neuron and can be induced to productively replicate by interfering with PI3-Kinase / Akt signaling or the simple withdrawal of nerve growth factor(1). A recombinant HSV-1 encoding EGFP fused to the viral lytic protein Us11 provides a functional, real-time marker for replication resulting from reactivation that is readily quantified. In addition to chemical treatments, genetic methodologies such as RNA-interference or gene delivery via lentiviral vectors can be successfully applied to the system permitting mechanistic studies that are very difficult, if not impossible, in animals. In summary, the SCG-based HSV-1 latency / reactivation system provides a powerful, necessary tool to unravel the molecular mechanisms controlling HSV1 latency and reactivation in neurons, a long standing puzzle in virology whose solution may offer fresh insights into developing new therapies that target the latent herpesvirus reservoir.
Collapse
Affiliation(s)
- Mariko Kobayashi
- Department of Microbiology, New York University School of Medicine, NY, USA
| | | | | | | | | | | | | |
Collapse
|
30
|
Howard AJ, Sherman DM, Visalli MA, Burnside DM, Visalli RJ. The Varicella-zoster virus ORF54 gene product encodes the capsid portal protein, pORF54. Virus Res 2012; 167:102-5. [PMID: 22475744 DOI: 10.1016/j.virusres.2012.03.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2011] [Revised: 03/07/2012] [Accepted: 03/19/2012] [Indexed: 10/28/2022]
Abstract
The Varicella-zoster virus (VZV) ORF54 gene was characterized using a guinea pig antiserum prepared to a GST-pORF54 fusion protein. A protein of the predicted size, 87kDa, was detected in VZV-infected MeWo cells but not in mock-infected cells. Sucrose density gradient fractionation of pORF54 expressed in a recombinant baculovirus system resulted in samples containing enriched amounts of pORF54. Electron microscopic analysis suggested that the ORF54 gene encodes a protein that assembles into ring-like portal structures similar to those observed for numerous bacteriophages and other herpesviruses.
Collapse
Affiliation(s)
- Alexander J Howard
- Department of Biology, Indiana University-Purdue University Fort Wayne, Fort Wayne, IN 46805, United States
| | | | | | | | | |
Collapse
|
31
|
Baines JD. Herpes simplex virus capsid assembly and DNA packaging: a present and future antiviral drug target. Trends Microbiol 2011; 19:606-13. [DOI: 10.1016/j.tim.2011.09.001] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2011] [Revised: 08/30/2011] [Accepted: 09/13/2011] [Indexed: 10/16/2022]
|
32
|
Camarena V, Kobayashi M, Kim JY, Roehm P, Perez R, Gardner J, Wilson AC, Mohr I, Chao MV. Nature and duration of growth factor signaling through receptor tyrosine kinases regulates HSV-1 latency in neurons. Cell Host Microbe 2011; 8:320-30. [PMID: 20951966 DOI: 10.1016/j.chom.2010.09.007] [Citation(s) in RCA: 129] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2010] [Revised: 07/10/2010] [Accepted: 08/20/2010] [Indexed: 11/16/2022]
Abstract
Herpes simplex virus-1 (HSV-1) establishes life-long latency in peripheral neurons where productive replication is suppressed. While periodic reactivation results in virus production, the molecular basis of neuronal latency remains incompletely understood. Using a primary neuronal culture system of HSV-1 latency and reactivation, we show that continuous signaling through the phosphatidylinositol 3-kinase (PI3-K) pathway triggered by nerve growth factor (NGF)-binding to the TrkA receptor tyrosine kinase (RTK) is instrumental in maintaining latent HSV-1. The PI3-K p110α catalytic subunit, but not the β or δ isoforms, is specifically required to activate 3-phosphoinositide-dependent protein kinase-1 (PDK1) and sustain latency. Disrupting this pathway leads to virus reactivation. EGF and GDNF, two other growth factors capable of activating PI3-K and PDK1 but that differ from NGF in their ability to persistently activate Akt, do not fully support HSV-1 latency. Thus, the nature of RTK signaling is a critical host parameter that regulates the HSV-1 latent-lytic switch.
