Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.192
Filter
1.
Retrovirology ; 21(1): 12, 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38886829

ABSTRACT

An essential regulatory hub for retroviral replication events, the 5' untranslated region (UTR) encodes an ensemble of cis-acting replication elements that overlap in a logical manner to carry out divergent RNA activities in cells and in virions. The primer binding site (PBS) and primer activation sequence initiate the reverse transcription process in virions, yet overlap with structural elements that regulate expression of the complex viral proteome. PBS-segment also encompasses the attachment site for Integrase to cut and paste the 3' long terminal repeat into the host chromosome to form the provirus and purine residues necessary to execute the precise stoichiometry of genome-length transcripts and spliced viral RNAs. Recent genetic mapping, cofactor affinity experiments, NMR and SAXS have elucidated that the HIV-1 PBS-segment folds into a three-way junction structure. The three-way junction structure is recognized by the host's nuclear RNA helicase A/DHX9 (RHA). RHA tethers host trimethyl guanosine synthase 1 to the Rev/Rev responsive element (RRE)-containing RNAs for m7-guanosine Cap hyper methylation that bolsters virion infectivity significantly. The HIV-1 trimethylated (TMG) Cap licenses specialized translation of virion proteins under conditions that repress translation of the regulatory proteins. Clearly host-adaption and RNA shapeshifting comprise the fundamental basis for PBS-segment orchestrating both reverse transcription of virion RNA and the nuclear modification of m7G-Cap for biphasic translation of the complex viral proteome. These recent observations, which have exposed even greater complexity of retroviral RNA biology than previously established, are the impetus for this article. Basic research to fully comprehend the marriage of PBS-segment structures and host RNA binding proteins that carry out retroviral early and late replication events is likely to expose an immutable virus-specific therapeutic target to attenuate retrovirus proliferation.


Subject(s)
5' Untranslated Regions , HIV-1 , RNA, Viral , Virus Replication , RNA, Viral/genetics , RNA, Viral/metabolism , Humans , HIV-1/physiology , HIV-1/genetics , Binding Sites , Gene Expression Regulation, Viral , Reverse Transcription , Retroviridae/physiology , Retroviridae/genetics
2.
Viruses ; 16(6)2024 Jun 08.
Article in English | MEDLINE | ID: mdl-38932225

ABSTRACT

The innate immune system, particularly the interferon (IFN) system, constitutes the initial line of defense against viral infections. IFN signaling induces the expression of interferon-stimulated genes (ISGs), and their products frequently restrict viral infection. Retroviruses like the human immunodeficiency viruses and the human T-lymphotropic viruses cause severe human diseases and are targeted by ISG-encoded proteins. Here, we discuss ISGs that inhibit the translation of retroviral mRNAs and thereby retrovirus propagation. The Schlafen proteins degrade cellular tRNAs and rRNAs needed for translation. Zinc Finger Antiviral Protein and RNA-activated protein kinase inhibit translation initiation factors, and Shiftless suppresses translation recoding essential for the expression of retroviral enzymes. We outline common mechanisms that underlie the antiviral activity of multifunctional ISGs and discuss potential antiretroviral therapeutic approaches based on the mode of action of these ISGs.


Subject(s)
Interferons , Protein Biosynthesis , Retroviridae , Humans , Interferons/immunology , Interferons/metabolism , Interferons/genetics , Retroviridae/genetics , Retroviridae/physiology , Immunity, Innate , Animals , Signal Transduction , Retroviridae Infections/virology , Retroviridae Infections/immunology , Retroviridae Infections/genetics
3.
Viruses ; 16(4)2024 04 13.
Article in English | MEDLINE | ID: mdl-38675945

