243 results on '"Knipe DM"'
Search Results
2. Local synthesis of C3 in lymphoid tissues is sufficient to restore humoral responses to peripheral herpes simplex virus (HSV-1) infection in C3-deficient mice
- Author
-
Verschoor, A, primary, Brockman, MA, additional, Knipe, DM, additional, and Carroll, MC, additional
- Published
- 2000
- Full Text
- View/download PDF
3. Comparison of the oncolytic activity of a replication-competent and a replication-deficient herpes simplex virus 1.
- Author
-
Lindner G, Walter A, Magnus CL, Rosenhammer K, Holoborodko B, Koch V, Hirsch S, Grossmann L, Li S, Knipe DM, DeLuca N, Schuler-Thurner B, Gross S, Schwertner B, Toelge M, Rohrhofer A, Stöckl S, Bauer RJ, Knoll G, Ehrenschwender M, Haferkamp S, Schmidt B, and Schuster P
- Subjects
- Humans, Cell Line, Tumor, Caspases metabolism, Animals, Melanoma therapy, Melanoma immunology, Herpesvirus 1, Human physiology, Virus Replication, Oncolytic Virotherapy methods, Oncolytic Viruses genetics, Oncolytic Viruses physiology, Apoptosis
- Abstract
In 2015, the oncolytic herpes simplex virus 1 (HSV-1) T-VEC (talimogene laherparepvec) was approved for intratumoral injection in non-resectable malignant melanoma. To determine whether viral replication is required for oncolytic activity, we compared replication-deficient HSV-1 d106S with replication-competent T-VEC. High infectious doses of HSV-1 d106S killed melanoma (n = 10), head-and-neck squamous cell carcinoma (n = 11), and chondrosarcoma cell lines (n = 2) significantly faster than T-VEC as measured by MTT metabolic activity, while low doses of T-VEC were more effective over time. HSV-1 d106S and, to a lesser extent T-VEC, triggered caspase-dependent early apoptosis as shown by pan-caspase inhibition and specific induction of caspases 3/7, 8, and 9. HSV-1 d106S induced a higher ratio of apoptosis-inducing infected cell protein (ICP) 0 to apoptosis-blocking ICP6 than T-VEC. T-VEC was oncolytic for an extended period of time as viral replication continued, which could be partially blocked by the antiviral drug aciclovir. High doses of T-VEC, but not HSV-1 d106S, increased interferon-β mRNA as part of the intrinsic immune response. When markers of immunogenic cell death were assessed, ATP was released more efficiently in the context of T-VEC than HSV-1 d106S infection, whereas HMGB1 was induced comparatively well. Overall, the early oncolytic effect on three different tumour entities was stronger with the non-replicative strain, while the replication-competent virus elicited a stronger innate immune response and more pronounced immunogenic cell death., (© 2024 The Authors. Immunology published by John Wiley & Sons Ltd.)
- Published
- 2024
- Full Text
- View/download PDF
4. Sp1 facilitates continued HSV-1 gene expression in the absence of key viral transactivators.
- Author
-
Sodroski CN, Oh HS, Chou S-F, and Knipe DM
- Subjects
- Humans, Trans-Activators genetics, Transcription Factors metabolism, Virus Replication, Gene Expression, Gene Expression Regulation, Viral, Herpesvirus 1, Human physiology, Immediate-Early Proteins genetics, Herpes Simplex, Herpesviridae Infections, Latent Infection
- Abstract
Productive replication of herpes simplex virus (HSV) relies upon a well-ordered transcriptional cascade flowing from immediate-early (IE) to early (E) to late (L) gene products. While several virus-encoded transcriptional activators are involved in this process, IE and E gene promoters also contain multiple binding sites for the ubiquitously expressed cellular transcription factor Sp1. Sp1 has been previously implicated in activating HSV-1 gene transcription downstream of these sites, but why Sp1-binding sites are maintained in the promoters of genes activated by virus-encoded activators remains unclear. We hypothesized that Sp1 enables continued HSV-1 transcription and replication when viral transactivators are limited. We used a depletion-based approach in human foreskin fibroblasts to investigate the specific contribution of Sp1 to the initiation and progression of the HSV-1 lytic gene cascade. We found that Sp1 increased viral transcript levels, protein expression, and replication following infection with VP16- or ICP0-deficient viruses but had little to no effect on rescued viruses or during wild-type (WT) HSV-1 infection. Moreover, Sp1 promoted WT virus transcription and replication following interferon treatment of fibroblasts and thus may contribute to viral immune evasion. Interestingly, we observed reduced expression of Sp1 and Sp1-family transcription factors in differentiated sensory neurons compared to undifferentiated cells, suggesting that reduced Sp1 levels may also contribute to HSV-1 latent infection. Overall, these findings indicate that Sp1 can promote HSV-1 gene expression in the absence of key viral transactivators; thus, HSV-1 may use Sp1 to maintain its gene expression and replication under adverse conditions.IMPORTANCEHerpes simplex virus (HSV) is a common human pathogen that actively replicates in the epithelia but can persist for the lifetime of the infected host via a stable, latent infection in neurons. A key feature of the HSV replication cycle is a complex transcriptional program in which virus and host-cell factors coordinate to regulate expression of the viral gene products necessary for continued viral replication. Multiple binding sites for the cellular transcription factor Sp1 are located in the promoters of HSV-1 genes, but how Sp1 binding contributes to transcription and replication of wild-type virus is not fully understood. In this study, we identified a specific role for Sp1 in maintaining HSV-1 gene transcription under adverse conditions, as when virus-encoded transcriptional activators were absent or limited. Preservation of Sp1-binding sites in HSV-1 gene promoters may thus benefit the virus as it navigates diverse cell types and host-cell conditions during infection., Competing Interests: The authors declare no conflict of interest.
- Published
- 2024
- Full Text
- View/download PDF
5. Neuronal miR-9 promotes HSV-1 epigenetic silencing and latency by repressing Oct-1 and Onecut family genes.
- Author
-
Deng Y, Lin Y, Chen S, Xiang Y, Chen H, Qi S, Oh HS, Das B, Komazin-Meredith G, Pesola JM, Knipe DM, Coen DM, and Pan D
- Subjects
- Humans, Male, Animals, Mice, Neurons, Transcription Factors, Epigenesis, Genetic, Homeodomain Proteins, Herpesvirus 1, Human genetics, Induced Pluripotent Stem Cells, MicroRNAs genetics, Herpes Simplex genetics
- Abstract
Herpes simplex virus 1 (HSV-1) latent infection entails repression of viral lytic genes in neurons. By functional screening using luciferase-expressing HSV-1, we identify ten neuron-specific microRNAs potentially repressing HSV-1 neuronal replication. Transfection of miR-9, the most active candidate from the screen, decreases HSV-1 replication and gene expression in Neuro-2a cells. Ectopic expression of miR-9 from lentivirus or recombinant HSV-1 suppresses HSV-1 replication in male primary mouse neurons in culture and mouse trigeminal ganglia in vivo, and reactivation from latency in the primary neurons. Target prediction and validation identify transcription factors Oct-1, a known co-activator of HSV transcription, and all three Onecut family members as miR-9 targets. Knockdown of ONECUT2 decreases HSV-1 yields in Neuro-2a cells. Overexpression of each ONECUT protein increases HSV-1 replication in Neuro-2a cells, human induced pluripotent stem cell-derived neurons, and primary mouse neurons, and accelerates reactivation from latency in the mouse neurons. Mutagenesis, ChIP-seq, RNA-seq, ChIP-qPCR and ATAC-seq results suggest that ONECUT2 can nonspecifically bind to viral genes via its CUT domain, globally stimulate viral gene transcription, reduce viral heterochromatin and enhance the accessibility of viral chromatin. Thus, neuronal miR-9 promotes viral epigenetic silencing and latency by targeting multiple host transcription factors important for lytic gene activation., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
6. Nuclear interferon-stimulated gene product maintains heterochromatin on the herpes simplex viral genome to limit lytic infection.
- Author
-
Sodroski CN and Knipe DM
- Subjects
- Humans, Heterochromatin genetics, Heterochromatin metabolism, Interferons genetics, Interferons metabolism, Phosphoproteins genetics, Phosphoproteins metabolism, Nuclear Proteins genetics, Nuclear Proteins metabolism, Ubiquitin-Protein Ligases genetics, Ubiquitin-Protein Ligases metabolism, Genome, Viral, Virus Replication genetics, Herpesvirus 1, Human physiology, Herpes Simplex
- Abstract
Interferons (IFN) are expressed in and secreted from cells in response to virus infection, and they induce the expression of a variety of genes called interferon-stimulated genes (ISGs) in infected and surrounding cells to block viral infection and limit spread. The mechanisms of action of a number of cytoplasmic ISGs have been well defined, but little is known about the mechanism of action of nuclear ISGs. Constitutive levels of nuclear interferon-inducible protein 16 (IFI16) serve to induce innate signaling and epigenetic silencing of herpes simplex virus (HSV), but only when the HSV infected cell protein 0 (ICP0) E3 ligase, which promotes IFI16 degradation, is inactivated. In this study, we found that following IFN induction, the pool of IFI16 within the infected cell remains high and can restrict wild-type viral gene expression and replication due to both the induced levels of IFI16 and the IFI16-mediated repression of ICP0 levels. Restriction of viral gene expression is achieved by IFI16 promoting the maintenance of heterochromatin on the viral genome, which silences it epigenetically. These results indicate that a nuclear ISG can restrict gene expression and replication of a nuclear DNA virus by maintaining or preventing the removal of repressive heterochromatin associated with the viral genome.
- Published
- 2023
- Full Text
- View/download PDF
7. Herpes simplex virus infected cell protein 8 is required for viral inhibition of the cGAS pathway.
- Author
-
Broekema N, Mertens ME, Angelova M, Orzalli MH, Oh HS, and Knipe DM
- Subjects
- Humans, Nucleotidyltransferases genetics, Nucleotidyltransferases metabolism, Virus Replication, DNA metabolism, Antiviral Agents pharmacology, Antiviral Agents metabolism, Herpesvirus 1, Human physiology, Herpes Simplex genetics
- Abstract
DNA virus infection triggers an antiviral type I interferon (IFN) response in cells that suppresses infection of surrounding cells. Consequently, viruses have evolved mechanisms to inhibit the IFN response for efficient replication. The cellular cGAS protein binds to double-stranded DNA and synthesizes the small molecule cGAMP to initiate DNA-dependent type I IFN production. We showed previously that cGAMP production is relatively low during HSV-1 infection compared to plasmid DNA transfection. Therefore, we hypothesized that HSV-1 produces antagonists of the cGAS DNA sensing pathway. In this study, we found that the HSV-1 ICP8 protein is required for viral inhibition of the cGAS pathway by reducing cGAMP levels stimulated by double-stranded DNA transfection. ICP8 alone inhibited the cGAMP response and may inhibit cGAS action by direct interaction with DNA, cGAS, or other infected cell proteins. Our results reveal another cGAS antiviral pathway inhibitor and highlight the importance of countering IFN for efficient viral replication., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2023. Published by Elsevier Inc.)
- Published
- 2023
- Full Text
- View/download PDF
8. Blockade of innate inflammatory cytokines TNF α , IL-1 β , or IL-6 overcomes virotherapy-induced cancer equilibrium to promote tumor regression.
- Author
-
Walsh MJ, Ali LR, Lenehan P, Kureshi CT, Kureshi R, Dougan M, Knipe DM, and Dougan SK
- Abstract
Cancer therapeutics can lead to immune equilibrium in which the immune response controls tumor cell expansion without fully eliminating the cancer. The factors involved in this equilibrium remain incompletely understood, especially those that would antagonize the anti-tumor immune response and lead to tumor outgrowth. We previously demonstrated that continuous treatment with a non-replicating herpes simplex virus 1 expressing interleukin (IL)-12 induces a state of cancer immune equilibrium highly dependent on interferon-γ. We profiled the IL-12 virotherapy-induced immune equilibrium in murine melanoma, identifying blockade of innate inflammatory cytokines, tumor necrosis factor alpha (TNFα), IL-1β, or IL-6 as possible synergistic interventions. Antibody depletions of each of these cytokines enhanced survival in mice treated with IL-12 virotherapy and helped to overcome equilibrium in some tumors. Single-cell RNA-sequencing demonstrated that blockade of inflammatory cytokines resulted in downregulation of overlapping inflammatory pathways in macrophages, shifting immune equilibrium towards tumor clearance, and raising the possibility that TNFα blockade could synergize with existing cancer immunotherapies., Competing Interests: M.D. has received research funding from Eli Lilly; he has received consulting fees from Genentech, ORIC Pharmaceuticals, Partner Therapeutics, SQZ Biotech, AzurRx, Eli Lilly, Mallinckrodt Pharmaceuticals, Aditum, Foghorn Therapeutics, Palleon, Sorriso Pharmaceuticals, Generate Biomedicines, and Moderna; he is a member of the Scientific Advisory Board for Neoleukin Therapeutics, Veravas and Cerberus Therapeutics. S.K.D. received research funding unrelated to this project from Eli Lilly, Novartis, and Bristol-Myers Squibb and is a founder, science advisory board member (SAB), and equity holder in Kojin. M.J.W., D.M.K., and S.K.D. together have a patent application “Immunotherapeutic Virus for Treatment of Cancer” pending for using d106S as an immunomodulatory agent. Other authors declare no other competing interests., (© The Author(s) 2023. Published by Oxford University Press on behalf of the British Society for Immunology.)