Collapse
Affiliation(s)
- Vladimir Camarena
- Molecular Neurobiology Program, Skirball Institute for Biomolecular Medicine, New York University School of Medicine, New York, NY 10016, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
A 128-base-pair sequence containing the pac1 and a presumed cryptic pac2 sequence includes cis elements sufficient to mediate efficient genome maturation of human cytomegalovirus. J Virol 2011; 85:4432-9. [PMID: 21345955 DOI: 10.1128/jvi.02307-10] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Herpesvirus DNA replication proceeds via concatemeric replicative intermediates that are comprised of head-to-tail linked genomes. Genome maturation is carried out by the terminase, an enzyme complex that mediates both the insertion of concatemer DNA into capsids and its subsequent cleavage to release genomes within these capsids. This cleavage is sequence specific, but the governing cis-acting DNA sequences are only partially characterized. Two highly conserved motifs, the pac1 and pac2 motifs, lie near the ends of herpesvirus genomes and are known to be critical for genome maturation. In murine cytomegalovirus, poorly conserved sequences distal to the pac2 motif up to 150 bp from the point of cleavage are also important for cleavage. Here, we sought to identify the cleavage/packaging signals of human cytomegalovirus. Our results show that a previously proposed pac2-like poly(A) tract is dispensable for cleavage/packaging function and suggest that human cytomegalovirus may utilize a cryptic pac2 motif that lacks a poly(A) tract characteristic of pac2 motifs in other herpesviruses. Additional distal sequences 47 to 100 bp from the point of cleavage were found to enhance cleavage efficiency. These results should facilitate the identification of trans-acting factors that bind to these cis elements and elucidation of their functions. Such information will be critical for understanding the molecular basis of this complex process.
Collapse
|
34
|
Visalli RJ, Knepper J, Goshorn B, Vanover K, Burnside DM, Irven K, McGauley R, Visalli M. Characterization of the Varicella-zoster virus ORF25 gene product: pORF25 interacts with multiple DNA encapsidation proteins. Virus Res 2009; 144:58-64. [PMID: 19720242 DOI: 10.1016/j.virusres.2009.03.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2008] [Revised: 03/29/2009] [Accepted: 03/30/2009] [Indexed: 11/18/2022]
Abstract
The Herpesviridae contain a group of highly conserved proteins designated the Herpes UL33 Superfamily (pfam03581). The Varicella-zoster virus (VZV) homolog, encoded by the ORF25 gene, was used to generate a GST-ORF25 fusion protein. Purified GST-ORF25 was used to generate a polyclonal rabbit antiserum that detected the 17.5 kDa ORF25 protein (pORF25) in VZV infected cells. In pull-down assays, GST-ORF25 interacted with a number of encapsidation proteins including ORF30, ORF42 (the second exon of ORF45/42) and itself. The self-interaction was confirmed via a yeast two-hybrid assay. Additionally, pORF25 and pORF30 were shown to co-immunoprecipitate from VZV infected cells. Our results suggest that pORF25 is part of the trimeric terminase complex for VZV. However, combined with data from previous studies on HSV-1 and Kaposi's sarcoma associated herpesvirus (KSVH), we hypothesize that VZV pORF25 and the Herpes UL33 Superfamily homologs are not encapsidation proteins per se but instead work to bring viral proteins together to form functional complexes.
Collapse
Affiliation(s)
- Robert J Visalli
- Department of Biology, Indiana University Purdue University Fort Wayne, 2101 E. Coliseum Blvd., Fort Wayne, IN 46805, USA.
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Abstract
The murine cytomegalovirus (MCMV) M56 is one of three proteins that combine to form the MCMV terminase, required for cleavage and packaging of viral DNA into capsids. Deletion of M56 from a bacterial artificial chromosome (BAC) clone of the MCMV genome was considered lethal, as the mutant BAC failed to reconstitute infectious virus. Reintroduction of M56 at an ectopic locus complemented the deletion, allowing reconstitution of a virus that replicated with wild-type efficiency. However, neither the reintroduction of M56 sequences encoding an N-terminal epitope fusion nor a mutation targeting a region in M56 implicated as an ATPase active site was capable of restoring virus viability. In contrast, a frame shift mutation in M56a, a putative open reading frame that overlaps M56, had no effect on viral replication. We conclude that M56a is dispensable, whereas M56 residues comprising the proposed ATPase active site are critical for terminase function and viral replication.