ABSTRACT

The field of retroviral integration research has a long history that started with the provirus hypothesis and subsequent discoveries of the retroviral reverse transcriptase and integrase enzymes. Because both enzymes are essential for retroviral replication, they became valued targets in the effort to discover effective compounds to inhibit HIV-1 replication. In 2007, the first integrase strand transfer inhibitor was licensed for clinical use, and subsequently approved second-generation integrase inhibitors are now commonly co-formulated with reverse transcriptase inhibitors to treat people living with HIV. International meetings specifically focused on integrase and retroviral integration research first convened in 1995, and this paper is part of the Viruses Special Issue on the 7th International Conference on Retroviral Integration, which was held in Boulder Colorado in the summer of 2023. Herein, we overview key historical developments in the field, especially as they pertain to the development of the strand transfer inhibitor drug class. Starting from the mid-1990s, research advancements are presented through the lens of the international conferences. Our overview highlights the impact that regularly scheduled, subject-specific international meetings can have on community-building and, as a result, on field-specific collaborations and scientific advancements.


Subject(s)
Congresses as Topic , Retroviridae , Virus Integration , Humans , Virus Integration/drug effects , Retroviridae/physiology , Retroviridae/drug effects , Retroviridae/genetics , HIV Infections/drug therapy , HIV Infections/virology , HIV-1/drug effects , HIV-1/physiology , HIV-1/genetics , History, 21st Century , History, 20th Century
4.
Proc Natl Acad Sci U S A ; 119(11): e2119415119, 2022 03 15.
Article in English | MEDLINE | ID: mdl-35259018

ABSTRACT

SignificanceHosts often target the relatively conserved regions in rapidly mutating retroviruses to inhibit their replication. One of these regions is called a primer binding site (PBS), which has to be complementary to the host tRNA to initiate reverse transcription. By analyzing endogenous retroviral elements, we found that host cells use this sequence as a target in efforts to block the expression of viral elements. A specific type of zinc finger protein targets the PBS in a host genome, which not only inhibits the transcription of endogenous viruses but also inhibits the replication of exogenous retroviruses with the same PBS. Thus, our study sheds light on a strategy for searching for host restriction factors targeting retroviruses.


Subject(s)
Gene Expression Regulation, Viral , Host-Pathogen Interactions , RNA, Viral/genetics , RNA, Viral/metabolism , Repressor Proteins/metabolism , Retroviridae/physiology , Zinc Fingers , Base Sequence , Binding Sites , Chromosome Mapping , Endogenous Retroviruses , Genome-Wide Association Study , Humans , Nucleotide Motifs , Retroviridae/classification , Transcription, Genetic , Virus Replication
5.
Viruses ; 14(2)2022 02 05.
Article in English | MEDLINE | ID: mdl-35215917

ABSTRACT

Retroviruses must selectively recognize their unspliced RNA genome (gRNA) among abundant cellular and spliced viral RNAs to assemble into newly formed viral particles. Retroviral gRNA packaging is governed by Gag precursors that also orchestrate all the aspects of viral assembly. Retroviral life cycles, and especially the HIV-1 one, have been previously extensively analyzed by several methods, most of them based on molecular biology and biochemistry approaches. Despite these efforts, the spatio-temporal mechanisms leading to gRNA packaging and viral assembly are only partially understood. Nevertheless, in these last decades, progress in novel bioimaging microscopic approaches (as FFS, FRAP, TIRF, and wide-field microscopy) have allowed for the tracking of retroviral Gag and gRNA in living cells, thus providing important insights at high spatial and temporal resolution of the events regulating the late phases of the retroviral life cycle. Here, the implementation of these recent bioimaging tools based on highly performing strategies to label fluorescent macromolecules is described. This report also summarizes recent gains in the current understanding of the mechanisms employed by retroviral Gag polyproteins to regulate molecular mechanisms enabling gRNA packaging and the formation of retroviral particles, highlighting variations and similarities among the different retroviruses.