- Published
- 2023
- Full Text
- View/download PDF
9. IFNγ is a central node of cancer immune equilibrium.
- Author
-
Walsh MJ, Stump CT, Kureshi R, Lenehan P, Ali LR, Dougan M, Knipe DM, and Dougan SK
- Subjects
- Animals, Humans, Mice, CD8-Positive T-Lymphocytes, Interleukin-12 metabolism, Perforin, Interferon-gamma metabolism, Neoplasms
- Abstract
Tumors in immune equilibrium are held in balance between outgrowth and destruction by the immune system. The equilibrium phase defines the duration of clinical remission and stable disease, and escape from equilibrium remains a major clinical problem. Using a non-replicating HSV-1 vector expressing interleukin-12 (d106S-IL12), we developed a mouse model of therapy-induced immune equilibrium, a phenomenon previously seen only in humans. This immune equilibrium was centrally reliant on interferon-γ (IFNγ). CD8
+ T cell direct recognition of MHC class I, perforin/granzyme-mediated cytotoxicity, and extrinsic death receptor signaling such as Fas/FasL were all individually dispensable for equilibrium. IFNγ was critically important and played redundant roles in host and tumor cells such that IFNγ sensing in either compartment was sufficient for immune equilibrium. We propose that these redundant mechanisms of action are integrated by IFNγ to protect from oncogenic or chronic viral threats and establish IFNγ as a central node in therapy-induced immune equilibrium., Competing Interests: Declaration of interests M.D. receives research funding from Novartis and is on the Scientific Advisory Board for Neoleukin Therapeutics. S.K.D. receives research funding from Novartis, Bristol-Meyers Squibb, and Eli Lilly and is co-founder and on the Scientific Advisory Board of Kojin. M.J.W., D.M.K., and S.K.D. together have a patent application “Immunotherapeutic Virus for Treatment of Cancer” pending for using d106S as an immunomodulatory agent., (Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
10. Replication Compartments of Eukaryotic and Bacterial DNA Viruses: Common Themes Between Different Domains of Host Cells.
- Author
-
Knipe DM, Prichard A, Sharma S, and Pogliano J
- Subjects
- Bacteria genetics, Cell Nucleus, DNA Viruses genetics, DNA, Bacterial, DNA, Viral genetics, Eukaryota genetics, Eukaryotic Cells, Virus Replication, Bacteriophages genetics, Viruses genetics
- Abstract
Subcellular organization is essential for life. Cells organize their functions into organelles to concentrate their machinery and supplies for optimal efficiency. Likewise, viruses organize their replication machinery into compartments or factories within their host cells for optimal replicative efficiency. In this review, we discuss how DNA viruses that infect both eukaryotic cells and bacteria assemble replication compartments for synthesis of progeny viral DNA and transcription of the viral genome. Eukaryotic DNA viruses assemble replication compartments in the nucleus of the host cell while DNA bacteriophages assemble compartments called phage nuclei in the bacterial cytoplasm. Thus, DNA viruses infecting host cells from different domains of life share common replication strategies.
- Published
- 2022
- Full Text
- View/download PDF
11. The US3 Kinase of Herpes Simplex Virus Phosphorylates the RNA Sensor RIG-I To Suppress Innate Immunity.
- Author
-
van Gent M, Chiang JJ, Muppala S, Chiang C, Azab W, Kattenhorn L, Knipe DM, Osterrieder N, and Gack MU
- Subjects
- Alphaherpesvirinae genetics, Alphaherpesvirinae metabolism, Alphaherpesvirinae physiology, Amino Acid Sequence, DEAD Box Protein 58 chemistry, HEK293 Cells, Herpesvirus 1, Human genetics, Herpesvirus 1, Human metabolism, Humans, Immunity, Innate, Interferon Type I metabolism, Phosphorylation, Protein Binding, Protein Serine-Threonine Kinases genetics, Receptors, Immunologic chemistry, Viral Proteins genetics, DEAD Box Protein 58 metabolism, Herpesvirus 1, Human immunology, Immune Evasion, Protein Serine-Threonine Kinases metabolism, Receptors, Immunologic metabolism, Viral Proteins metabolism
- Abstract
Recent studies have demonstrated that the signaling activity of the cytosolic pathogen sensor retinoic acid-inducible gene-I (RIG-I) is modulated by a variety of posttranslational modifications (PTMs) to fine-tune the antiviral type I interferon (IFN) response. Whereas K63-linked ubiquitination of the RIG-I caspase activation and recruitment domains (CARDs) catalyzed by TRIM25 or other E3 ligases activates RIG-I, phosphorylation of RIG-I at S8 and T170 represses RIG-I signal transduction by preventing the TRIM25-RIG-I interaction and subsequent RIG-I ubiquitination. While strategies to suppress RIG-I signaling by interfering with its K63-polyubiquitin-dependent activation have been identified for several viruses, evasion mechanisms that directly promote RIG-I phosphorylation to escape antiviral immunity are unknown. Here, we show that the serine/threonine (Ser/Thr) kinase US3 of herpes simplex virus 1 (HSV-1) binds to RIG-I and phosphorylates RIG-I specifically at S8. US3-mediated phosphorylation suppressed TRIM25-mediated RIG-I ubiquitination, RIG-I-MAVS binding, and type I IFN induction. We constructed a mutant HSV-1 encoding a catalytically-inactive US3 protein (K220A) and found that, in contrast to the parental virus, the US3 mutant HSV-1 was unable to phosphorylate RIG-I at S8 and elicited higher levels of type I IFNs, IFN-stimulated genes (ISGs), and proinflammatory cytokines in a RIG-I-dependent manner. Finally, we show that this RIG-I evasion mechanism is conserved among the alphaherpesvirus US3 kinase family. Collectively, our study reveals a novel immune evasion mechanism of herpesviruses in which their US3 kinases phosphorylate the sensor RIG-I to keep it in the signaling-repressed state. IMPORTANCE Herpes simplex virus 1 (HSV-1) establishes lifelong latency in the majority of the human population worldwide. HSV-1 occasionally reactivates to produce infectious virus and to facilitate dissemination. While often remaining subclinical, both primary infection and reactivation occasionally cause debilitating eye diseases, which can lead to blindness, as well as life-threatening encephalitis and newborn infections. To identify new therapeutic targets for HSV-1-induced diseases, it is important to understand the HSV-1-host interactions that may influence infection outcome and disease. Our work uncovered direct phosphorylation of the pathogen sensor RIG-I by alphaherpesvirus-encoded kinases as a novel viral immune escape strategy and also underscores the importance of RNA sensors in surveilling DNA virus infection.
- Published
- 2022
- Full Text
- View/download PDF
12. A virus-specific monocyte inflammatory phenotype is induced by SARS-CoV-2 at the immune-epithelial interface.
- Author
-
Leon J, Michelson DA, Olejnik J, Chowdhary K, Oh HS, Hume AJ, Galván-Peña S, Zhu Y, Chen F, Vijaykumar B, Yang L, Crestani E, Yonker LM, Knipe DM, Mühlberger E, and Benoist C
- Subjects
- Adult, B-Lymphocytes immunology, COVID-19 pathology, Child, Coculture Techniques, Ebolavirus pathogenicity, Epithelial Cells virology, Gene Expression Profiling, Humans, Inflammation, Influenza A virus pathogenicity, Lung immunology, Myeloid Cells immunology, Species Specificity, Viral Proteins immunology, COVID-19 immunology, Epithelial Cells immunology, Monocytes immunology, SARS-CoV-2 pathogenicity
- Abstract
Infection by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) provokes a potentially fatal pneumonia with multiorgan failure, and high systemic inflammation. To gain mechanistic insight and ferret out the root of this immune dysregulation, we modeled, by in vitro coculture, the interactions between infected epithelial cells and immunocytes. A strong response was induced in monocytes and B cells, with a SARS-CoV-2-specific inflammatory gene cluster distinct from that seen in influenza A or Ebola virus-infected cocultures, and which reproduced deviations reported in blood or lung myeloid cells from COVID-19 patients. A substantial fraction of the effect could be reproduced after individual transfection of several SARS-CoV-2 proteins (Spike and some nonstructural proteins), mediated by soluble factors, but not via transcriptional induction. This response was greatly muted in monocytes from healthy children, perhaps a clue to the age dependency of COVID-19. These results suggest that the inflammatory malfunction in COVID-19 is rooted in the earliest perturbations that SARS-CoV-2 induces in epithelia., (Copyright © 2021 the Author(s). Published by PNAS.)
- Published
- 2022
- Full Text
- View/download PDF
13. CRISPR-Cas9 Expressed in Stably Transduced Cell Lines Promotes Recombination and Selects for Herpes Simplex Virus Recombinants.
- Author
-
Oh HS, Diaz FM, Zhou C, Carpenter N, and Knipe DM
- Abstract
Recombinant herpes simplex virus strains can be constructed by several methods, including homologous recombination, bacterial artificial chromosome manipulation, and yeast genetic methods. Homologous recombination may have the advantage of introducing fewer genetic alterations in the viral genome, but the low level of recombinants can make this method more time consuming if there is no screen or selection. In this study we used complementing cell lines that express Cas9 and guide RNAs targeting the parental virus to rapidly generate recombinant viruses. Analysis of the progeny viruses indicated that CRISPR-Cas9 both promoted recombination to increase recombinant viruses and selected against parental viruses in the transfection progeny viruses. This approach can also be used to enrich for recombinants made by any of the current methods.
- Published
- 2022
- Full Text
- View/download PDF
14. Tissue-Resident-Memory CD8 + T Cells Bridge Innate Immune Responses in Neighboring Epithelial Cells to Control Human Genital Herpes.
- Author
-
Peng T, Phasouk K, Sodroski CN, Sun S, Hwangbo Y, Layton ED, Jin L, Klock A, Diem K, Magaret AS, Jing L, Laing K, Li A, Huang ML, Mertens M, Johnston C, Jerome KR, Koelle DM, Wald A, Knipe DM, Corey L, and Zhu J
- Subjects
- Adaptive Immunity genetics, Adult, Aged, Antigens, Viral immunology, CD8-Positive T-Lymphocytes metabolism, CD8-Positive T-Lymphocytes virology, Cells, Cultured, Epithelial Cells metabolism, Epithelial Cells virology, Female, Gene Expression Profiling, Herpes Genitalis genetics, Herpes Genitalis metabolism, Herpes Genitalis virology, Herpesvirus 2, Human pathogenicity, Host-Pathogen Interactions, Humans, Interferon-gamma genetics, Interferon-gamma metabolism, Male, Memory T Cells metabolism, Memory T Cells virology, Middle Aged, Phenotype, Skin metabolism, Skin virology, Transcriptome, CD8-Positive T-Lymphocytes immunology, Epithelial Cells immunology, Herpes Genitalis immunology, Herpesvirus 2, Human immunology, Immunity, Innate genetics, Immunologic Memory, Memory T Cells immunology, Skin immunology
- Abstract
Tissue-resident-memory T cells (TRM) populate the body's barrier surfaces, functioning as frontline responders against reencountered pathogens. Understanding of the mechanisms by which CD8TRM achieve effective immune protection remains incomplete in a naturally recurring human disease. Using laser capture microdissection and transcriptional profiling, we investigate the impact of CD8TRM on the tissue microenvironment in skin biopsies sequentially obtained from a clinical cohort of diverse disease expression during herpes simplex virus 2 (HSV-2) reactivation. Epithelial cells neighboring CD8TRM display elevated and widespread innate and cell-intrinsic antiviral signature expression, largely related to IFNG expression. Detailed evaluation via T-cell receptor reconstruction confirms that CD8TRM recognize viral-infected cells at the specific HSV-2 peptide/HLA level. The hierarchical pattern of core IFN- γ signature expression is well-conserved in normal human skin across various anatomic sites, while elevation of IFI16, TRIM 22, IFITM2, IFITM3, MX1, MX2, STAT1, IRF7, ISG15, IFI44, CXCL10 and CCL5 expression is associated with HSV-2-affected asymptomatic tissue. In primary human cells, IFN- γ pretreatment reduces gene transcription at the immediate-early stage of virus lifecycle, enhances IFI16 restriction of wild-type HSV-2 replication and renders favorable kinetics for host protection. Thus, the adaptive immune response through antigen-specific recognition instructs innate and cell-intrinsic antiviral machinery to control herpes reactivation, a reversal of the canonical thinking of innate activating adaptive immunity in primary infection. Communication from CD8TRM to surrounding epithelial cells to activate broad innate resistance might be critical in restraining various viral diseases., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest. The reviewer has declared a shared affiliation with some of the authors CS, MM, DK to the handling Editor at the time of review., (Copyright © 2021 Peng, Phasouk, Sodroski, Sun, Hwangbo, Layton, Jin, Klock, Diem, Magaret, Jing, Laing, Li, Huang, Mertens, Johnston, Jerome, Koelle, Wald, Knipe, Corey and Zhu.)
- Published
- 2021
- Full Text
- View/download PDF
15. Vesicular Stomatitis Virus Chimeras Expressing the Oropouche Virus Glycoproteins Elicit Protective Immune Responses in Mice.