Collapse
Affiliation(s)
- Jian Ben Wang
- Department of Pediatrics, Medical College of Virginia campus of Virginia Commonwealth University, 1101 E. Marshall Street, Richmond, Virginia 23298-0163, USA
| | - Michael A McVoy
- Department of Pediatrics, Medical College of Virginia campus of Virginia Commonwealth University, 1101 E. Marshall Street, Richmond, Virginia 23298-0163, USA
| |
Collapse
|
36
|
Visalli RJ, Nicolosi DM, Irven KL, Goshorn B, Khan T, Visalli MA. The Varicella-zoster virus DNA encapsidation genes: Identification and characterization of the putative terminase subunits. Virus Res 2007; 129:200-11. [PMID: 17868947 PMCID: PMC2669082 DOI: 10.1016/j.virusres.2007.07.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2007] [Revised: 07/24/2007] [Accepted: 07/24/2007] [Indexed: 11/23/2022]
Abstract
The putative DNA encapsidation genes encoded by open reading frames (ORFs) 25, 26, 30, 34, 43, 45/42 and 54 were cloned from Varicella-zoster virus (VZV) strain Ellen. Sequencing revealed that the Ellen ORFs were highly conserved at the amino acid level when compared to those of 19 previously published VZV isolates. Additionally, RT-PCR provided the first evidence that ORF45/42 was expressed as a spliced transcript in VZV-infected cells. All seven ORFs were expressed in vitro and full length products were identified using a C-terminal V5 epitope tag. The in vitro products of the putative VZV terminase subunits encoded by ORFs 30 and 45/42 proved useful in protein-protein interaction assays. Previous studies have reported the formation of a heterodimeric terminase complex involved in DNA encapsidation for both herpes simplex virus-type 1 (HSV-1) and human cytomegalovirus (HCMV). Here we report that the C-terminal portion of exon II of ORF45/42 (ORF42-C269) interacted in GST-pull down experiments with in vitro synthesized ORF30 and ORF45/42. The interactions were maintained in the presence of anionic detergents and in buffers of increasing ionic strength. Cells transiently transfected with epitope tagged ORF45/42 or ORF30 showed primarily cytoplasmic staining. In contrast, an antiserum directed to the N-terminal portion of ORF45 showed nearly exclusive nuclear localization of the ORF45/42 gene product in infected cells. An ORF30 specific antiserum detected an 87 kDa protein in both the cytoplasmic and nuclear fractions of VZV infected cells. The results were consistent with the localization and function of herpesviral terminase subunits. This is the first study aimed at the identification and characterization of the VZV DNA encapsidation gene products.
Collapse
Affiliation(s)
- Robert J Visalli
- Department of Biology, Indiana University Purdue University Fort Wayne, 2101 E. Coliseum Blvd., Fort Wayne, IN 46805-1499, USA.
| | | | | | | | | | | |
Collapse
|
37
|
Trujillo C, Mó O, Yáñez M, Salpin JY, Tortajada J. Gas-Phase Reactions Between Thiourea and Ca2+: New Evidence for the Formation of [Ca(NH3)]2+ and Other Doubly Charged Species. Chemphyschem 2007; 8:1330-7. [PMID: 17492701 DOI: 10.1002/cphc.200700113] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The gas-phase reactions between Ca(2+) and thiourea are investigated by means of electrospray ionization/mass spectrometry experiments. The MS/MS spectra of [Ca(thiourea)](2+) complexes show the appearance of new doubly charged species formed by the loss of NH(3) and HNCS. Other intense peaks at m/z 43, 56, 60, 73, 76 and 98 are also observed, and assigned to monocations produced in different coulomb-explosion processes. The structures and bonding characteristics of the different stationary points of the [Ca(thiourea)](2+) potential energy surface (PES) were theoretically studied by DFT calculations carried out at B3LYP/cc-pWCVTZ level. The analysis of the topology of this PES permits to propose different mechanisms for the loss of ammonia and HNCS, and to identify, the m/z 43, 56, 60, 73, 76 and 98 peaks as H(2)NCNH(+), CaNH(2) (+), H(2)NCS(+), CaSH(+), thiourea(+) and CaNCS(+) ions respectively. There are significant dissimilarities between the reactivity of urea and thiourea, which are related to the lower ionization energy of the latter, and to the fact that thioenols are intrinsically more stable than enols with respect to the corresponding keto forms.