Subject(s)
Capsid/metabolism , Gene Products, gag/metabolism , Retroviridae Infections/virology , Retroviridae/physiology , Virus Assembly , Animals , Gene Products, gag/genetics , Genome, Viral , Humans , Retroviridae/genetics , Virion/genetics , Virion/physiology
6.
J Gen Virol ; 102(12)2021 12.
Article in English | MEDLINE | ID: mdl-34939563

ABSTRACT

Viruses in the family Retroviridae are found in a wide variety of vertebrate hosts. Enveloped virions are 80-100 nm in diameter with an inner core containing the viral genome and replicative enzymes. Core morphology is often characteristic for viruses within the same genus. Replication involves reverse transcription and integration into host cell DNA, resulting in a provirus. Integration into germline cells can result in a heritable provirus known as an endogenous retrovirus. This is a summary of the International Committee on Taxonomy of Viruses (ICTV) Report on the family Retroviridae, which is available at ictv.global/report/retroviridae.


Subject(s)
DNA Viruses/classification , Retroviridae/classification , Animals , DNA Viruses/genetics , DNA Viruses/physiology , DNA Viruses/ultrastructure , Genome, Viral , Host Specificity , Retroviridae/genetics , Retroviridae/physiology , Retroviridae/ultrastructure , Vertebrates/virology , Virion/ultrastructure , Virus Replication
7.
Viruses ; 13(12)2021 12 15.
Article in English | MEDLINE | ID: mdl-34960784

ABSTRACT

Understanding the molecular mechanisms of retroviral assembly has been a decades-long endeavor. With the recent discovery of inositol hexakisphosphate (IP6) acting as an assembly co-factor for human immunodeficiency virus (HIV), great strides have been made in retroviral research. In this review, the enzymatic pathways to synthesize and metabolize inositol phosphates (IPs) relevant to retroviral assembly are discussed. The functions of these enzymes and IPs are outlined in the context of the cellular biology important for retroviruses. Lastly, the recent advances in understanding the role of IPs in retroviral biology are surveyed.


Subject(s)
Inositol Phosphates/metabolism , Retroviridae/physiology , Virus Assembly/physiology , Biosynthetic Pathways , HIV Infections/virology , Humans , Phytic Acid/metabolism , Virion/metabolism , gag Gene Products, Human Immunodeficiency Virus/chemistry , gag Gene Products, Human Immunodeficiency Virus/metabolism
8.
Viruses ; 13(11)2021 11 04.
Article in English | MEDLINE | ID: mdl-34835027

ABSTRACT

Paradigm shifts throughout the history of microbiology have typically been ignored, or met with skepticism and resistance, by the scientific community. This has been especially true in the field of virology, where the discovery of a "contagium vivum fluidum", or infectious fluid remaining after excluding bacteria by filtration, was initially ignored because it did not coincide with the established view of microorganisms. Subsequent studies on such infectious agents, eventually termed "viruses", were met with skepticism. However, after an abundance of proof accumulated, viruses were eventually acknowledged as defined microbiological entities. Next, the proposed role of viruses in oncogenesis in animals was disputed, as was the unique mechanism of genome replication by reverse transcription of RNA by the retroviruses. This same pattern of skepticism holds true for the prediction of the existence of retroviral "antisense" transcripts and genes. From the time of their discovery, it was thought that retroviruses encoded proteins on only one strand of proviral DNA. However, in 1988, it was predicted that human immunodeficiency virus type 1 (HIV-1), and other retroviruses, express an antisense protein encoded on the DNA strand opposite that encoding the known viral proteins. Confirmation came quickly with the characterization of the antisense protein, HBZ, of the human T-cell leukemia virus type 1 (HTLV-1), and the finding that both the protein and its antisense mRNA transcript play key roles in viral replication and pathogenesis. However, acceptance of the existence, and potential importance, of a corresponding antisense transcript and protein (ASP) in HIV-1 infection and pathogenesis has lagged, despite gradually accumulating theoretical and experimental evidence. The most striking theoretical evidence is the finding that asp is highly conserved in group M viruses and correlates exclusively with subtypes, or clades, responsible for the AIDS pandemic. This review outlines the history of the major shifts in thought pertaining to the nature and characteristics of viruses, and in particular retroviruses, and details the development of the hypothesis that retroviral antisense transcripts and genes exist. We conclude that there is a need to accelerate studies on ASP, and its transcript(s), with the view that both may be important, and overlooked, targets in anti-HIV therapeutic and vaccine strategies.


