Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Cancer Immunol Res ; 11(2): 145-149, 2023 02 03.
Article in English | MEDLINE | ID: mdl-36734102

ABSTRACT

From September 28 to October 1, 2022, in New York City, the Sixth International Cancer Immunotherapy Conference was hosted jointly by the Cancer Research Institute (CRI), the American Association for Cancer Research, and the European Network for Cancer Immunotherapy. The four-day event covered the latest advances in cancer immunology and immunotherapy.


Subject(s)
Neoplasms , Humans , Neoplasms/therapy , Immunotherapy
2.
Virology ; 518: 221-231, 2018 05.
Article in English | MEDLINE | ID: mdl-29547809

ABSTRACT

During viral replication in the nucleus, the DNA genomes of adenoviruses are accessible to cellular DNA-binding proteins. Human adenovirus type 5 (Ad5) targets the cellular Mre11-Rad50-Nbs1 complex (MRN) to evade detection by the DNA damage response (DDR). Ad5 mutants that cannot target MRN have reduced viral propagation. Previous studies showed that diverse adenovirus serotypes interact differently with MRN. While these studies revealed diverse MRN interactions among serotypes, it remains unclear how these differences influence viral replication. Here, we examined effects of the DDR on several adenovirus serotypes. We demonstrate that wild-type Ad9 and Ad12 do not overcome MRN impairment. We also examined viral proteins involved in targeting MRN and found that unlike Ad5-E4orf3, expression of Ad9-E4orf3 is not sufficient for MRN mislocalization observed during infection. We conclude that adenovirus serotypes target MRN in distinct ways, and the MRN complex can impair DNA replication of wild-type viruses across the adenovirus family.


Subject(s)
Adenoviruses, Human/immunology , Adenoviruses, Human/physiology , Cell Cycle Proteins/metabolism , DNA Repair Enzymes/metabolism , DNA-Binding Proteins/metabolism , Host-Pathogen Interactions , MRE11 Homologue Protein/metabolism , Nuclear Proteins/metabolism , Virus Replication , Acid Anhydride Hydrolases , Cell Line , Humans , Serogroup
3.
Mol Cell Proteomics ; 16(12): 2079-2097, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28972080

ABSTRACT

Viral DNA genomes replicating in cells encounter a myriad of host factors that facilitate or hinder viral replication. Viral proteins expressed early during infection modulate host factors interacting with viral genomes, recruiting proteins to promote viral replication, and limiting access to antiviral repressors. Although some host factors manipulated by viruses have been identified, we have limited knowledge of pathways exploited during infection and how these differ between viruses. To identify cellular processes manipulated during viral replication, we defined proteomes associated with viral genomes during infection with adenovirus, herpes simplex virus and vaccinia virus. We compared enrichment of host factors between virus proteomes and confirmed association with viral genomes and replication compartments. Using adenovirus as an illustrative example, we uncovered host factors deactivated by early viral proteins, and identified a subgroup of nucleolar proteins that aid virus replication. Our data sets provide valuable resources of virus-host interactions that affect proteins on viral genomes.


Subject(s)
Dependovirus/physiology , Proteome/metabolism , Simplexvirus/physiology , Vaccinia virus/physiology , Viral Proteins/metabolism , Virus Diseases/metabolism , A549 Cells , Cell Line, Tumor , DNA Replication , Genome, Viral , HeLa Cells , Host-Pathogen Interactions , Humans , Protein Interaction Maps , Proteomics/methods , Virus Replication
4.
Philos Trans R Soc Lond B Biol Sci ; 372(1732)2017 Oct 19.
Article in English | MEDLINE | ID: mdl-28893936

ABSTRACT

Viruses regulate cellular processes to facilitate viral replication. Manipulation of nuclear proteins and pathways by nuclear replicating viruses often causes cellular genome instability that contributes to transformation. The cellular DNA damage response (DDR) safeguards the host to maintain genome integrity, but DNA tumour viruses can manipulate the DDR to promote viral propagation. In this review, we describe the interactions of DNA tumour viruses with the phosphatidylinositol 3-kinase-like protein kinase (PIKK) pathways, which are central regulatory arms of the DDR. We review how signalling through the ataxia telangiectasia mutated (ATM), ataxia telangiectasia and Rad3 related (ATR), and DNA-dependent protein kinases (DNA-PK) influences viral life cycles, and how their manipulation by viral proteins may contribute to tumour formation.This article is part of the themed issue 'Human oncogenic viruses'.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/physiology , Carcinogenesis/genetics , DNA Tumor Viruses/physiology , DNA-Activated Protein Kinase/physiology , Signal Transduction , Viral Proteins/metabolism , DNA Damage , DNA Repair , Humans , Protein Kinases
5.
J Virol ; 91(20)2017 Oct 15.
Article in English | MEDLINE | ID: mdl-28794020