Collapse
Affiliation(s)
- Cristina Trujillo
- Departamento de Química, Universidad Autónoma de Madrid, Cantoblanco, 28049 Madrid, Spain
| | | | | | | | | |
Collapse
|
38
|
Dodson AW, Taylor TJ, Knipe DM, Coen DM. Inhibitors of the sodium potassium ATPase that impair herpes simplex virus replication identified via a chemical screening approach. Virology 2007; 366:340-8. [PMID: 17544048 PMCID: PMC2099250 DOI: 10.1016/j.virol.2007.05.001] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2007] [Revised: 03/27/2007] [Accepted: 05/01/2007] [Indexed: 01/07/2023]
Abstract
Small molecules can provide valuable tools to investigate virus biology. We developed a chemical screening approach to identify small molecule inhibitors of poorly understood, pre-early gene expression steps in herpes simplex virus infection, using green fluorescent protein fused to an early protein. Our assay identified ouabain, a cardiac glycoside. Ouabain reversibly decreased viral yield by 100-fold without affecting cellular metabolic activity in an overnight assay. The antiviral potencies of other cardiac glycosides correlated with their potencies against the known target of these compounds, the cellular sodium potassium ATPase. Ouabain had a reduced effect if added 8 h post-infection. It did not inhibit viral attachment or entry, but did reduce the expression of viral immediate-early and early genes by at least 5-fold. Collectively, these results implicate a cellular target that was hitherto not considered important for a stage of HSV replication prior to viral gene expression.
Collapse
Affiliation(s)
- Allen W. Dodson
- Department of Biological Chemistry and Molecular Pharmacology
| | - Travis J Taylor
- Department of Microbiology and Molecular Genetics
- Committee on Virology, Harvard Medical School Boston, MA 02115, USA
| | - David M. Knipe
- Department of Microbiology and Molecular Genetics
- Committee on Virology, Harvard Medical School Boston, MA 02115, USA
| | - Donald M. Coen
- Department of Biological Chemistry and Molecular Pharmacology
- Committee on Virology, Harvard Medical School Boston, MA 02115, USA
- To Whom Correspondence should be addressed: Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, 250 Longwood Avenue, Boston, MA, 02215. Phone: +1 617 432-1691. Fax: +1 617 432-3833,
| |
Collapse
|
39
|
Pesola JM, Coen DM. In vivo fitness and virulence of a drug-resistant herpes simplex virus 1 mutant. J Gen Virol 2007; 88:1410-1414. [PMID: 17412967 DOI: 10.1099/vir.0.82787-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Two important issues regarding a virus mutant that is resistant to an antiviral drug are its ability to replicate in animal hosts (in vivo fitness) relative to other genetic variants, including wild type, and its ability to cause disease. These issues have been investigated for a herpes simplex virus 1 mutant that is resistant to thiourea compounds, which inhibit encapsidation of viral DNA. Following corneal inoculation of mice, the mutant virus replicated very similarly to its wild-type parent in the eye, trigeminal ganglion and brain. The mutant virus was as lethal to mice as its wild-type parent following this route of inoculation. Indeed, it exhibited increased virulence. Thus, unlike most drug-resistant virus mutants, this mutant retained in vivo fitness and virulence.