- Author
-
Stubbs SH, Cornejo Pontelli M, Mishra N, Zhou C, de Paula Souza J, Mendes Viana RM, Lipkin WI, Knipe DM, Arruda E, and Whelan SPJ
- Subjects
- Animals, Antibodies, Neutralizing, Bunyaviridae Infections immunology, Male, Mice, Mice, Inbred C57BL, Vesicular Stomatitis virology, Virus Replication, Bunyaviridae Infections prevention & control, Genome, Viral, Orthobunyavirus genetics, Vesiculovirus genetics, Vesiculovirus immunology, Viral Envelope Proteins genetics
- Abstract
Oropouche virus (OROV) infection of humans is associated with a debilitating febrile illness that can progress to meningitis or encephalitis. First isolated from a forest worker in Trinidad and Tobago in 1955, the arbovirus OROV has since been detected throughout the Amazon basin with an estimated 500,000 human infections over 60 years. Like other members of the family Peribunyaviridae , the viral genome exists as 3 single-stranded negative-sense RNA segments. The medium-sized segment encodes a viral glycoprotein complex (GPC) that is proteolytically processed into two viral envelope proteins, Gn and Gc, responsible for attachment and membrane fusion. There are no therapeutics or vaccines to combat OROV infection, and we have little understanding of protective immunity to infection. Here, we generated a replication competent chimeric vesicular stomatitis virus (VSV), in which the endogenous glycoprotein was replaced by the GPC of OROV. Serum from mice immunized by intramuscular injection with VSV-OROV specifically neutralized wild-type OROV, and using peptide arrays we mapped multiple epitopes within an N-terminal variable region of Gc recognized by the immune sera. VSV-OROV lacking this variable region of Gc was also immunogenic in mice producing neutralizing sera that recognize additional regions of Gc. Challenge of both sets of immunized mice with wild-type OROV shows that the VSV-OROV chimeras reduce wild-type viral infection and suggest that antibodies that recognize the variable N terminus of Gc afford less protection than those that target more conserved regions of Gc. IMPORTANCE Oropouche virus (OROV), an orthobunyavirus found in Central and South America, is an emerging public health challenge that causes debilitating febrile illness. OROV is transmitted by arthropods, and increasing mobilization has the potential to significantly increase the spread of OROV globally. Despite this, no therapeutics or vaccines have been developed to combat infection. Using vesicular stomatitis (VSV) as a backbone, we developed a chimeric virus bearing the OROV glycoproteins (VSV-OROV) and tested its ability to elicit a neutralizing antibody response. Our results demonstrate that VSV-OROV produces a strong neutralizing antibody response that is at least partially targeted to the N-terminal region of Gc. Importantly, vaccination with VSV-OROV reduces viral loads in mice challenged with wild-type virus. These data provide novel evidence that targeting the OROV glycoproteins may be an effective vaccination strategy to combat OROV infection.
- Published
- 2021
- Full Text
- View/download PDF
16. A recombinant herpes virus expressing influenza hemagglutinin confers protection and induces antibody-dependent cellular cytotoxicity.
- Author
-
Kaugars K, Dardick J, de Oliveira AP, Weiss KA, Lukose R, Kim J, Leung L, Rajagopalan S, Wolin S, Akabas L, Knipe DM, Bajic G, and Jacobs WR Jr
- Subjects
- Animals, Antibodies, Viral blood, Antibodies, Viral immunology, Female, Herpes Simplex prevention & control, Herpesvirus 1, Human physiology, Herpesvirus 2, Human physiology, Influenza Vaccines immunology, Male, Mice, Mice, Inbred C57BL, Orthomyxoviridae Infections immunology, Antibody-Dependent Cell Cytotoxicity immunology, Hemagglutinin Glycoproteins, Influenza Virus immunology, Herpes Simplex immunology, Influenza Vaccines administration & dosage, Orthomyxoviridae immunology, Orthomyxoviridae Infections prevention & control
- Abstract
Despite widespread yearly vaccination, influenza leads to significant morbidity and mortality across the globe. To make a more broadly protective influenza vaccine, it may be necessary to elicit antibodies that can activate effector functions in immune cells, such as antibody-dependent cellular cytotoxicity (ADCC). There is growing evidence supporting the necessity for ADCC in protection against influenza and herpes simplex virus (HSV), among other infectious diseases. An HSV-2 strain lacking the essential glycoprotein D (gD), was used to create ΔgD-2, which is a highly protective vaccine against lethal HSV-1 and HSV-2 infection in mice. It also elicits high levels of IgG2c antibodies that bind FcγRIV, a receptor that activates ADCC. To make an ADCC-eliciting influenza vaccine, we cloned the hemagglutinin ( HA ) gene from an H1N1 influenza A strain into the ΔgD-2 HSV vector. Vaccination with ΔgD-2::HA
PR8 was protective against homologous influenza challenge and elicited an antibody response against HA that inhibits hemagglutination (HAI+ ), is predominantly IgG2c, strongly activates FcγRIV, and protects against influenza challenge following passive immunization of naïve mice. Prior exposure of mice to HSV-1, HSV-2, or a replication-defective HSV-2 vaccine ( dl5-29 ) does not reduce protection against influenza by ΔgD-2::HAPR8 This vaccine also continues to elicit protection against both HSV-1 and HSV-2, including high levels of IgG2c antibodies against HSV-2. Mice lacking the interferon-α/β receptor and mice lacking the interferon-γ receptor were also protected against influenza challenge by ΔgD-2::HAPR8 Our results suggest that ΔgD-2 can be used as a vaccine vector against other pathogens, while also eliciting protective anti-HSV immunity., Competing Interests: Competing interest statement: The laboratories of W.R.J. receive financial support for sponsored research from X-Vax Technology, Inc., which holds licenses to several patents and patent applications related to ΔgD-2 vaccines, antibodies, and their use. W.R.J. serves as scientific advisor and consultant for the company. W.R.J. has equity interests in X-Vax Technology, Inc. W.R.J. is a coinventor on US patent no. 9,999,665 B2 “RECOMBINANT HERPES SIMPLEX VIRUS 2 (HSV-2) VACCINE VECTORS” and other patents related to ΔgD-2 vaccines, antibodies, and their use. D.M.K. is a coinventor on a patent on HSV-2 dl5-29 vaccine technology that is licensed by Harvard University to Sanofi Pasteur.- Published
- 2021
- Full Text
- View/download PDF
17. Expression of SARS coronavirus 1 spike protein from a herpesviral vector induces innate immune signaling and neutralizing antibody responses.
- Author
-
Kurt-Jones EA, Dudek TE, Watanabe D, Mandell L, Che J, Zhou S, Cao L, Greenough T, Babcock GJ, Diaz F, Oh HS, Zhou C, Finberg RW, and Knipe DM
- Subjects
- Animals, Antibodies, Neutralizing blood, Antibodies, Viral blood, Antibodies, Viral immunology, Cell Fusion, Cell Line, Cytokines immunology, Genetic Vectors, Humans, Mice, Severe acute respiratory syndrome-related coronavirus genetics, Signal Transduction, Spike Glycoprotein, Coronavirus genetics, Spike Glycoprotein, Coronavirus metabolism, Vaccines, Virus-Like Particle immunology, Viral Vaccines immunology, Antibodies, Neutralizing immunology, Herpesvirus 1, Human genetics, Immunity, Innate, Severe acute respiratory syndrome-related coronavirus immunology, Spike Glycoprotein, Coronavirus immunology
- Abstract
SARS coronavirus 1 (SARS-CoV-1) causes a respiratory infection that can lead to acute respiratory distress characterized by inflammation and high levels of cytokines in the lung tissue. In this study we constructed a herpes simplex virus 1 replication-defective mutant vector expressing SARS-CoV-1 spike protein as a potential vaccine vector and to probe the effects of spike protein on host cells. The spike protein expressed from this vector is functional in that it localizes to the surface of infected cells and induces fusion of ACE2-expressing cells. In immunized mice, the recombinant vector induced antibodies that bind to spike protein in an ELISA assay and that show neutralizing activity. The spike protein expressed from this vector can induce the expression of cytokines in an ACE2-independent, MyD88-dependent process. These results argue that the SARS-CoV-1 spike protein intrinsically activates signaling pathways that induce cytokines and contribute directly to the inflammatory process of SARS., (Copyright © 2021 Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
18. Regulation of host and virus genes by neuronal miR-138 favours herpes simplex virus 1 latency.
- Author
-
Sun B, Yang X, Hou F, Yu X, Wang Q, Oh HS, Raja P, Pesola JM, Vanni EAH, McCarron S, Morris-Love J, Ng AHM, Church GM, Knipe DM, Coen DM, and Pan D
- Subjects
- Animals, Gene Expression Regulation, Viral, Herpes Simplex genetics, Herpesvirus 1, Human metabolism, Host-Pathogen Interactions, Humans, Immediate-Early Proteins genetics, Immediate-Early Proteins metabolism, Male, Mice, MicroRNAs genetics, Neurons virology, Organic Cation Transporter 1 genetics, Organic Cation Transporter 1 metabolism, RNA, Viral genetics, RNA, Viral metabolism, Ubiquitin-Protein Ligases genetics, Ubiquitin-Protein Ligases metabolism, Herpes Simplex metabolism, Herpes Simplex virology, Herpesvirus 1, Human genetics, MicroRNAs metabolism, Neurons metabolism, Virus Latency
- Abstract
MicroRNA miR-138, which is highly expressed in neurons, represses herpes simplex virus 1 (HSV-1) lytic cycle genes by targeting viral ICP0 messenger RNA, thereby promoting viral latency in mice. We found that overexpressed miR-138 also represses lytic processes independently of ICP0 in murine and human neuronal cells; therefore, we investigated whether miR-138 has targets besides ICP0. Using genome-wide RNA sequencing/photoactivatable ribonucleoside-enhanced crosslinking and immunoprecipitation followed by short interfering RNA knockdown of candidate targets, we identified the host Oct-1 and Foxc1 messenger mRNAs as miR-138's targets, whose gene products are transcription factors important for HSV-1 replication in neuronal cells. OCT-1 has a known role in the initiation of HSV transcription. Overexpression of FOXC1, which was not known to affect HSV-1, promoted HSV-1 replication in murine neurons and ganglia. CRISPR-Cas9 knockout of FOXC1 reduced viral replication, lytic gene expression and miR-138 repression in murine neuronal cells. FOXC1 also collaborated with ICP0 to decrease heterochromatin on viral genes and compensated for the defect of an ICP0-null virus. In summary, miR-138 targets ICP0, Oct-1 and Foxc1 to repress HSV-1 lytic cycle genes and promote epigenetic gene silencing, which together enable favourable conditions for latent infection.
- Published
- 2021
- Full Text
- View/download PDF
19. ATRX limits the accessibility of histone H3-occupied HSV genomes during lytic infection.
- Author
-
Cabral JM, Cushman CH, Sodroski CN, and Knipe DM
- Subjects
- Cells, Cultured, Gene Expression Regulation, Viral genetics, Genome, Viral genetics, Herpes Simplex genetics, Herpes Simplex pathology, Herpes Simplex virology, Herpesvirus 1, Human genetics, Herpesvirus 1, Human metabolism, Heterochromatin metabolism, Histones metabolism, Host-Pathogen Interactions genetics, Humans, Virus Physiological Phenomena genetics, Herpesvirus 1, Human physiology, Virus Replication genetics, X-linked Nuclear Protein physiology
- Abstract
Histones are rapidly loaded on the HSV genome upon entry into the nucleus of human fibroblasts, but the effects of histone loading on viral replication have not been fully defined. We showed recently that ATRX is dispensable for de novo deposition of H3 to HSV genomes after nuclear entry but restricted infection through maintenance of viral heterochromatin. To further investigate the roles that ATRX and other histone H3 chaperones play in restriction of HSV, we infected human fibroblasts that were systematically depleted of nuclear H3 chaperones. We found that the ATRX/DAXX complex is unique among nuclear H3 chaperones in its capacity to restrict ICP0-null HSV infection. Only depletion of ATRX significantly alleviated restriction of viral replication. Interestingly, no individual nuclear H3 chaperone was required for deposition of H3 onto input viral genomes, suggesting that during lytic infection, H3 deposition may occur through multiple pathways. ChIP-seq for total histone H3 in control and ATRX-KO cells infected with ICP0-null HSV showed that HSV DNA is loaded with high levels of histones across the entire viral genome. Despite high levels of H3, ATAC-seq analysis revealed that HSV DNA is highly accessible, especially in regions of high GC content, and is not organized largely into ordered nucleosomes during lytic infection. ATRX reduced accessibility of viral DNA to the activity of a TN5 transposase and enhanced accumulation of viral DNA fragment sizes associated with nucleosome-like structures. Together, these findings support a model in which ATRX restricts viral infection by altering the structure of histone H3-loaded viral chromatin that reduces viral DNA accessibility for transcription. High GC rich regions of the HSV genome, especially the S component inverted repeats of the HSV-1 genome, show increased accessibility, which may lead to increased ability to transcribe the IE genes encoded in these regions during initiation of infection., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2021
- Full Text
- View/download PDF
20. Herpes Simplex Virus 1 Manipulates Host Cell Antiviral and Proviral DNA Damage Responses.
- Author
-
Mertens ME and Knipe DM
- Subjects
- Ataxia Telangiectasia Mutated Proteins genetics, Ataxia Telangiectasia Mutated Proteins metabolism, Cell Line, Clustered Regularly Interspaced Short Palindromic Repeats genetics, DNA, Viral genetics, Fibroblasts virology, Foreskin cytology, Genome, Viral, Humans, Male, Virus Replication physiology, Antiviral Agents pharmacology, DNA Damage, Herpesvirus 1, Human drug effects, Herpesvirus 1, Human genetics, Host-Pathogen Interactions, Proviruses genetics
- Abstract
Cells activate their DNA damage response (DDR) in response to DNA virus infection, including adenoviruses, papillomaviruses, polyomaviruses, and herpesviruses. In this study, we found that the DDR kinase pathways activated in normal human fibroblasts by herpes simplex virus 1 (HSV-1) input genomic DNA, HSV-1 replicating DNA, and progeny DNA and in uninfected cells treated with etoposide are different. We also found using clustered regularly interspaced palindromic repeat (CRISPR)-Cas9 technology that different host gene products are required for the DDR in uninfected versus infected cells. Individual DDR components can be proviral or antiviral in that ataxia-telangiectasia mutated (ATM) and p53 promote and Mre11 restricts replication of ICP0-null HSV-1, but ICP0 expression eliminates these DDR effects. Thus, in total, these results argue that HSV-1 manipulates the host cell DDR to utilize specific components for its optimal replication while inactivating the antiviral aspects of the DDR. IMPORTANCE We investigated the relationship between the DNA damage response, a collection of vital cellular pathways that repair potentially lethal damage to the genome, and the DNA virus herpes simplex virus 1. We found that infection by the virus triggers the DNA damage response, and key proteins that mediate this response have opposing effects on the replication and production of progeny viruses. Our work provides novel insights into the relationship between DNA virus infection and the cellular response to the viral genome. We speculate that viral gene products modulate this response, providing potentially novel targets for therapeutic intervention against the virus., (Copyright © 2021 Mertens and Knipe.)