Subject(s)
RNA, Antisense/genetics , RNA, Messenger/genetics , Retroviridae Proteins/genetics , Retroviridae/genetics , Carcinogenesis/genetics , Genome, Viral , HIV-1/genetics , HIV-1/pathogenicity , HIV-1/physiology , History, 20th Century , History, 21st Century , Human Immunodeficiency Virus Proteins/genetics , Human T-lymphotropic virus 1/genetics , Human T-lymphotropic virus 1/pathogenicity , Human T-lymphotropic virus 1/physiology , Humans , Open Reading Frames , Retroviridae/pathogenicity , Retroviridae/physiology , Transcription, Genetic , Viral Envelope Proteins/genetics , Virology/history , Virus Replication
9.
Viruses ; 13(11)2021 11 09.
Article in English | MEDLINE | ID: mdl-34835055

ABSTRACT

Retroviral infection delivers an RNA genome into the cytoplasm that serves as the template for the synthesis of a linear double-stranded DNA copy by the viral reverse transcriptase. Within the nucleus this linear DNA gives rise to extrachromosomal circular forms, and in a key step of the life cycle is inserted into the host genome to form the integrated provirus. The unintegrated DNA forms, like those of DNAs entering cells by other means, are rapidly loaded with nucleosomes and heavily silenced by epigenetic histone modifications. This review summarizes our present understanding of the silencing machinery for the DNAs of the mouse leukemia viruses and human immunodeficiency virus type 1. We consider the potential impact of the silencing on virus replication, on the sensing of the virus by the innate immune system, and on the formation of latent proviruses. We also speculate on the changeover to high expression from the integrated proviruses in permissive cell types, and briefly consider the silencing of proviruses even after integration in embryonic stem cells and other developmentally primitive cell types.


Subject(s)
DNA, Viral/genetics , Gene Silencing , Retroviridae/genetics , Animals , HIV-1/genetics , HIV-1/physiology , Histone Code , Humans , Leukemia Virus, Murine/genetics , Leukemia Virus, Murine/physiology , Proviruses/genetics , Proviruses/physiology , Retroviridae/physiology , Transcription, Genetic , Virus Integration , Virus Replication
10.
Viruses ; 13(9)2021 09 17.
Article in English | MEDLINE | ID: mdl-34578434

ABSTRACT

The small cellular molecule inositol hexakisphosphate (IP6) has been known for ~20 years to promote the in vitro assembly of HIV-1 into immature virus-like particles. However, the molecular details underlying this effect have been determined only recently, with the identification of the IP6 binding site in the immature Gag lattice. IP6 also promotes formation of the mature capsid protein (CA) lattice via a second IP6 binding site, and enhances core stability, creating a favorable environment for reverse transcription. IP6 also enhances assembly of other retroviruses, from both the Lentivirus and the Alpharetrovirus genera. These findings suggest that IP6 may have a conserved function throughout the family Retroviridae. Here, we discuss the different steps in the viral life cycle that are influenced by IP6, and describe in detail how IP6 interacts with the immature and mature lattices of different retroviruses.


Subject(s)
HIV-1/physiology , Phytic Acid/metabolism , Retroviridae/physiology , Virus Assembly , Binding Sites , Capsid Proteins , Human Immunodeficiency Virus Proteins/metabolism , Mutation , Retroviridae Proteins/metabolism , Reverse Transcription , Rous sarcoma virus/physiology , Virus Replication , gag Gene Products, Human Immunodeficiency Virus/genetics , gag Gene Products, Human Immunodeficiency Virus/metabolism
11.
Viruses ; 13(8)2021 07 22.
Article in English | MEDLINE | ID: mdl-34452291