Collapse
Affiliation(s)
- Jean M Pesola
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, 250 Longwood Avenue, Boston, MA 02115, USA
| | - Donald M Coen
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, 250 Longwood Avenue, Boston, MA 02115, USA
| |
Collapse
|
40
|
Yang K, Homa F, Baines JD. Putative terminase subunits of herpes simplex virus 1 form a complex in the cytoplasm and interact with portal protein in the nucleus. J Virol 2007; 81:6419-33. [PMID: 17392365 PMCID: PMC1900116 DOI: 10.1128/jvi.00047-07] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Herpes simplex virus (HSV) terminase is an essential component of the molecular motor that translocates DNA through the portal vertex in the capsid during DNA packaging. The HSV terminase is believed to consist of the UL15, UL28, and UL33 gene products (pUL15, pUL28, and pUL33, respectively), whereas the HSV type 1 portal vertex is encoded by UL6. Immunoprecipitation reactions revealed that pUL15, pUL28, and pUL33 interact in cytoplasmic and nuclear lysates. Deletion of a canonical nuclear localization signal (NLS) from pUL15 generated a dominant-negative protein that, when expressed in an engineered cell line, decreased the replication of wild-type virus up to 80-fold. When engineered into the genome of recombinant HSV, this mutation did not interfere with the coimmunoprecipitation of pUL15, pUL28, and pUL33 from cytoplasmic lysates of infected cells but prevented viral replication, most nuclear import of both pUL15 and pUL28, and coimmunoprecipitation of pUL15, pUL28, and pUL33 from nuclear lysates. When the pUL15/pUL28 interaction was reduced in infected cells by the truncation of the C terminus of pUL28, pUL28 remained in the cytoplasm. Whether putative terminase components localized in the nucleus or cytoplasm, pUL6 localized in infected cell nuclei, as viewed by indirect immunofluorescence. The finding that the portal and terminase do eventually interact was supported by the observation that pUL6 coimmunoprecipitated strongly with pUL15 and weakly with pUL28 from extracts of infected cells in 1.0 M NaCl. These data are consistent with the hypothesis that the pUL15/pUL28/pUL33 complex forms in the cytoplasm and that an NLS in pUL15 is used to import the complex into the nucleus where at least pUL15 and pUL28 interact with the portal to mediate DNA packaging.
Collapse
Affiliation(s)
- Kui Yang
- Department of Microbiology and Immunology, Cornell University, New York School of Veterinary Medicine, Ithaca, NY 14853, USA
| | | | | |
Collapse
|
41
|
Kleymann G. Agents and strategies in development for improved management of herpes simplex virus infection and disease. Expert Opin Investig Drugs 2006; 14:135-61. [PMID: 15757392 DOI: 10.1517/13543784.14.2.135] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The quiet pandemic of herpes simplex virus (HSV) infections has plagued humanity since ancient times, causing mucocutaneous infection such as herpes labialis and herpes genitalis. Disease symptoms often interfere with every-day activities and occasionally HSV infections are the cause of life-threatening or sight-impairing disease, especially in neonates and the immuno-compromised patient population. After infection the virus persists for life in neurons of the host in a latent form, periodically reactivating and often resulting in significant psychosocial distress for the patient. Currently no cure is available. So far, vaccines, ILs, IFNs, therapeutic proteins, antibodies, immunomodulators and small-molecule drugs with specific or non-specific modes of action lacked either efficacy or the required safety profile to replace the nucleosidic drugs acyclovir, valacyclovir, penciclovir and famciclovir as the first choice of treatment. The recently discovered inhibitors of the HSV helicase-primase are the most potent development candidates today. These antiviral agents act by a novel mechanism of action and display low resistance rates in vitro and superior efficacy in animal models. This review summarises the current therapeutic options, discusses the potential of preclinical or investigational drugs and provides an up-to-date interpretation of the challenge to establish novel treatments for herpes simplex disease.