- Published
- 2021
- Full Text
- View/download PDF
21. Tripartite Motif 22 (TRIM22) protein restricts herpes simplex virus 1 by epigenetic silencing of viral immediate-early genes.
- Author
-
Reddi TS, Merkl PE, Lim SY, Letvin NL, and Knipe DM
- Subjects
- Cell Line, Disease Susceptibility immunology, Herpesvirus 1, Human genetics, Herpesvirus 1, Human immunology, Heterochromatin metabolism, Histones metabolism, Humans, Minor Histocompatibility Antigens immunology, Repressor Proteins immunology, Tripartite Motif Proteins immunology, Virus Replication genetics, Epigenesis, Genetic, Gene Silencing, Genes, Immediate-Early, Herpesvirus 1, Human physiology, Minor Histocompatibility Antigens physiology, Repressor Proteins physiology, Tripartite Motif Proteins physiology
- Abstract
Intrinsic resistance is a crucial line of defense against virus infections, and members of the Tripartite Ring Interaction Motif (TRIM) family of proteins are major players in this system, such as cytoplasmic TRIM5α or nuclear promyelocytic leukemia (PML/TRIM19) protein. Previous reports on the antiviral function of another TRIM protein, TRIM22, emphasized its innate immune role as a Type I and Type II interferon-stimulated gene against RNA viruses. This study shows that TRIM22 has an additional intrinsic role against DNA viruses. Here, we report that TRIM22 is a novel restriction factor of HSV-1 and limits ICP0-null virus replication by increasing histone occupancy and heterochromatin, thereby reducing immediate-early viral gene expression. The corresponding wild-type equivalent of the virus evades the TRIM22-specific restriction by a mechanism independent of ICP0-mediated degradation. We also demonstrate that TRIM22 inhibits other DNA viruses, including representative members of the β- and γ- herpesviruses. Allelic variants in TRIM22 showed different degrees of anti-herpesviral activity; thus, TRIM22 genetic variability may contribute to the varying susceptibility to HSV-1 infection in humans. Collectively, these results argue that TRIM22 is a novel restriction factor and expand the list of restriction factors functioning in the infected cell nucleus to counter DNA virus infection., Competing Interests: I have read the journal's policy and the authors of this manuscript have the following competing interests: Author Norman L. Letvin was unable to confirm their authorship contributions. On their behalf, the corresponding author has reported their contributions to the best of their knowledge.
- Published
- 2021
- Full Text
- View/download PDF
22. Ensuring vaccine safety.
- Author
-
Knipe DM, Levy O, Fitzgerald KA, and Mühlberger E
- Subjects
- Animal Experimentation, Animals, COVID-19 Vaccines immunology, Humans, Immunogenicity, Vaccine, Mice, United States, United States Food and Drug Administration, COVID-19 prevention & control, COVID-19 Vaccines adverse effects
- Published
- 2020
- Full Text
- View/download PDF
23. Screening Method for CRISPR/Cas9 Inhibition of a Human DNA Virus: Herpes Simplex Virus.
- Author
-
Neuhausser WM, Oh HS, Eggan P, Angelova M, Kirchner R, Eggan KC, and Knipe DM
- Abstract
The efficiency of cleavage of individual CRISPR/Cas9-sgRNAs remains difficult to predict based on the CRISPR target sequence alone. Different intracellular environments (dependent on cell type or cell cycle state for example) may affect sgRNA efficiency by altering accessibility of genomic DNA through DNA modifications such as epigenetic marks and DNA-binding proteins ( e.g. , histones) as well as alteration of the chromatin state of genomic DNA within the nucleus. We recently reported a multi-step screening method for the identification of efficient sgRNAs targeting the Herpes simplex virus (HSV-1) genome and reported a differential mechanism for viral inhibition by CRISPR-Cas9 in the latent versus lytic phase. The screening platform detailed in this protocol allows step-by-step testing of the efficiency of cleavage in a cell-free system and in the context of viral target cells such as human foreskin fibroblasts followed by functional testing of the effects of CRISPR/sgRNA on viral protein expression, replication, and reactivation. This strategy could be readily applied to other target cells such as pluripotent stem cell-derived human sensory neurons or other human DNA viruses., Competing Interests: Competing interestsThe authors declare no conflicts of interest or competing interests., (Copyright © The Authors; exclusive licensee Bio-protocol LLC.)
- Published
- 2020
- Full Text
- View/download PDF
24. Vaccination Route as a Determinant of Protective Antibody Responses against Herpes Simplex Virus.
- Author
-
Burn Aschner C, Pierce C, Knipe DM, and Herold BC
- Abstract
Herpes simplex viruses (HSV) are significant global health problems associated with mucosal and neurologic disease. Prior experimental vaccines primarily elicited neutralizing antibodies targeting glycoprotein D (gD), but those that advanced to clinical efficacy trials have failed. Preclinical studies with an HSV-2 strain deleted in gD (ΔgD-2) administered subcutaneously demonstrated that it elicited a high titer, weakly neutralizing antibodies that activated Fcg receptors to mediate antibody-dependent cellular cytotoxicity (ADCC), and completely protected mice against lethal disease and latency following vaginal or skin challenge with HSV-1 or HSV-2. Vaccine efficacy, however, may be impacted by dose and route of immunization. Thus, the current studies were designed to compare immunogenicity and efficacy following different routes of vaccination with escalating doses of ΔgD-2. We compared ΔgD-2 with two other candidates: recombinant gD protein combined with aluminum hydroxide and monophosphoryl lipid A adjuvants and a replication-defective virus deleted in two proteins involved in viral replication, dl 5-29. Compared to the subcutaneous route, intramuscular and/or intradermal immunization resulted in increased total HSV antibody responses for all three vaccines and boosted the ADCC, but not the neutralizing response to ΔgD and dl 5-29. The adjuvanted gD protein vaccine provided only partial protection and failed to elicit ADCC independent of route of administration. In contrast, the increased ADCC following intramuscular or intradermal administration of DgD-2 or dl 5-29 translated into significantly increased protection. The DgD-2 vaccine provided 100% protection at doses as low as 5 × 10
4 pfu when administered intramuscularly or intradermally, but not subcutaneously. However, administration of a combination of low dose subcutaneous DgD-2 and adjuvanted gD protein resulted in greater protection than low dose DgD-2 alone indicating that gD neutralizing antibodies may contribute to protection. Taken together, these results demonstrate that ADCC provides a more predictive correlate of protection against HSV challenge in mice and support intramuscular or intradermal routes of vaccination.- Published
- 2020
- Full Text
- View/download PDF
25. Model of vaccine efficacy against HSV-2 superinfection of HSV-1 seropositive mice demonstrates protection by antibodies mediating cellular cytotoxicity.
- Author
-
Burn Aschner C, Knipe DM, and Herold BC
- Abstract
A majority of the world's population is infected with HSV-1, highlighting the need for vaccines that are effective in HSV-1-seropositive hosts. We established a superinfection model by infecting mice intranasally with a sublethal dose of HSV-1, which results in high rates of seropositive, latently infected mice susceptible to HSV-2 superinfection. Sublethal HSV-1 induced a predominantly neutralizing antibody response. Vaccination of HSV-1-seropositive mice with recombinant adjuvanted glycoprotein D (rgD-2) failed to significantly boost HSV total or neutralizing antibody responses and provided no significant increased protection against HSV-2 superinfection compared to control-vaccinated HSV-1-seropositive mice. In contrast, immunization with a single-cycle virus deleted in gD (ΔgD-2) significantly boosted total HSV-specific antibody titers and elicited new antibody-dependent cell-mediated cytotoxicity responses, providing complete protection from death following HSV-2 superinfection. This model recapitulates clinical responses to natural infection and the rgD-2 vaccine trial outcomes and suggests that ΔgD-2 may prove protective in HSV-1-seropositive hosts., Competing Interests: Competing interestsB.C.H. is an inventor on patents on the HSV-2 ΔgD-2 vaccine and is a scientific consultant for X-VAX, Technologies. None of the other authors report any conflicts of interest., (© The Author(s) 2020.)
- Published
- 2020
- Full Text
- View/download PDF
26. Neoleukin-2 enhances anti-tumour immunity downstream of peptide vaccination targeted by an anti-MHC class II VHH.
- Author
-
Crowley SJ, Bruck PT, Bhuiyan MA, Mitchell-Gears A, Walsh MJ, Zhangxu K, Ali LR, Jeong HJ, Ingram JR, Knipe DM, Ploegh HL, Dougan M, and Dougan SK
- Subjects
- Animals, Antigens, Neoplasm immunology, Autoantigens immunology, CD8-Positive T-Lymphocytes immunology, Camelids, New World immunology, Cancer Vaccines administration & dosage, Cancer Vaccines immunology, Cell Line, Tumor, Drug Synergism, Interleukin-2 immunology, Melanoma immunology, Mice, Peptides immunology, Recombinant Proteins administration & dosage, Recombinant Proteins immunology, Vaccines, Subunit, Xenograft Model Antitumor Assays, Histocompatibility Antigens Class II immunology, Interleukin-2 administration & dosage, Melanoma drug therapy, Peptides administration & dosage, Single-Domain Antibodies metabolism
- Abstract
Cancer-specific mutations can lead to peptides of unique sequence presented on MHC class I to CD8 T cells. These neoantigens can be potent tumour-rejection antigens, appear to be the driving force behind responsiveness to anti-CTLA-4 and anti-PD1/L1-based therapies and have been used to develop personalized vaccines. The platform for delivering neoantigen-based vaccines has varied, and further optimization of both platform and adjuvant will be necessary to achieve scalable vaccine products that are therapeutically effective at a reasonable cost. Here, we developed a platform for testing potential CD8 T cell tumour vaccine candidates. We used a high-affinity alpaca-derived VHH against MHC class II to deliver peptides to professional antigen-presenting cells. We show in vitro and in vivo that peptides derived from the model antigen ovalbumin are better able to activate naive ovalbumin-specific CD8 T cells when conjugated to an MHC class II-specific VHH when compared with an irrelevant control VHH. We then used the VHH-peptide platform to evaluate a panel of candidate neoantigens in vivo in a mouse model of pancreatic cancer. None of the candidate neoantigens tested led to protection from tumour challenge; however, we were able to show vaccine-induced CD8 T cell responses to a melanoma self-antigen that was augmented by combination therapy with the synthetic cytokine mimetic Neo2/15.
- Published
- 2020
- Full Text
- View/download PDF
27. Herpesviral lytic gene functions render the viral genome susceptible to novel editing by CRISPR/Cas9.
- Author
-
Oh HS, Neuhausser WM, Eggan P, Angelova M, Kirchner R, Eggan KC, and Knipe DM
- Subjects
- Animals, Base Sequence, Cell Line, DNA Repair genetics, DNA, Viral genetics, Gene Expression Regulation, Viral, Herpesvirus 1, Human genetics, Humans, Models, Genetic, Mutagenesis genetics, RNA, Guide, CRISPR-Cas Systems genetics, Virus Replication genetics, CRISPR-Associated Protein 9 metabolism, CRISPR-Cas Systems genetics, Gene Editing, Genes, Viral, Herpesviridae genetics
- Abstract
Herpes simplex virus (HSV) establishes lifelong latent infection and can cause serious human disease, but current antiviral therapies target lytic but not latent infection. We screened for sgRNAs that cleave HSV-1 DNA sequences efficiently in vitro and used these sgRNAs to observe the first editing of quiescent HSV-1 DNA. The sgRNAs targeted lytic replicating viral DNA genomes more efficiently than quiescent genomes, consistent with the open structure of lytic chromatin. Editing of latent genomes caused short indels while editing of replicating genomes produced indels, linear molecules, and large genomic sequence loss around the gRNA target site. The HSV ICP0 protein and viral DNA replication increased the loss of DNA sequences around the gRNA target site. We conclude that HSV, by promoting open chromatin needed for viral gene expression and by inhibiting the DNA damage response, makes the genome vulnerable to a novel form of editing by CRISPR-Cas9 during lytic replication., Competing Interests: HO, WN, MA, KE has patent applications pending. U.S. Patent application No. 62/365,826, International Patent Application PCT/US2017/043225, PE, RK No competing interests declared, DK Reviewing editor, eLife, (© 2019, Oh et al.)