ABSTRACT

Viral manipulation of cellular proteins allows viruses to suppress host defenses and generate infectious progeny. Due to the linear double-stranded DNA nature of the adenovirus genome, the cellular DNA damage response (DDR) is considered a barrier to successful infection. The adenovirus genome is packaged with protein VII, a virally encoded histone-like core protein that is suggested to protect incoming viral genomes from detection by the cellular DNA damage machinery. We showed that protein VII localizes to host chromatin during infection, leading us to hypothesize that protein VII may affect DNA damage responses on the cellular genome. Here we show that protein VII at cellular chromatin results in a significant decrease in accumulation of phosphorylated H2AX (γH2AX) following irradiation, indicating that protein VII inhibits DDR signaling. The oncoprotein SET was recently suggested to modulate the DDR by affecting access of repair proteins to chromatin. Since protein VII binds SET, we investigated a role for SET in DDR inhibition by protein VII. We show that knockdown of SET partially rescues the protein VII-induced decrease in γH2AX accumulation on the host genome, suggesting that SET is required for inhibition. Finally, we show that knockdown of SET also allows ATM to localize to incoming viral genomes bound by protein VII during infection with a mutant lacking early region E4. Together, our data suggest that the protein VII-SET interaction contributes to DDR evasion by adenovirus. Our results provide an additional example of a strategy used by adenovirus to abrogate the host DDR and show how viruses can modify cellular processes through manipulation of host chromatin.IMPORTANCE The DNA damage response (DDR) is a cellular network that is crucial for maintaining genome integrity. DNA viruses replicating in the nucleus challenge the resident genome and must overcome cellular responses, including the DDR. Adenoviruses are prevalent human pathogens that can cause a multitude of diseases, such as respiratory infections and conjunctivitis. Here we describe how a small adenovirus core protein that localizes to host chromatin during infection can globally downregulate the DDR. Our study focuses on key players in the damage signaling pathway and highlights how viral manipulation of chromatin may influence access of DDR proteins to the host genome.

6.
Cell Host Microbe ; 20(2): 178-88, 2016 Aug 10.
Article in English | MEDLINE | ID: mdl-27512903

ABSTRACT

Humans occasionally transmit herpes simplex virus 1 (HSV-1) to captive primates, who reciprocally harbor alphaherpesviruses poised for zoonotic transmission to humans. To understand the basis for the species-specific restriction of HSV-1 in primates, we simulated what might happen during the cross-species transmission of HSV-1 and found that the DNA repair protein Nbs1 from only some primate species is able to promote HSV-1 infection. The Nbs1 homologs that promote HSV-1 infection also interact with the HSV-1 ICP0 protein. ICP0 interaction mapped to a region of structural disorder in the Nbs1 protein. Chimeras reversing patterns of disorder in Nbs1 reversed titers of HSV-1 produced in the cell. By extending this analysis to 1,237 virus-interacting mammalian proteins, we show that proteins that interact with viruses are highly enriched in disorder, suggesting that viruses commonly interact with host proteins through intrinsically disordered domains.


Subject(s)
Cell Cycle Proteins/metabolism , DNA Repair Enzymes/metabolism , Herpesvirus 1, Human/immunology , Herpesvirus 1, Human/physiology , Host-Pathogen Interactions , Immediate-Early Proteins/metabolism , Nuclear Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Virus Replication , Animals , Cell Cycle Proteins/genetics , Cell Line , DNA Repair Enzymes/genetics , Humans , Nuclear Proteins/genetics , Primates , Protein Binding , Protein Interaction Domains and Motifs , Protein Interaction Mapping , Sequence Homology, Amino Acid , Viral Load
7.
Nature ; 535(7610): 173-7, 2016 07 07.
Article in English | MEDLINE | ID: mdl-27362237

ABSTRACT

Viral proteins mimic host protein structure and function to redirect cellular processes and subvert innate defenses. Small basic proteins compact and regulate both viral and cellular DNA genomes. Nucleosomes are the repeating units of cellular chromatin and play an important part in innate immune responses. Viral-encoded core basic proteins compact viral genomes, but their impact on host chromatin structure and function remains unexplored. Adenoviruses encode a highly basic protein called protein VII that resembles cellular histones. Although protein VII binds viral DNA and is incorporated with viral genomes into virus particles, it is unknown whether protein VII affects cellular chromatin. Here we show that protein VII alters cellular chromatin, leading us to hypothesize that this has an impact on antiviral responses during adenovirus infection in human cells. We find that protein VII forms complexes with nucleosomes and limits DNA accessibility. We identified post-translational modifications on protein VII that are responsible for chromatin localization. Furthermore, proteomic analysis demonstrated that protein VII is sufficient to alter the protein composition of host chromatin. We found that protein VII is necessary and sufficient for retention in the chromatin of members of the high-mobility-group protein B family (HMGB1, HMGB2 and HMGB3). HMGB1 is actively released in response to inflammatory stimuli and functions as a danger signal to activate immune responses. We showed that protein VII can directly bind HMGB1 in vitro and further demonstrated that protein VII expression in mouse lungs is sufficient to decrease inflammation-induced HMGB1 content and neutrophil recruitment in the bronchoalveolar lavage fluid. Together, our in vitro and in vivo results show that protein VII sequesters HMGB1 and can prevent its release. This study uncovers a viral strategy in which nucleosome binding is exploited to control extracellular immune signaling.


Subject(s)
Adenoviridae/chemistry , Immunity, Innate , Nucleosomes/metabolism , Viral Core Proteins/metabolism , Alarmins/metabolism , Animals , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Cell Line , Chromatin Assembly and Disassembly/drug effects , HMGB1 Protein/metabolism , Histones/metabolism , Humans , Immunity, Innate/drug effects , Inflammation/immunology , Inflammation/metabolism , Lung/immunology , Lung/metabolism , Male , Mice , Neutrophil Infiltration/drug effects , Neutrophil Infiltration/immunology , Nucleosomes/chemistry , Nucleosomes/drug effects , Nucleosomes/genetics , Protein Binding , Protein Processing, Post-Translational , Proteomics , Viral Core Proteins/chemistry , Viral Core Proteins/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...