ABSTRACT

HIV-1 can infect non-dividing cells. The nuclear envelope therefore represents a barrier that HIV-1 must traverse in order to gain access to the host cell chromatin for integration. Hence, nuclear entry is a critical step in the early stages of HIV-1 replication. Following membrane fusion, the viral capsid (CA) lattice, which forms the outer face of the retroviral core, makes numerous interactions with cellular proteins that orchestrate the progress of HIV-1 through the replication cycle. The ability of CA to interact with nuclear pore proteins and other host factors around the nuclear pore determines whether nuclear entry occurs. Uncoating, the process by which the CA lattice opens and/or disassembles, is another critical step that must occur prior to integration. Both early and delayed uncoating have detrimental effects on viral infectivity. How uncoating relates to nuclear entry is currently hotly debated. Recent technological advances have led to intense discussions about the timing, location, and requirements for uncoating and have prompted the field to consider alternative uncoating scenarios that presently focus on uncoating at the nuclear pore and within the nuclear compartment. This review describes recent advances in the study of HIV-1 nuclear entry, outlines the interactions of the retroviral CA protein, and discusses the challenges of investigating HIV-1 uncoating.


Subject(s)
Capsid Proteins/metabolism , Capsid/metabolism , Cell Nucleus/virology , HIV Infections/virology , HIV-1/physiology , Virus Uncoating , Animals , Cell Nucleus/metabolism , HIV-1/genetics , Host-Pathogen Interactions , Humans , Nuclear Envelope/physiology , Nuclear Envelope/virology , Nuclear Pore/physiology , Nuclear Pore/virology , Retroviridae/physiology , Reverse Transcription
12.
Brief Bioinform ; 22(6)2021 11 05.
Article in English | MEDLINE | ID: mdl-34463709

ABSTRACT

Oncovirus integrations cause copy number variations and complex structural variations (SVs) on host genomes. However, the understanding of how inserted viral DNA impacts the local genome remains limited. The linear structure of the oncovirus integrated local genomic map (LGM) will lay the foundations to understand how oncovirus integrations emerge and compromise the host genome's functioning. We propose a conjugate graph model to reconstruct the rearranged LGM at integrated loci. Simulation tests prove the reliability and credibility of the algorithm. Applications of the algorithm to whole-genome sequencing data of human papillomavirus (HPV) and hepatitis B virus (HBV)-infected cancer samples gained biological insights on oncovirus integrations. We observed four affection patterns of oncovirus integrations from the HPV and HBV-integrated cancer samples, including the coding-frame truncation, hyper-amplification of tumor gene, the viral cis-regulation inserted at the single intron and at the intergenic region. We found that the focal duplicates and host SVs are frequent in the HPV-integrated LGMs, while the focal deletions are prevalent in HBV-integrated LGMs. Furthermore, with the results yields from our method, we found the enhanced microhomology-mediated end joining might lead to both HPV and HBV integrations and conjectured that the HPV integrations might mainly occur during the DNA replication process. The conjugate graph algorithm code and LGM construction pipeline, available at https://github.com/deepomicslab/FuseSV.


Subject(s)
Computational Biology/methods , DNA Copy Number Variations , Genome, Human , Retroviridae/physiology , User-Computer Interface , Virus Integration , Algorithms , Base Sequence , DNA, Viral , Databases, Genetic , Humans , Neoplasms/etiology
13.
Viruses ; 13(7)2021 06 26.
Article in English | MEDLINE | ID: mdl-34206771

ABSTRACT

Gene therapy vectors derived from different viral species have become a fixture in biomedicine, both for direct therapeutic intervention and as tools to facilitate cell-based therapies, such as chimeric antigen receptor-based immunotherapies. On the contrary, extracellular vesicles have only recently gained a massive increase in interest and, concomitantly, knowledge in the field has drastically risen. Viral infections and extracellular vesicle biology overlap in many ways, both with pro- and antiviral outcomes. In this review, we take a closer look at these interactions for the most prominent groups of viral vectors (Adenoviral, Adeno-associated and Retro/Lentiviral vectors) and the possible implications of these overlaps for viral vector technology and its biomedical applications.