- Published
- 2019
- Full Text
- View/download PDF
28. Immunization of BLT Humanized Mice Redirects T Cell Responses to Gag and Reduces Acute HIV-1 Viremia.
- Author
-
Claiborne DT, Dudek TE, Maldini CR, Power KA, Ghebremichael M, Seung E, Mellors EF, Vrbanac VD, Krupp K, Bisesi A, Tager AM, Knipe DM, Boutwell CL, and Allen TM
- Subjects
- Acute Disease, Animals, Biological Evolution, CD4-Positive T-Lymphocytes, CD8-Positive T-Lymphocytes, Disease Models, Animal, HIV Infections metabolism, Host-Pathogen Interactions, Humans, Immunization, Mice, Mice, Transgenic, T-Lymphocytes metabolism, Viral Load, HIV Infections immunology, HIV Infections virology, HIV-1 immunology, T-Lymphocytes immunology, T-Lymphocytes virology, Viremia, gag Gene Products, Human Immunodeficiency Virus immunology
- Abstract
BLT (bone marrow-liver-thymus) humanized mice, which reconstitute a functional human immune system, develop prototypic human virus-specific CD8
+ T cell responses following infection with human immunodeficiency virus type 1 (HIV-1). We explored the utility of the BLT model for HIV-1 vaccine development by immunizing BLT mice against the conserved viral Gag protein, utilizing a rapid prime-boost protocol of poly(lactic-co-glycolic) acid microparticles and a replication-defective herpes simplex virus (HSV) recombinant vector. After HIV-1 challenge, the mice developed broad, proteome-wide gamma interferon-positive (IFN-γ+ ) T cell responses against HIV-1 that reached magnitudes equivalent to what is observed in HIV-1-infected individuals. The functionality of these responses was underscored by the consistent emergence of escape mutations in multiple CD8+ T cell epitopes during the course of infection. Although prechallenge vaccine-induced responses were largely undetectable, the Gag immunization increased both the magnitude and the kinetics of anamnestic Gag-specific T cell responses following HIV-1 infection, and the magnitude of these postchallenge Gag-specific responses was inversely correlated with acute HIV-1 viremia. Indeed, Gag immunization was associated with a modest but significant 0.5-log reduction in HIV-1 viral load when analyzed across four experimental groups of BLT mice. Notably, the HSV vector induced elevated plasma concentrations of polarizing cytokines and chemotactic factors, including interleukin-12p70 (IL-12p70) and MIP-1α, which were positively correlated with the magnitude of Gag-specific responses. Overall, these results support the ability of BLT mice to recapitulate human pathogen-specific T cell responses and to respond to immunization; however, additional improvements to the model are required to develop a robust system for testing HIV-1 vaccine efficacy. IMPORTANCE Advances in the development of humanized mice have raised the possibility of a small-animal model for preclinical testing of an HIV-1 vaccine. Here, we describe the capacity of BLT humanized mice to mount broadly directed HIV-1-specific human T cell responses that are functionally active, as indicated by the rapid emergence of viral escape mutations. Although immunization of BLT mice with the conserved viral Gag protein did not result in detectable prechallenge responses, it did increase the magnitude and kinetics of postchallenge Gag-specific T cell responses, which was associated with a modest but significant reduction in acute HIV-1 viremia. Additionally, the BLT model revealed immunization-associated increases in the plasma concentrations of immunomodulatory cytokines and chemokines that correlated with more robust T cell responses. These data support the potential utility of the BLT humanized mouse for HIV-1 vaccine development but suggest that additional improvements to the model are warranted., (Copyright © 2019 American Society for Microbiology.)- Published
- 2019
- Full Text
- View/download PDF
29. ABIN-1 heterozygosity sensitizes to innate immune response in both RIPK1-dependent and RIPK1-independent manner.
- Author
-
Su Z, Dziedzic SA, Hu D, Barrett VJ, Broekema N, Li W, Qian L, Jia N, Ofengeim D, Najafov A, Zhu H, Knipe DM, and Yuan J
- Subjects
- Adaptor Proteins, Signal Transducing genetics, Animals, Cells, Cultured, Female, Genotype, Male, Mice, Mice, Knockout, Receptor-Interacting Protein Serine-Threonine Kinases antagonists & inhibitors, Receptor-Interacting Protein Serine-Threonine Kinases genetics, Adaptor Proteins, Signal Transducing immunology, Immunity, Innate immunology, Receptor-Interacting Protein Serine-Threonine Kinases immunology
- Abstract
ABIN-1 (encoded by the gene Tnip1) is a ubiquitin-binding protein that can interact with ubiquitin-editing enzyme A20 (encoded by the gene TNFAIP3) to restrain the activation of necroptosis and NF-κB activation. Genetic variants in the genes Tnip1 and TNFAIP3 are both strongly associated with susceptibility to autoimmune chronic inflammatory diseases such as psoriasis vulgaris and systemic lupus erythematosus (SLE) in humans. Here we investigated the mechanism by which ABIN-1 regulated innate immune responses. We show that ABIN-1 heterozygosity sensitizes cells to antiviral response by mediating NF-κB-dependent and RIPK1-independent expression of pattern recognition molecules, including TLR3, RIG-I, and MDA5, in MEFs. Furthermore, we demonstrate that increased interaction of ABIN-1 and A20 with prolonged poly(I:C) stimulation of WT cells leads to A20-dependent reduction of ABIN-1 protein. Finally, we show that ABIN-1 heterozygosity sensitizes innate immune response of Abin-1
+/ - mice in vivo by promoting the production of proinflammatory cytokines, which can be blocked upon inhibition of RIPK1 kinase. Inhibition of RIPK1 kinase activity in vivo partially reduces the expression of MDA5, RIG-I, and caspase-11 in Abin-1+/ - mice but not in WT mice. Thus, we conclude that ABIN-1 is a suppressor of innate immune response and the interaction of ABIN-1 with A20 controls innate immunity response through the NF-κB pathway and in both RIPK1 kinase activity-independent and dependent manner.- Published
- 2019
- Full Text
- View/download PDF
30. Maternal immunization confers protection against neonatal herpes simplex mortality and behavioral morbidity.
- Author
-
Patel CD, Backes IM, Taylor SA, Jiang Y, Marchant A, Pesola JM, Coen DM, Knipe DM, Ackerman ME, and Leib DA
- Subjects
- Animals, Animals, Newborn, Antibodies, Neutralizing immunology, Antibodies, Viral blood, Antibodies, Viral immunology, Anxiety etiology, Female, Herpes Simplex blood, Herpes Simplex virology, Herpes Simplex Virus Vaccines immunology, Herpesvirus 1, Human immunology, Humans, Immunoglobulin G blood, Mice, Morbidity, Pregnancy, Pregnancy Complications, Infectious blood, Pregnancy Complications, Infectious virology, Trigeminal Ganglion pathology, Trigeminal Ganglion virology, Vaccination, Behavior, Animal, Herpes Simplex immunology, Herpes Simplex prevention & control, Immunization, Pregnancy Complications, Infectious immunology, Pregnancy Complications, Infectious prevention & control
- Abstract
Neonatal herpes simplex virus (nHSV) infections cause devastating morbidity and mortality in infants. Most nHSV cases are associated with primary maternal infection, consistent with the hypothesis that maternal immunity is protective. In humans, we found HSV-specific neutralizing antibodies in newborns of immune mothers, indicating that placentally transferred HSV-specific antibody is protective. Using a murine model, we showed that passive administration of HSV-specific antibody to dams prevented disseminated infection and mortality in pups. Maternal immunization with an HSV-2 replication-defective vaccine candidate, dl 5-29, led to transfer of HSV-specific antibodies into neonatal circulation that protected against nHSV neurological disease and death. Furthermore, we observed considerable anxiety-like behavior in adult mice that had been infected with low doses of HSV as neonates, despite a notable lack of signs of infection. This phenotype suggests that nHSV infection can have an unsuspected and permanent impact on behavior. These behavioral sequelae of nHSV were prevented by maternal immunization with dl 5-29, demonstrating an unexpected benefit of immunization. These findings also support the general concept that maternal immunization can prevent neurotropic neonatal infections and associated morbidity and mortality., (Copyright © 2019 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)
- Published
- 2019
- Full Text
- View/download PDF
31. Classification of human Herpesviridae proteins using Domain-architecture Aware Inference of Orthologs (DAIO).
- Author
-
Zmasek CM, Knipe DM, Pellett PE, and Scheuermann RH
- Subjects
- Capsid Proteins genetics, Capsid Proteins metabolism, Gene Duplication, Gene Expression Regulation, Viral, Peptide Hydrolases, Protein Domains, Uracil-DNA Glycosidase chemistry, Uracil-DNA Glycosidase genetics, Uracil-DNA Glycosidase metabolism, Herpesviridae genetics, Herpesviridae metabolism, Phylogeny, Viral Proteins chemistry, Viral Proteins genetics
- Abstract
We developed a computational approach called Domain-architecture Aware Inference of Orthologs (DAIO) for the analysis of protein orthology by combining phylogenetic and protein domain-architecture information. Using DAIO, we performed a systematic study of the proteomes of all human Herpesviridae species to define Strict Ortholog Groups (SOGs). In addition to assessing the taxonomic distribution for each protein based on sequence similarity, we performed a protein domain-architecture analysis for every protein family and computationally inferred gene duplication events. While many herpesvirus proteins have evolved without any detectable gene duplications or domain rearrangements, numerous herpesvirus protein families do exhibit complex evolutionary histories. Some proteins acquired additional domains (e.g., DNA polymerase), whereas others show a combination of domain acquisition and gene duplication (e.g., betaherpesvirus US22 family), with possible functional implications. This novel classification system of SOGs for human Herpesviridae proteins is available through the Virus Pathogen Resource (ViPR, www.viprbrc.org)., (Copyright © 2019. Published by Elsevier Inc.)
- Published
- 2019
- Full Text
- View/download PDF
32. Herpes Simplex Virus 1 Lytic Infection Blocks MicroRNA (miRNA) Biogenesis at the Stage of Nuclear Export of Pre-miRNAs.
- Author
-
Pan D, Li G, Morris-Love J, Qi S, Feng L, Mertens ME, Jurak I, Knipe DM, and Coen DM
- Subjects
- Animals, Blotting, Northern, Cell Line, Disease Models, Animal, Gene Expression Regulation, Herpes Simplex pathology, Humans, Immediate-Early Proteins genetics, Immediate-Early Proteins metabolism, Mice, Mutation, RNA-Binding Proteins genetics, RNA-Binding Proteins metabolism, Trigeminal Ganglion pathology, Trigeminal Ganglion virology, Viral Proteins genetics, Viral Proteins metabolism, Virus Latency, Active Transport, Cell Nucleus, Herpes Simplex virology, Herpesvirus 1, Human growth & development, Host-Pathogen Interactions, MicroRNAs metabolism, RNA Precursors metabolism
- Abstract
Herpes simplex virus 1 (HSV-1) switches between two infection programs, productive ("lytic") and latent infection. Some HSV-1 microRNAs (miRNAs) have been hypothesized to help control this switch, and yet little is known about regulation of their expression. Using Northern blot analyses, we found that, despite inherent differences in biogenesis efficiency among six HSV-1 miRNAs, all six exhibited high pre-miRNA/miRNA ratios during lytic infection of different cell lines and, when detectable, in acutely infected mouse trigeminal ganglia. In contrast, considerably lower ratios were observed in latently infected ganglia and in cells transduced with lentiviral vectors expressing the miRNAs, suggesting that HSV-1 lytic infection blocks miRNA biogenesis. This phenomenon is not specific to viral miRNAs, as a host miRNA expressed from recombinant HSV-1 also exhibited high pre-miRNA/miRNA ratios late during lytic infection. The levels of most of the mature miRNAs remained stable during infection in the presence of actinomycin D, indicating that the high ratios are due to inefficient pre-miRNA conversion to miRNA. Cellular fractionation experiments showed that late (but not early) during infection, pre-miRNAs were enriched in the nucleus and depleted in the cytoplasm, indicating that nuclear export was blocked. A mutation eliminating ICP27 expression or addition of acyclovir reduced pre-miRNA/miRNA ratios, but mutations drastically reducing Us11 expression did not. Thus, HSV-1 lytic infection inhibits miRNA biogenesis at the step of nuclear export and does so in an ICP27- and viral DNA synthesis-dependent manner. This mechanism may benefit the virus by reducing expression of repressive miRNAs during lytic infection while permitting elevated expression during latency. IMPORTANCE Various mechanisms have been identified by which viruses target host small RNA biogenesis pathways to achieve optimal infection outcomes. Herpes simplex virus 1 (HSV-1) is a ubiquitous human pathogen whose successful persistence in the host entails both productive ("lytic") and latent infection. Although many HSV-1 miRNAs have been discovered and some are thought to help control the lytic/latent switch, little is known about regulation of their biogenesis. By characterizing expression of both pre-miRNAs and mature miRNAs under various conditions, this study revealed striking differences in miRNA biogenesis between lytic and latent infection and uncovered a regulatory mechanism that blocks pre-miRNA nuclear export and is dependent on viral protein ICP27 and viral DNA synthesis. This mechanism represents a new virus-host interaction that could limit the repressive effects of HSV-1 miRNAs hypothesized to promote latency and may shed light on the regulation of miRNA nuclear export, which has been relatively unexplored., (Copyright © 2019 Pan et al.)