Collapse
|
42
|
Pesola JM, Zhu J, Knipe DM, Coen DM. Herpes simplex virus 1 immediate-early and early gene expression during reactivation from latency under conditions that prevent infectious virus production. J Virol 2005; 79:14516-25. [PMID: 16282451 PMCID: PMC1287588 DOI: 10.1128/jvi.79.23.14516-14525.2005] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2005] [Accepted: 09/12/2005] [Indexed: 01/20/2023] Open
Abstract
The program of gene expression exhibited by herpes simplex virus during productive infection of cultured cells is well established; however, less is known about the regulatory controls governing reactivation from latency in neurons. One difficulty in examining gene regulation during reactivation lies in distinguishing between events occurring in initial reactivating cells versus events occurring in secondarily infected cells. Thus, two inhibitors were employed to block production of infectious virus: acyclovir, which inhibits viral DNA synthesis, and WAY-150138, which permits viral DNA synthesis but inhibits viral DNA encapsidation. Latently infected murine ganglia were explanted in the presence of either inhibitor, and then amounts of RNA, DNA, or infectious virus were quantified. In ganglia explanted for 48 h, the levels of five immediate-early and early RNAs did not exhibit meaningful differences between acyclovir and WAY-150138 treatments when analyzed by in situ hybridization or quantitative reverse transcription-PCR. However, comparative increases in viral DNA and RNA content in untreated ganglia suggested that virus was produced before 48 h postexplant. This was confirmed by the detection of infectious virus as early as 14 h postexplant. Together, these results suggest that high levels of viral gene expression at 48 h postexplant are due largely to the production of infectious virus and subsequent spread through the tissue. These results lead to a reinterpretation of previous results indicating a role for DNA replication in immediate-early and early viral gene expression; however, it remains possible that viral gene expression is regulated differently in neurons than in cultured cells.
Collapse
Affiliation(s)
- Jean M Pesola
- Department of Biological Chemistry, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | |
Collapse
|
43
|
Abstract
The long-term treatment of herpesvirus infections with current antivirals leads to the development of drug-resistant viruses. Because currently available antivirals finally target the viral DNA polymerase, mutant resistant to one drug often shows cross-resistance to other drugs. This evidence highlights the need for the development of new antivirals that have the different viral targets. Recently, high-through-put screening of large compound collections for inhibiting specific viral enzymes, or in vitro cell culture assay, has identified several new antivirals. These include the inhibitors of helicase/primase complex, terminase complex, portal protein and UL97 protein kinase. This review will focus on these new compounds that directly inhibit viral replication.
Collapse
Affiliation(s)
- Yoshito Eizuru
- Center for Chronic Viral Diseases, Graduate School of Medical and Dental Sciences, Kagoshima University, Japan.
| |
Collapse
|
44
|
Di Grandi MJ, Curran KJ, Feigelson G, Prashad A, Ross AA, Visalli R, Fairhurst J, Feld B, Bloom JD. Thiourea inhibitors of herpesviruses. Part 3: Inhibitors of varicella zoster virus. Bioorg Med Chem Lett 2005; 14:4157-60. [PMID: 15261261 DOI: 10.1016/j.bmcl.2004.06.025] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2004] [Revised: 06/09/2004] [Accepted: 06/09/2004] [Indexed: 11/20/2022]
Abstract
The preparation of alpha-methylbenzyl thioureas and their biological activity against varicella zoster virus is described. Several analogs demonstrated IC50s<0.1 microM and their SAR are discussed. These compounds represent a novel class of potent and selective nonnucleoside inhibitors of varicella zoster virus.