Subject(s)
Adenoviridae/genetics , Dependovirus/genetics , Extracellular Vesicles , Genetic Vectors , Retroviridae/genetics , Adenoviridae/physiology , Dependovirus/physiology , Genetic Therapy , Humans , Lentivirus/genetics , Retroviridae/physiology , Virus Diseases/virology
14.
Elife ; 102021 07 05.
Article in English | MEDLINE | ID: mdl-34223819

ABSTRACT

Early events in retrovirus transmission are determined by interactions between incoming viruses and frontline cells near entry sites. Despite their importance for retroviral pathogenesis, very little is known about these events. We developed a bioluminescence imaging (BLI)-guided multiscale imaging approach to study these events in vivo. Engineered murine leukemia reporter viruses allowed us to monitor individual stages of retrovirus life cycle including virus particle flow, virus entry into cells, infection and spread for retroorbital, subcutaneous, and oral routes. BLI permitted temporal tracking of orally administered retroviruses along the gastrointestinal tract as they traversed the lumen through Peyer's patches to reach the draining mesenteric sac. Importantly, capture and acquisition of lymph-, blood-, and milk-borne retroviruses spanning three routes was promoted by a common host factor, the I-type lectin CD169, expressed on sentinel macrophages. These results highlight how retroviruses co-opt the immune surveillance function of tissue-resident sentinel macrophages for establishing infection.


Subject(s)
Retroviridae Infections/diagnostic imaging , Retroviridae Infections/transmission , Retroviridae/physiology , Sialic Acid Binding Ig-like Lectin 1/metabolism , Animals , Disease Models, Animal , Female , Humans , Leukemia Virus, Murine , Life Cycle Stages , Lymph Nodes , Macrophages/virology , Male , Mammary Glands, Human/diagnostic imaging , Mammary Glands, Human/virology , Mice , Retroviridae/genetics , Retroviridae Infections/metabolism , Retroviridae Infections/pathology , Sialic Acid Binding Ig-like Lectin 1/genetics , Spleen/diagnostic imaging , Virion , Virus Internalization
15.
Viruses ; 13(6)2021 06 13.
Article in English | MEDLINE | ID: mdl-34199191

ABSTRACT

The proper assembly and dissemination of progeny virions is a fundamental step in virus replication. As a whole, viruses have evolved a myriad of strategies to exploit cellular compartments and mechanisms to ensure a successful round of infection. For enveloped viruses such as retroviruses and herpesviruses, acquisition and incorporation of cellular membrane is an essential process during the formation of infectious viral particles. To do this, these viruses have evolved to hijack the host Endosomal Sorting Complexes Required for Transport (ESCRT-I, -II, and -III) to coordinate the sculpting of cellular membrane at virus assembly and dissemination sites, in seemingly different, yet fundamentally similar ways. For instance, at the plasma membrane, ESCRT-I recruitment is essential for HIV-1 assembly and budding, while it is dispensable for the release of HSV-1. Further, HSV-1 was shown to recruit ESCRT-III for nuclear particle assembly and egress, a process not used by retroviruses during replication. Although the cooption of ESCRTs occurs in two separate subcellular compartments and at two distinct steps for these viral lifecycles, the role fulfilled by ESCRTs at these sites appears to be conserved. This review discusses recent findings that shed some light on the potential parallels between retroviral budding and nuclear egress and proposes a model where HSV-1 nuclear egress may occur through an ESCRT-dependent mechanism.


Subject(s)
Endosomal Sorting Complexes Required for Transport/metabolism , Host Microbial Interactions , Retroviridae/physiology , Virus Assembly , Virus Release , DNA Viruses/physiology , HIV-1/physiology , Humans , Protein Transport , Virion/metabolism , Virus Replication
16.
BMC Immunol ; 22(1): 49, 2021 07 28.
Article in English | MEDLINE | ID: mdl-34320928