- Published
- 2019
- Full Text
- View/download PDF
33. Role for a Filamentous Nuclear Assembly of IFI16, DNA, and Host Factors in Restriction of Herpesviral Infection.
- Author
-
Merkl PE and Knipe DM
- Subjects
- Cell Line, Epigenesis, Genetic, Gene Expression Regulation, Viral, Gene Silencing, Humans, Microscopy, DNA, Viral metabolism, Herpesvirus 1, Human immunology, Herpesvirus 1, Human physiology, Nuclear Proteins metabolism, Phosphoproteins metabolism, Protein Multimerization, Virus Replication
- Abstract
Several host cell nuclear factors are known to restrict herpes simplex virus 1 (HSV-1) replication, but their mechanisms of action remain to be defined. Interferon-inducible protein 16 (IFI16) and the nuclear domain 10-associated proteins, such as promyelocytic leukemia (PML) protein, localize to input viral genomes, but they are also capable of restricting progeny viral transcription. In this study, we used structured illumination microscopy to show that after HSV DNA replication, IFI16 forms nuclear filamentous structures on DNA within a subset of nuclear replication compartments in HSV-1 ICP0-null mutant virus-infected human cells. The ability to form filaments in different cell types correlates with the efficiency of restriction, and the kinetics of filament formation and epigenetic changes are similar. Thus, both are consistent with the filamentous structures being involved in epigenetic silencing of viral progeny DNA. IFI16 filaments recruit other restriction factors, including PML, Sp100, and ATRX, to aid in the restriction. Although the filaments are only in a subset of the replication compartments, IFI16 reduces the levels of elongation-competent RNA polymerase II (Pol II) in all replication compartments. Therefore, we propose that IFI16 filaments with associated restriction factors that form in replication compartments constitute a "restrictosome" structure that signals in cis and trans to silence the progeny viral DNA throughout the infected cell nucleus. The IFI16 filamentous structure may constitute the first known nuclear supramolecular organizing center for signaling in the cell nucleus. IMPORTANCE Mammalian cells exhibit numerous strategies to recognize and contain viral infections. The best-characterized antiviral responses are those that are induced within the cytosol by receptors that activate interferon responses or shut down translation. Antiviral responses also occur in the nucleus, yet these intranuclear innate immune responses are poorly defined at the receptor-proximal level. In this study, we explored the ability of cells to restrict infection by assembling viral DNA into transcriptionally silent heterochromatin within the nucleus. We found that the IFI16 restriction factor forms filaments on DNA within infected cells. These filaments recruit antiviral restriction factors to prevent viral replication in various cell types. Mechanistically, IFI16 filaments inhibit the recruitment of RNA polymerase II to viral genes. We propose that IFI16 filaments with associated restriction factors constitute a "restrictosome" structure that can signal to other parts of the nucleus where foreign DNA is located that it should be silenced., (Copyright © 2019 Merkl and Knipe.)
- Published
- 2019
- Full Text
- View/download PDF
34. Generation of an Oncolytic Herpes Simplex Virus 1 Expressing Human MelanA.
- Author
-
Boscheinen JB, Thomann S, Knipe DM, DeLuca N, Schuler-Thurner B, Gross S, Dörrie J, Schaft N, Bach C, Rohrhofer A, Werner-Klein M, Schmidt B, and Schuster P
- Subjects
- Antigen-Presenting Cells immunology, Antigen-Presenting Cells metabolism, CD8-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes metabolism, Cell Line, Tumor, Fibroblasts metabolism, Gene Targeting, Genetic Engineering, Humans, Leukocytes, Mononuclear immunology, Leukocytes, Mononuclear metabolism, Melanoma, T-Lymphocytes immunology, T-Lymphocytes metabolism, Gene Expression, Genetic Vectors genetics, Herpesvirus 1, Human genetics, MART-1 Antigen genetics, Oncolytic Viruses genetics, Transgenes
- Abstract
Robust anti-tumor immunity requires innate as well as adaptive immune responses. We have shown that plasmacytoid dendritic cells develop killer cell-like activity in melanoma cell cocultures after exposure to the infectious but replication-deficient herpes simplex virus 1 (HSV-1) d 106S. To combine this innate effect with an enhanced adaptive immune response, the gene encoding human MelanA/MART-1 was inserted into HSV-1 d 106S via homologous recombination to increase direct expression of this tumor antigen. Infection of Vero cells using this recombinant virus confirmed MelanA expression by Western blotting, flow cytometry, and immunofluorescence. HSV-1 d 106S-MelanA induced expression of the transgene in fibroblast and melanoma cell lines not naturally expressing MelanA. Infection of a melanoma cell line with CRISPR-Cas9-mediated knockout of MelanA confirmed de novo expression of the transgene in the viral context. Dependent on MelanA expression, infected fibroblast and melanoma cell lines induced degranulation of HLA-matched MelanA-specific CD8
+ T cells, followed by killing of infected cells. To study infection of immune cells, we exposed peripheral blood mononuclear cells and in vitro -differentiated macrophages to the parental HSV-1 d 106S, resulting in expression of the transgene GFP in CD11c+ cells and macrophages. These data provide evidence that the application of MelanA-encoding HSV-1 d 106S could enhance adaptive immune responses and re-direct MelanA-specific CD8+ T cells to tumor lesions, which have escaped adaptive immune responses via downregulation of their tumor antigen. Hence, HSV-1 d 106S-MelanA harbors the potential to induce innate immune responses in conjunction with adaptive anti-tumor responses by CD8+ T cells, which should be evaluated in further studies.- Published
- 2019
- Full Text
- View/download PDF
35. CNBP controls IL-12 gene transcription and Th1 immunity.
- Author
-
Chen Y, Sharma S, Assis PA, Jiang Z, Elling R, Olive AJ, Hang S, Bernier J, Huh JR, Sassetti CM, Knipe DM, Gazzinelli RT, and Fitzgerald KA
- Subjects
- Animals, Interferon-gamma genetics, Interferon-gamma immunology, Interleukin-12 Subunit p40 genetics, Mice, Mice, Knockout, NF-kappa B genetics, NF-kappa B immunology, Proto-Oncogene Proteins c-rel genetics, Proto-Oncogene Proteins c-rel immunology, RNA-Binding Proteins genetics, Th1 Cells cytology, Immunity, Cellular, Interleukin-12 Subunit p40 immunology, RNA-Binding Proteins immunology, Th1 Cells immunology, Transcription, Genetic immunology
- Abstract
An inducible program of inflammatory gene expression is a hallmark of antimicrobial defenses. Recently, cellular nucleic acid-binding protein (CNBP) was identified as a regulator of nuclear factor-kappaB (NF-κB)-dependent proinflammatory cytokine gene expression. Here, we generated mice lacking CNBP and found that CNBP regulates a very restricted gene signature that includes IL-12β. CNBP resides in the cytosol of macrophages and translocates to the nucleus in response to diverse microbial pathogens and pathogen-derived products. Cnbp -deficient macrophages induced canonical NF-κB/Rel signaling normally but were impaired in their ability to control the activation of c-Rel, a key driver of IL-12β gene transcription. The nuclear translocation and DNA-binding activity of c-Rel required CNBP. Lastly, Cnbp -deficient mice were more susceptible to acute toxoplasmosis associated with reduced production of IL-12β, as well as a reduced T helper type 1 (Th1) cell IFN-γ response essential to controlling parasite replication. Collectively, these findings identify CNBP as important regulator of c-Rel-dependent IL-12β gene transcription and Th1 immunity., (© 2018 Chen et al.)
- Published
- 2018
- Full Text
- View/download PDF
36. ATRX promotes maintenance of herpes simplex virus heterochromatin during chromatin stress.
- Author
-
Cabral JM, Oh HS, and Knipe DM
- Subjects
- Cell Line, Cell Line, Tumor, DNA, Viral genetics, Fibroblasts virology, Gene Expression Regulation, Viral, Gene Knockout Techniques, HEK293 Cells, Herpesvirus 1, Human genetics, Herpesvirus 1, Human physiology, Histones metabolism, Host-Pathogen Interactions, Humans, Models, Genetic, Virus Latency genetics, X-linked Nuclear Protein genetics, DNA, Viral metabolism, Fibroblasts metabolism, Herpesvirus 1, Human metabolism, X-linked Nuclear Protein metabolism
- Abstract
The mechanisms by which mammalian cells recognize and epigenetically restrict viral DNA are not well defined. We used herpes simplex virus with bioorthogonally labeled genomes to detect host factors recruited to viral DNA shortly after its nuclear entry and found that the cellular IFI16, PML, and ATRX proteins colocalized with viral DNA by 15 min post infection. HSV-1 infection of ATRX-depleted fibroblasts resulted in elevated viral mRNA and accelerated viral DNA accumulation. Despite the early association of ATRX with vDNA, we found that initial viral heterochromatin formation is ATRX-independent. However, viral heterochromatin stability required ATRX from 4 to 8 hr post infection. Inhibition of transcription blocked viral chromatin loss in ATRX-knockout cells; thus, ATRX is uniquely required for heterochromatin maintenance during chromatin stress. These results argue that the initial formation and the subsequent maintenance of viral heterochromatin are separable mechanisms, a concept that likely extrapolates to host cell chromatin and viral latency., Competing Interests: JC, HO No competing interests declared, DK Reviewing editor, eLife, (© 2018, Cabral et al.)
- Published
- 2018
- Full Text
- View/download PDF
37. Mechanisms of Host IFI16, PML, and Daxx Protein Restriction of Herpes Simplex Virus 1 Replication.
- Author
-
Merkl PE, Orzalli MH, and Knipe DM
- Subjects
- Adaptor Proteins, Signal Transducing genetics, Cell Line, Co-Repressor Proteins, DNA Replication genetics, DNA Replication physiology, DNA, Viral biosynthesis, DNA, Viral genetics, Gene Expression Regulation, Viral, Genome, Viral genetics, HEK293 Cells, Herpesvirus 1, Human genetics, Herpesvirus 1, Human growth & development, Host-Pathogen Interactions, Humans, Molecular Chaperones, Nuclear Proteins genetics, Phosphoproteins genetics, Promyelocytic Leukemia Protein genetics, RNA Interference, RNA, Small Interfering genetics, Adaptor Proteins, Signal Transducing metabolism, Herpesvirus 1, Human physiology, Nuclear Proteins metabolism, Phosphoproteins metabolism, Promyelocytic Leukemia Protein metabolism, Virus Replication genetics
- Abstract
The initial events after DNA virus infection involve a race between epigenetic silencing of the incoming viral DNA by host cell factors and expression of viral genes. Several host gene products, including the nuclear domain 10 (ND10) components PML (promyelocytic leukemia) and Daxx (death domain-associated protein 6), as well as IFI16 (interferon-inducible protein 16), have been shown to restrict herpes simplex virus 1 (HSV-1) replication. Whether IFI16 and ND10 components work together or separately to restrict HSV-1 replication is not known. To determine the combinatorial effects of IFI16 and ND10 proteins on viral infection, we depleted Daxx or PML in primary human foreskin fibroblasts (HFFs) in the presence or absence of IFI16. Daxx or IFI16 depletion resulted in higher ICP0 mutant viral yields, and the effects were additive. Surprisingly, small interfering RNA (siRNA) depletion of PML in the HFF cells led to decreased ICP0-null virus replication, while short hairpin RNA (shRNA) depletion led to increased ICP0-null virus replication, arguing that different PML isoforms or PML-related proteins may have restrictive or proviral functions. In normal human cells, viral DNA replication increases expression of all classes of HSV-1 genes. We observed that IFI16 repressed transcription from both parental and progeny DNA genomes. Taken together, our results show that the mechanisms of action of IFI16 and ND10 proteins are independent, at least in part, and that IFI16 exerts restrictive effects on both input and replicated viral genomes. These results raise the potential for distinct mechanisms of action of IFI16 on parental and progeny viral DNA molecules. IMPORTANCE Many human DNA viruses transcribe their genomes and replicate in the nucleus of a host cell, where they exploit the host cell nuclear machinery for their own replication. Host factors attempt to restrict viral replication by blocking such events, and viruses have evolved mechanisms to neutralize the host restriction factors. In this study, we provide information about the mechanisms of action of three host cell factors that restrict replication of herpes simplex virus (HSV). We found that these factors function independently and that one acts to restrict viral transcription from parental and progeny viral DNA genomes. These results provide new information about how cells counter DNA virus replication in the nucleus and provide possible approaches to enhance the ability of human cells to resist HSV infection., (Copyright © 2018 American Society for Microbiology.)
- Published
- 2018
- Full Text
- View/download PDF
38. CCCTC-Binding Factor Acts as a Heterochromatin Barrier on Herpes Simplex Viral Latent Chromatin and Contributes to Poised Latent Infection.