Collapse
|
45
|
Evers DL, Wang X, Huong SM, Andreoni KA, Huang ES. Inhibition of human cytomegalovirus signaling and replication by the immunosuppressant FK778. Antiviral Res 2005; 65:1-12. [PMID: 15652966 DOI: 10.1016/j.antiviral.2004.03.007] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2003] [Accepted: 03/31/2004] [Indexed: 11/27/2022]
Abstract
FK778 (Fujisawa Healthcare Inc.) is an immunosuppressant structurally similar to A771726, the active metabolite of leflunomide (Aventis Pharmaceuticals), but with a clinically relevant shorter serum half-life. Leflunomide, a tolerated and efficacious immunosuppressive agent in patients receiving allograft transplantations, was reported to be active against HCMV and HSV-1. Here we report that FK778 is a potent and effective inhibitor of HCMV, and that its mode of antiviral action appears to mirror the biochemical mechanisms elsewhere described to be responsible for its immunosuppressive properties: inhibition of protein tyrosine phosphorylation and inhibition of cellular de novo pyrimidine biosynthesis. Initial HCMV-mediated activation of the EGF receptor/phosphatidylinositol 3-kinase (PI3-K) pathways and Sp1 and NF-kappaB were partially inhibited by FK778. The second tier (phase) of PI3-K, Sp1, and NF-kappaB induction by HCMV was more sensitive to FK778. Treatment of HCMV-infected cells with FK778 prevented the appearance of HCMV proteins some 12-24h post infection, and inhibited viral DNA synthesis. In our assays, leflunomide also reduced HCMV DNA levels. The antiviral activity of FK778 was reversed in cell culture by treatment with uridine, consistent with specific inhibition of dihydroorotate dehydrogenase (DHODH), a required enzyme in the de novo biosynthesis of pyrimidines. This report substantiates the clinical possibility of a single drug treatment to achieve immunosuppression and inhibit opportunistic herpesvirus infections. Our results differ from descriptions of leflunomide acting as an inhibitor of HCMV cytoplasmic capsid formation. Additionally, this study indicates that DHODH may be an effective cellular antiviral target.
Collapse
Affiliation(s)
- David L Evers
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | | | | | | | | |
Collapse
|
46
|
Evers DL, Komazin G, Ptak RG, Shin D, Emmer BT, Townsend LB, Drach JC. Inhibition of human cytomegalovirus replication by benzimidazole nucleosides involves three distinct mechanisms. Antimicrob Agents Chemother 2004; 48:3918-27. [PMID: 15388453 PMCID: PMC521925 DOI: 10.1128/aac.48.10.3918-3927.2004] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The benzimidazole nucleosides 2-bromo-5,6-dichloro-1-(beta-d-ribofuranosyl)benzimidazole (BDCRB) and 2-isopropylamino-5,6-dichloro-1-(beta-l-ribofuranosyl)benzimidazole (1263W94, or maribavir) are potent and selective inhibitors of human cytomegalovirus (HCMV) replication. These inhibitors act by two different mechanisms: BDCRB blocks the processing and maturation of viral DNA, whereas maribavir prevents viral DNA synthesis and capsid nuclear egress. In order to determine by which of these two mechanisms other benzimidazole nucleosides acted, we performed time-of-addition studies and other experiments with selected new analogs. We found that the erythrofuranosyl analog and the alpha-lyxofuranosyl analog acted late in the viral replication cycle, similar to BDCRB. In marked contrast, the alpha-5'-deoxylyxofuranosyl analog of 2,5,6-trichloro-1-(beta-d-ribofuranosyl)benzimidazole (compound UMJD1311) acted early in the replication cycle, too early to be consistent with either mechanism. Similar to other reports on early acting inhibitors of herpesviruses, compound 1311 was multiplicity of infection dependent, an observation that could not be reproduced with UV-inactivated virus. HCMV isolates resistant to BDCRB and maribavir were sensitive to compound 1311, as were viruses resistant to ganciclovir, cidofovir, and foscarnet. The preincubation of host cells with compound 1311 and removal prior to the addition of HCMV did not produce an antiviral cellular response. We conclude that this newly discovered early mode of action occurs at a stage of viral replication after entry to cells but prior to viral DNA synthesis, thereby strongly suggesting that the trisubstituted benzimidazole nucleoside series possesses three distinct biochemical modes of action for inhibition of HCMV replication.
Collapse
Affiliation(s)
- David L Evers
- School of Dentistry, 1011 N. University Ave., University of Michigan, Ann Arbor, MI 48109-1078, USA
| | | | | | | | | | | | | |
Collapse
|
47
|
Abstract
The long-term treatment of herpesvirus infections with current antivirals in immunocompromised hosts leads to the development of drug-resistant viruses. Because nearly all currently available antivirals finally target viral DNA polymerase, virus resistant to one drug often shows cross-resistance to other drugs. In addition, nearly all the antivirals show various kinds of side effects or poor bioavailability. This evidence highlights the need for developing new antivirals for herpesviruses that have the different viral targets. Recently, high-throughput screening of large compound collections for inhibiting specific viral enzymes, or in vitro cell culture assay, has identified several new antivirals that target different viral proteins. These include the inhibitors of helicase/primase complex, terminase complex, portal protein and UL97 protein kinase. In addition, non-nucleoside inhibitors for viral DNA polymerase have been also developed. This review will focus on these new compounds that directly inhibit viral replication.