- Author
-
Lee JS, Raja P, Pan D, Pesola JM, Coen DM, and Knipe DM
- Subjects
- Animals, Binding Sites, Chlorocebus aethiops, DNA, Viral genetics, Disease Models, Animal, HeLa Cells, Herpes Simplex pathology, Herpes Simplex virology, Humans, Mutation, Vero Cells, CCCTC-Binding Factor metabolism, Chromatin metabolism, DNA, Viral metabolism, Herpesvirus 1, Human physiology, Host-Pathogen Interactions, Virus Activation
- Abstract
Herpes simplex virus 1 (HSV-1) establishes latent infection in neurons via a variety of epigenetic mechanisms that silence its genome. The cellular CCCTC-binding factor (CTCF) functions as a mediator of transcriptional control and chromatin organization and has binding sites in the HSV-1 genome. We constructed an HSV-1 deletion mutant that lacked a pair of CTCF-binding sites ( CTRL2 ) within the latency-associated transcript ( LAT ) coding sequences and found that loss of these CTCF-binding sites did not alter lytic replication or levels of establishment of latent infection, but their deletion reduced the ability of the virus to reactivate from latent infection. We also observed increased heterochromatin modifications on viral chromatin over the LAT promoter and intron. We therefore propose that CTCF binding at the CTRL2 sites acts as a chromatin insulator to keep viral chromatin in a form that is poised for reactivation, a state which we call poised latency. IMPORTANCE Herpes simplex virus 1 (HSV-1) is a human pathogen that persists for the lifetime of the host as a result of its ability to establish latent infection within sensory neurons. The mechanism by which HSV-1 transitions from the lytic to latent infection program is largely unknown; however, HSV-1 is able to coopt cellular silencing mechanisms to facilitate the suppression of lytic gene expression. Here, we demonstrate that the cellular CCCTC-binding factor (CTCF)-binding site within the latency associated transcript ( LAT ) region is critical for the maintenance of a specific local chromatin structure. Additionally, loss of CTCF binding has detrimental effects on the ability to reactivate from latent infection. These results argue that CTCF plays a critical role in epigenetic regulation of viral gene expression to establish and/or maintain a form of latent infection that can reactivate efficiently., (Copyright © 2018 Lee et al.)
- Published
- 2018
- Full Text
- View/download PDF
39. Intramuscular delivery of replication-defective herpes simplex virus gives antigen expression in muscle syncytia and improved protection against pathogenic HSV-2 strains.
- Author
-
Diaz F, Gregory S, Nakashima H, Viapiano MS, and Knipe DM
- Subjects
- Animals, Disease Models, Animal, Injections, Intramuscular, Mice, Inbred BALB C, Treatment Outcome, Vaccines, Attenuated administration & dosage, Vaccines, Attenuated immunology, Herpes Genitalis prevention & control, Herpesvirus 2, Human immunology, Herpesvirus Vaccines administration & dosage, Herpesvirus Vaccines immunology
- Abstract
Herpes simplex virus 2 (HSV-2) is the leading cause of genital herpes and increases the risk of HIV infection, but there is no effective vaccine. A replication-defective HSV-2 mutant virus, dl5-29, is effective in animal models and has been in a phase I trial. Previous studies have shown that dl5-29 gives higher antibody responses and better protection when inoculated intramuscularly (IM) as compared with subcutaneously (SC). However, the basis for this effect has not been defined. We confirmed that IM inoculation of dl5-29 is more immunogenic and provides better protection than SC inoculation. IM inoculation of HSV-2 strains produced higher levels of a luciferase transgene than SC inoculation, as measured by intravital bioluminescence imaging. Intramuscular immunization also showed better protection against infection with a highly pathogenic African HSV-2, demonstrating that this single vaccine can be efficacious against HSV-2 strains from different geographic regions., (Copyright © 2017 Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
40. A digenic human immunodeficiency characterized by IFNAR1 and IFNGR2 mutations.
- Author
-
Hoyos-Bachiloglu R, Chou J, Sodroski CN, Beano A, Bainter W, Angelova M, Al Idrissi E, Habazi MK, Alghamdi HA, Almanjomi F, Al Shehri M, Elsidig N, Alaa Eldin M, Knipe DM, AlZahrani M, and Geha RS
- Subjects
- Bacteremia genetics, Bacteremia immunology, Bacteremia microbiology, Cytomegalovirus immunology, Cytomegalovirus Infections genetics, Cytomegalovirus Infections immunology, Female, Fibroblasts immunology, Fibroblasts microbiology, Fibroblasts virology, Genetic Diseases, Inborn microbiology, Genetic Diseases, Inborn virology, Humans, Immunologic Deficiency Syndromes microbiology, Immunologic Deficiency Syndromes virology, Male, Mycobacterium Infections, Nontuberculous genetics, Mycobacterium Infections, Nontuberculous immunology, Mycobacterium abscessus immunology, Phosphorylation genetics, Phosphorylation immunology, STAT1 Transcription Factor genetics, STAT1 Transcription Factor immunology, STAT2 Transcription Factor genetics, STAT2 Transcription Factor immunology, Streptococcal Infections genetics, Streptococcal Infections immunology, Viremia genetics, Viremia immunology, Viremia virology, Viridans Streptococci immunology, Genetic Diseases, Inborn genetics, Genetic Diseases, Inborn immunology, Immunologic Deficiency Syndromes genetics, Immunologic Deficiency Syndromes immunology, Mutation, Receptor, Interferon alpha-beta genetics, Receptor, Interferon alpha-beta immunology, Receptors, Interferon genetics, Receptors, Interferon immunology
- Abstract
Primary immunodeficiencies are often monogenic disorders characterized by vulnerability to specific infectious pathogens. Here, we performed whole-exome sequencing of a patient with disseminated Mycobacterium abscessus, Streptococcus viridians bacteremia, and cytomegalovirus (CMV) viremia and identified mutations in 2 genes that regulate distinct IFN pathways. The patient had a homozygous frameshift deletion in IFNGR2, which encodes the signal transducing chain of the IFN-γ receptor, that resulted in minimal protein expression and abolished downstream signaling. The patient also harbored a homozygous deletion in IFNAR1 (IFNAR1*557Gluext*46), which encodes the IFN-α receptor signaling subunit. The IFNAR1*557Gluext*46 resulted in replacement of the stop codon with 46 additional codons at the C-terminus. The level of IFNAR1*557Gluext*46 mutant protein expressed in patient fibroblasts was comparable to levels of WT IFNAR1 in control fibroblasts. IFN-α-induced signaling was impaired in the patient fibroblasts, as evidenced by decreased STAT1/STAT2 phosphorylation, nuclear translocation of STAT1, and expression of IFN-α-stimulated genes critical for CMV immunity. Pretreatment with IFN-α failed to suppress CMV protein expression in patient fibroblasts, whereas expression of WT IFNAR1 restored IFN-α-mediated suppression of CMV. This study identifies a human IFNAR1 mutation and describes a digenic immunodeficiency specific to type I and type II IFNs.
- Published
- 2017
- Full Text
- View/download PDF
41. Role of Herpes Simplex Virus 1 γ34.5 in the Regulation of IRF3 Signaling.
- Author
-
Manivanh R, Mehrbach J, Knipe DM, and Leib DA
- Subjects
- Animals, Beclin-1 metabolism, Chlorocebus aethiops, Fibroblasts drug effects, Fibroblasts virology, Herpesvirus 1, Human genetics, Herpesvirus 1, Human pathogenicity, Humans, Immunity, Innate, Interferon-beta pharmacology, Interferons metabolism, Mice, Phosphorylation, Protein Serine-Threonine Kinases metabolism, Vero Cells, Viral Proteins chemistry, Viral Proteins genetics, Virus Replication genetics, Herpesvirus 1, Human metabolism, Host-Pathogen Interactions, Interferon Regulatory Factor-3 metabolism, Signal Transduction, Viral Proteins metabolism
- Abstract
During viral infection, pattern recognition receptors (PRRs) and their associated adaptors recruit TANK-binding kinase 1 (TBK1) to activate interferon regulatory factor 3 (IRF3), resulting in production of type I interferons (IFNs). ICP0 and ICP34.5 are among the proteins encoded by herpes simplex virus 1 (HSV-1) that modulate type I IFN signaling. We constructed a recombinant virus (ΔXX) that lacks amino acids 87 to 106, a portion of the previously described TBK1-binding domain of the γ34.5 gene (D. Verpooten, Y. Ma, S. Hou, Z. Yan, and B. He, J Biol Chem 284:1097-1105, 2009, https://doi.org/10.1074/JBC.M805905200). These 20 residues are outside the γ34.5 beclin1-binding domain (BBD) that interacts with beclin1 and regulates autophagy. Unexpectedly, ΔXX showed no deficit in replication in vivo in a variety of tissues and showed virulence comparable to that of wild-type and marker-rescued viruses following intracerebral infection. ΔXX was fully capable of mediating the dephosphorylation of eIF2α, and the virus was capable of controlling the phosphorylation of IRF3. In contrast, a null mutant in γ34.5 failed to control IRF3 phosphorylation due to an inability of the mutant to sustain expression of ICP0. Our data show that while γ34.5 regulates IRF3 phosphorylation, the TBK1-binding domain itself has no impact on IRF3 phosphorylation or on replication and pathogenesis in mice. IMPORTANCE Interferons (IFNs) are potent activators of a variety of host responses that serve to control virus infections. The Herpesviridae have evolved countermeasures to IFN responses. Herpes simplex virus 1 (HSV-1) encodes the multifunctional neurovirulence protein ICP34.5. In this study, we investigated the biological relevance of the interaction between ICP34.5 and TANK-binding kinase 1 (TBK1), an activator of IFN responses. Here, we establish that although ICP34.5 binds TBK1 under certain conditions through a TBK1-binding domain (TBD), there was no direct impact of the TBD on viral replication or virulence in mice. Furthermore, we showed that activation of IRF3, a substrate of TBK1, was independent of the TBD. Instead, we provided evidence that the ability of ICP34.5 to control IRF3 activation is through its ability to reverse translational shutoff and sustain the expression of other IFN inhibitors encoded by the virus. This work provides new insights into the immunomodulatory functions of ICP34.5., (Copyright © 2017 American Society for Microbiology.)
- Published
- 2017
- Full Text
- View/download PDF
42. Keratinocytes produce IL-17c to protect peripheral nervous systems during human HSV-2 reactivation.
- Author
-
Peng T, Chanthaphavong RS, Sun S, Trigilio JA, Phasouk K, Jin L, Layton ED, Li AZ, Correnti CE, De van der Schueren W, Vazquez J, O'Day DR, Glass IA, Knipe DM, Wald A, Corey L, and Zhu J
- Subjects
- Animals, Herpes Genitalis virology, Humans, Keratinocytes virology, Neurites physiology, Neuroblastoma physiopathology, Peripheral Nervous System physiopathology, Virus Activation physiology, Herpes Genitalis physiopathology, Herpesvirus 2, Human physiology, Interleukin-17 physiology, Keratinocytes metabolism, Peripheral Nervous System virology
- Abstract
Despite frequent herpes simplex virus (HSV) reactivation, peripheral nerve destruction and sensory anesthesia are rare. We discovered that skin biopsies obtained during asymptomatic human HSV-2 reactivation exhibit a higher density of nerve fibers relative to biopsies during virological and clinical quiescence. We evaluated the effects of HSV infection on keratinocytes, the initial target of HSV replication, to better understand this observation. Keratinocytes produced IL-17c during HSV-2 reactivation, and IL-17RE, an IL-17c-specific receptor, was expressed on nerve fibers in human skin and sensory neurons in dorsal root ganglia. In ex vivo experiments, exogenous human IL-17c provided directional guidance and promoted neurite growth and branching in microfluidic devices. Exogenous murine IL-17c pretreatment reduced apoptosis in HSV-2-infected primary neurons. These results suggest that IL-17c is a neurotrophic cytokine that protects peripheral nerve systems during HSV reactivation. This mechanism could explain the lack of nerve damage from recurrent HSV infection and may provide insight to understanding and treating sensory peripheral neuropathies., (© 2017 Peng et al.)
- Published
- 2017
- Full Text
- View/download PDF
43. Viral gene products actively promote latent infection by epigenetic silencing mechanisms.
- Author
-
Knipe DM, Raja P, and Lee J
- Subjects
- Gene Silencing, HIV genetics, Herpesviridae genetics, Host-Pathogen Interactions, Viral Proteins genetics, Epigenesis, Genetic, Gene Expression Regulation, Viral, HIV physiology, Herpesviridae physiology, Viral Proteins biosynthesis, Virus Latency
- Abstract
Many viruses undergo an acute infection in the host organism and then are cleared by the ensuing host immune response, but other viruses establish a persistent infection involving a latent infection or a chronic infection. Latent infection by the herpesviruses or human immunodeficiency virus involves epigenetic silencing of the DNA genome or proviral genome, respectively. Latent infection was previously thought to be a default pathway resulting from infection of a nonpermissive cell, but recent studies have shown that viral gene products can promote epigenetic silencing and latent infection. This review will summarize the viral gene products that have been shown to promote epigenetic silencing of the genomes and their potential for therapeutics to target these viral gene products and disrupt or lock in latent infection., (Copyright © 2017 Elsevier B.V. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
44. Innate Immune Mechanisms and Herpes Simplex Virus Infection and Disease.
- Author
-
Kurt-Jones EA, Orzalli MH, and Knipe DM
- Subjects
- Animals, Host-Pathogen Interactions immunology, Humans, Immune Evasion, Herpes Simplex immunology, Herpes Simplex virology, Herpesvirus 1, Human physiology, Immunity, Innate
- Abstract
Innate immune responses play a major role in the control of herpes simplex virus (HSV) infections, and a multiplicity of mechanisms have emerged as a result of human evolution to sense and respond to HSV infections. HSV in turn has evolved a number of ways to evade immune detection and to blunt human innate immune responses. In this review, we summarize the major host innate immune mechanisms and the HSV evasion mechanisms that have evolved. We further discuss how disease can result if this equilibrium between virus and host response is disrupted.
- Published
- 2017
- Full Text
- View/download PDF
45. Production of immunogenic West Nile virus-like particles using a herpes simplex virus 1 recombinant vector.
- Author
-
Taylor TJ, Diaz F, Colgrove RC, Bernard KA, DeLuca NA, Whelan SPJ, and Knipe DM
- Subjects
- Amino Acid Sequence, Animals, Antibodies, Neutralizing immunology, Antibodies, Viral immunology, Capsid immunology, Capsid ultrastructure, Cell Line, Gene Order, Humans, Immunization, Mice, Vaccines, Virus-Like Particle administration & dosage, Vaccines, Virus-Like Particle immunology, Vaccines, Virus-Like Particle ultrastructure, Viral Structural Proteins chemistry, Viral Structural Proteins immunology, West Nile virus immunology, Genetic Vectors genetics, Herpesvirus 1, Human genetics, Vaccines, Virus-Like Particle genetics, Viral Structural Proteins genetics, West Nile virus genetics
- Abstract
West Nile virus (WNV) is a flavivirus that swept rapidly across North America in 1999, declined in prevalence, and then resurged in 2012. To date, no vaccine is available to prevent infection in the human population. Herpes simplex virus (HSV) replication-defective vaccine vectors induce a durable immunity characterized by strong antibody and CD8(+) T cell responses even in HSV-immune animals. In this study, a WNV protein expression cassette was optimized for virus-like particle (VLP) production in transfection studies, and the cassette was recombined into an HSV-1 d106-WNV virus vector, which produced extracellular VLPs, as confirmed by immunoelectron microscopy. Immunization of mice with the d106-WNV recombinant vector elicited a specific anti-WNV IgG response. This study highlights the flavivirus coding sequences needed for efficient assembly of virus-like particles. This information will facilitate generation of additional vaccine vectors against other flaviviruses including the recently emerged Zika virus., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
46. Relative Contributions of Herpes Simplex Virus 1 ICP0 and vhs to Loss of Cellular IFI16 Vary in Different Human Cell Types.
- Author
-
Orzalli MH, Broekema NM, and Knipe DM
- Subjects
- Cells, Cultured, Humans, Down-Regulation, Herpesvirus 1, Human physiology, Host-Pathogen Interactions, Immediate-Early Proteins metabolism, Nuclear Proteins metabolism, Phosphoproteins metabolism, Ribonucleases metabolism, Ubiquitin-Protein Ligases metabolism, Viral Proteins metabolism
- Abstract
Unlabelled: The herpes simplex virus 1 (HSV-1) ICP0 protein is an E3 ubiquitin ligase that promotes the degradation of several host cell proteins. Most studies have found that ICP0 promotes the loss of IFI16 in infected cells, but one study reported that ICP0 was not necessary or sufficient for loss of IFI16 in a tumor-derived cell line. Therefore, in this study, we examined the requirement for ICP0 in promoting the loss of IFI16 in several normal and tumor-derived cell lines. HSV-1 infection resulted in an observable decrease of IFI16 protein levels in normal human foreskin fibroblasts (HFFs), normal oral keratinocytes (NOKs), and HeLa cells but not in U2OS cells. During infection with an ICP0-null virus, we observed a reduced loss of IFI16 in HFFs and NOKs but not in HeLa cells. Ectopic expression of ICP0 from a transfected plasmid was sufficient to promote the loss of IFI16 in HFFs and NOKs. In the absence of ICP0, we observed a delayed reduction of IFI16 protein that correlated with a reduction in the steady-state levels of IFI16 mRNA. In addition, we show that the ICP0-independent loss of IFI16 in HeLa cells is dependent in part on the activity of the viral virion host shutoff (vhs) tegument protein. Together, these results demonstrate that HSV-1 promotes the loss of IFI16 through at least two mechanisms: (i) by ICP0-dependent degradation of IFI16 and (ii) by vhs-dependent turnover of IFI16 mRNA. In addition, this study highlights a potential intrinsic difference between normal and tumor-derived cells for the activities of IFI16 and HSV-1 ICP0., Importance: HSV-1 is a ubiquitous virus that establishes a lifetime persistent infection in humans. The relative success of HSV-1 as a pathogen is, in part, dependent on the expression of viral proteins that counteract host intrinsic defense mechanisms and that modulate immune responses during viral infection. In this study, we examined the relative roles of two viral gene products for the ability to promote loss of the antiviral IFI16 DNA sensor. We demonstrate that the viral immediate early ICP0 protein plays a dominant role in the loss of IFI16 in normal, but not tumor-derived, human cell lines. In contrast, viral vhs-mediated loss of IFI16 by mRNA destabilization is revealed to be dominant in tumor-derived cells in which ICP0 is nonfunctional. Together, these results contribute to our understanding of how HSV-1 modulates IFI16 protein levels and highlight cell-type-dependent differences between normal and tumor-derived cells., (Copyright © 2016, American Society for Microbiology. All Rights Reserved.)
- Published
- 2016
- Full Text
- View/download PDF
47. HSV-1 ICP27 targets the TBK1-activated STING signalsome to inhibit virus-induced type I IFN expression.
- Author
-
Christensen MH, Jensen SB, Miettinen JJ, Luecke S, Prabakaran T, Reinert LS, Mettenleiter T, Chen ZJ, Knipe DM, Sandri-Goldin RM, Enquist LW, Hartmann R, Mogensen TH, Rice SA, Nyman TA, Matikainen S, and Paludan SR
- Subjects
- Cells, Cultured, Host-Pathogen Interactions, Humans, Protein Interaction Mapping, Herpesvirus 1, Human pathogenicity, Immediate-Early Proteins metabolism, Immune Evasion, Interferon Type I antagonists & inhibitors, Macrophages immunology, Membrane Proteins metabolism, Protein Serine-Threonine Kinases metabolism
- Abstract
Herpes simplex virus (HSV) 1 stimulates type I IFN expression through the cGAS-STING-TBK1 signaling axis. Macrophages have recently been proposed to be an essential source of IFN during viral infection. However, it is not known how HSV-1 inhibits IFN expression in this cell type. Here, we show that HSV-1 inhibits type I IFN induction through the cGAS-STING-TBK1 pathway in human macrophages, in a manner dependent on the conserved herpesvirus protein ICP27. This viral protein was expressed de novo in macrophages with early nuclear localization followed by later translocation to the cytoplasm where ICP27 prevented activation of IRF3. ICP27 interacted with TBK1 and STING in a manner that was dependent on TBK1 activity and the RGG motif in ICP27. Thus, HSV-1 inhibits expression of type I IFN in human macrophages through ICP27-dependent targeting of the TBK1-activated STING signalsome., (© 2016 The Authors.)
- Published
- 2016
- Full Text
- View/download PDF
48. A Herpesviral Lytic Protein Regulates the Structure of Latent Viral Chromatin.
- Author
-
Raja P, Lee JS, Pan D, Pesola JM, Coen DM, and Knipe DM
- Subjects
- Animals, Epigenesis, Genetic, Herpes Simplex virology, Heterochromatin, Mice, Mutation, Promoter Regions, Genetic, Sensory Receptor Cells virology, Viral Proteins metabolism, Chromatin chemistry, Gene Expression Regulation, Viral, Herpesvirus 1, Human genetics, Herpesvirus 1, Human physiology, Immediate-Early Proteins genetics, Immediate-Early Proteins metabolism, Ubiquitin-Protein Ligases genetics, Ubiquitin-Protein Ligases metabolism, Virus Latency genetics
- Abstract
Unlabelled: Latent infections by viruses usually involve minimizing viral protein expression so that the host immune system cannot recognize the infected cell through the viral peptides presented on its cell surface. Herpes simplex virus (HSV), for example, is thought to express noncoding RNAs such as latency-associated transcripts (LATs) and microRNAs (miRNAs) as the only abundant viral gene products during latent infection. Here we describe analysis of HSV-1 mutant viruses, providing strong genetic evidence that HSV-infected cell protein 0 (ICP0) is expressed during establishment and/or maintenance of latent infection in murine sensory neurons in vivo Studies of an ICP0 nonsense mutant virus showed that ICP0 promotes heterochromatin and latent and lytic transcription, arguing that ICP0 is expressed and functional. We propose that ICP0 promotes transcription of LATs during establishment or maintenance of HSV latent infection, much as it promotes lytic gene transcription. This report introduces the new concept that a lytic viral protein can be expressed during latent infection and can serve dual roles to regulate viral chromatin to optimize latent infection in addition to its role in epigenetic regulation during lytic infection. An additional implication of the results is that ICP0 might serve as a target for an antiviral therapeutic acting on lytic and latent infections., Importance: Latent infection by viruses usually involves minimizing viral protein synthesis so that the host immune system cannot recognize the infected cells and eliminate them. Herpes simplex virus has been thought to express only noncoding RNAs as abundant gene products during latency. In this study, we found genetic evidence that an HSV lytic protein is functional during latent infection, and this protein may provide a new target for antivirals that target both lytic and latent infections., (Copyright © 2016 Raja et al.)
- Published
- 2016
- Full Text
- View/download PDF
49. Protection from genital herpes disease, seroconversion and latent infection in a non-lethal murine genital infection model by immunization with an HSV-2 replication-defective mutant virus.
- Author
-
Diaz FM and Knipe DM
- Subjects
- Animals, Antibodies, Viral blood, Disease Models, Animal, Enzyme-Linked Immunosorbent Assay, Female, Herpes Genitalis virology, Herpesvirus 2, Human physiology, Herpesvirus Vaccines administration & dosage, Mice, Inbred BALB C, Real-Time Polymerase Chain Reaction, Seroconversion, Treatment Outcome, Vaccines, Attenuated administration & dosage, Vaccines, Attenuated immunology, Viral Proteins immunology, Herpes Genitalis immunology, Herpes Genitalis prevention & control, Herpesvirus 2, Human immunology, Herpesvirus Vaccines immunology, Virus Latency
- Abstract
Viral vaccines have traditionally protected against disease, but for viruses that establish latent infection, it is desirable for the vaccine to reduce infection to reduce latent infection and reactivation. While seroconversion has been used in clinical trials of herpes simplex virus (HSV) vaccines to measure protection from infection, this has not been modeled in animal infection systems. To measure the ability of a genital herpes vaccine candidate to protect against various aspects of infection, we established a non-lethal murine model of genital HSV-2 infection, an ELISA assay to measure antibodies specific for infected cell protein 8 (ICP8), and a very sensitive qPCR assay. Using these assays, we observed that immunization with HSV-2 dl5-29 virus reduced disease, viral shedding, seroconversion, and latent infection by the HSV-2 challenge virus. Therefore, it may be feasible to obtain protection against genital disease, seroconversion and latent infection by immunization, even if sterilizing immunity is not achieved., (Copyright © 2015 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
50. Herpesviral ICP0 Protein Promotes Two Waves of Heterochromatin Removal on an Early Viral Promoter during Lytic Infection.
- Author
-
Lee JS, Raja P, and Knipe DM
- Subjects
- Cells, Cultured, Fibroblasts virology, Humans, Heterochromatin metabolism, Host-Pathogen Interactions, Immediate-Early Proteins metabolism, Promoter Regions, Genetic, Ubiquitin-Protein Ligases metabolism
- Abstract
Unlabelled: Herpesviruses must contend with host cell epigenetic silencing responses acting on their genomes upon entry into the host cell nucleus. In this study, we confirmed that unchromatinized herpes simplex virus 1 (HSV-1) genomes enter primary human foreskin fibroblasts and are rapidly subjected to assembly of nucleosomes and association with repressive heterochromatin modifications such as histone 3 (H3) lysine 9-trimethylation (H3K9me3) and lysine 27-trimethylation (H3K27me3) during the first 1 to 2 h postinfection. Kinetic analysis of the modulation of nucleosomes and heterochromatin modifications over the course of lytic infection demonstrates a progressive removal that coincided with initiation of viral gene expression. We obtained evidence for three phases of heterochromatin removal from an early gene promoter: an initial removal of histones and heterochromatin not dependent on ICP0, a second ICP0-dependent round of removal of H3K9me3 that is independent of viral DNA synthesis, and a third phase of H3K27me3 removal that is dependent on ICP0 and viral DNA synthesis. The presence of ICP0 in transfected cells is also sufficient to promote removal of histones and H3K9me3 modifications of cotransfected genes. Overall, these results show that ICP0 promotes histone removal, a reduction of H3K9me3 modifications, and a later indirect reduction of H3K27me3 modifications following viral early gene expression and DNA synthesis. Therefore, HSV ICP0 promotes the reversal of host epigenetic silencing mechanisms by several mechanisms., Importance: The human pathogen herpes simplex virus (HSV) has evolved multiple strategies to counteract host-mediated epigenetic silencing during productive infection. However, the mechanisms by which viral and cellular effectors contribute to these processes are not well defined. The results from this study demonstrate that HSV counteracts host epigenetic repression in a dynamic stepwise process to remove histone 3 (H3) and subsequently target specific heterochromatin modifications in two distinct waves. This provides the first evidence of a stepwise reversal of host epigenetic silencing by viral proteins. This work also suggests that targets capable of disrupting the kinetics of epigenetic regulation could serve as potential antiviral therapeutic agents., (Copyright © 2016 Lee et al.)
- Published
- 2016
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.