Collapse
Affiliation(s)
- Yoshito Eizuru
- Division of Persistent & Oncogenic Viruses, Centre for Chronic Viral Diseases, Faculty of Medicine, Kagoshima University, Kagoshima, Japan.
| |
Collapse
|
48
|
Visalli RJ. Novel compounds for the treatment of varicella–zoster virus infections. Expert Opin Ther Pat 2004. [DOI: 10.1517/13543776.14.3.355] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
49
|
Jones TR, Lee SW, Johann SV, Razinkov V, Visalli RJ, Feld B, Bloom JD, O'Connell J. Specific inhibition of human cytomegalovirus glycoprotein B-mediated fusion by a novel thiourea small molecule. J Virol 2004; 78:1289-300. [PMID: 14722284 PMCID: PMC321382 DOI: 10.1128/jvi.78.3.1289-1300.2004] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
A novel small molecule inhibitor of human cytomegalovirus (HCMV) was identified as the result of screening a chemical library by using a whole-virus infected-cell assay. Synthetic chemistry efforts yielded the analog designated CFI02, a compound whose potency had been increased about 100-fold over an initial inhibitor. The inhibitory concentration of CFI02 in various assays is in the low nanomolar range. CFI02 is a selective and potent inhibitor of HCMV; it has no activity against other CMVs, alphaherpesviruses, or unrelated viruses. Mechanism-of-action studies indicate that CFI02 acts very early in the replication cycle, inhibiting virion envelope fusion with the cell plasma membrane. Mutants resistant to CFI02 have mutations in the abundant virion envelope glycoprotein B that are sufficient to confer resistance. Taken together, the data suggest that CFI02 inhibits glycoprotein B-mediated HCMV virion fusion. Furthermore, CFI02 inhibits the cell-cell spread of HCMV. This is the first study of a potent and selective small molecule inhibitor of CMV fusion and cell-cell spread.
Collapse
Affiliation(s)
- Thomas R Jones
- Infectious Disease Section, Wyeth Research, Pearl River, New York 10965, USA.
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Komazin G, Townsend LB, Drach JC. Role of a mutation in human cytomegalovirus gene UL104 in resistance to benzimidazole ribonucleosides. J Virol 2004; 78:710-5. [PMID: 14694102 PMCID: PMC368810 DOI: 10.1128/jvi.78.2.710-715.2004] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The benzimidazole D-ribonucleosides TCRB and BDCRB are potent and selective inhibitors of human cytomegalovirus (HCMV) replication. Two HCMV strains resistant to these compounds were selected and had resistance mutations in genes UL89 and UL56. Proteins encoded by these two genes are the two subunits of the HCMV "terminase" and are necessary for cleavage and packaging of viral genomic DNA, a process inhibited by TCRB and BDCRB. We now report that both strains also have a previously unidentified mutation in UL104, the HCMV portal protein. This mutation, which results in L21F substitution, was introduced into the genome of wild-type HCMV by utilizing a recently cloned genome of HCMV as a bacterial artificial chromosome. The virus with this mutation alone was not resistant to BDCRB, suggesting that this site is not involved in binding benzimidazole nucleosides. As in previous proposals for mutations in UL104 of murine cytomegalovirus and HCMV strains resistant to BAY 38-4766, we hypothesize that this mutation could compensate for conformational changes in mutant UL89 and UL56 proteins, since the HCMV terminase is likely to interact with the portal protein during cleavage and packaging of genomic DNA.
Collapse
Affiliation(s)
- Gloria Komazin
- Department of Biologic and Materials Sciences, School of Dentistry, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109, USA
| | | | | |
Collapse
|