ABSTRACT

BACKGROUND: Targeted immunotherapy is mostly associated with cancer treatment wherein designed molecules engage signaling pathways and mutant proteins critical to the survival of the cell. One of several genetic approaches is the use of in silico methods to develop immune epitopes targeting specific antigenic regions on related mutant proteins. In a recent study we showed a functional association between the gamma retrovirus HERV-H Long Terminal Associating (HHLA1, HHLA2 and HHLA3) proteins and melanoma associated antigen of the B class proteins (MAGEB5), with a resultant decrease in expression of HLA class I and II immune variants. HLA-C and HLA-DRB5 were the main HLA class I and II Immune variants, respectively, that showed expression changes across viral samples of interest. Specific immune variants for HLA-C and HLA-DRB5 were filtered for the top ten based on their relative frequency of counts across the samples. RESULTS: Protein variants for HHLA1, HHLA2, HHLA3 and MAGEB5 were used to predict antigenic epitope peptides to immune peptide-MHC class I and II binding using artificial neural networks. For IC50 peptide scores (PS) ≥ 0.5 with a transformed binding ability between 0 and 1, the top 5 epitopes identified for all targeted genes HHLA1,2 & 3 and MAGEB5 were qualified as strong or weak binders according to the threshold. Domain analysis using NCBI Conserved Domain Database (CDD) identified HHLA2 with immunoglobulin-like domains (Ig_C1-set) and MAGEB5 with the MAGE Homology Domain (MHD). Linear regression showed a statistical correlation (P < 0.001) for HHLA2 and MAGEB5 predicted epitope peptides to HLA-C but not HLA-DRB5. The prediction model identified HLA-C variant 9 (HLA-C9, BAA08825.1 HLA-B*1511) at 1.1% as the most valuable immune target for clinical considerations. Identification of the 9-mer epitope peptide within the domain showed for HHLA2: YANRTSLFY (PS = 0.5837) and VLAYYLSSSQNTIIN (PS = 0.77) for HLA-C and HLA-DRB5, respectively and for MAGEB5, peptides: FVRLTYLEY (PS = 0.5293) and YPAHYQFLWGPRAYT (PS = 0.62) for HLA-C and HLA-DRB5, respectively. CONCLUSION: Specific immune responses to targeted epitope peptides and their prediction models, suggested co-expression and co-evolution for HHLA2 and MAGEB5 in viral related diseases. HHLA2 and MAGEB5 could be considered markers for virus related tumors and targeted therapy for oncogenic diseases.


Subject(s)
Antigens, Neoplasm/metabolism , Epitopes, T-Lymphocyte/immunology , Immunoglobulins/immunology , Immunotherapy/methods , Melanoma/immunology , Neoplasm Proteins/metabolism , Retroviridae Infections/immunology , Retroviridae/physiology , Antigens, Neoplasm/genetics , Epitope Mapping , Epitopes, T-Lymphocyte/genetics , Evolution, Molecular , Gene Expression Regulation , Genetic Association Studies , HLA Antigens/genetics , HLA-C Antigens/genetics , HLA-DRB5 Chains/genetics , Humans , Immunoglobulins/genetics , Melanoma/genetics , Mutation/genetics , Neoplasm Proteins/genetics , Polymorphism, Genetic
17.
Virology ; 561: 6-16, 2021 09.
Article in English | MEDLINE | ID: mdl-34089997

ABSTRACT

Based on the previously reported 13-residue antibacterial peptide analog, brevinin-1EMa (FLGWLFKVASKVL, peptide B), we attempted to design a novel class of antiviral peptides. For this goal, we synthesized three peptides with different stapling positions (B-2S, B-8S, and B-5S). The most active antiviral peptide with the specific stapling position (B-5S) was further modified in combination with either cysteine (B-5S3C, B-5S7C, and B-5S10C) or hydrophilic amino acid substitution (Bsub and Bsub-5S). Overall, B, B-5S, and Bsub-5S peptides showed superior antiviral activities against enveloped viruses such as retrovirus, lentivirus, hepatitis C virus, and herpes simplex virus with EC50 values of 1-5 µM. Murine norovirus, a non-enveloped virus, was not susceptible to the virucidal actions of these peptides, suggesting the virus membrane disruption as their main antiviral mechanisms of action. We believe that these three novel peptides could serve as promising candidates for further development of membrane-targeting antiviral drugs in the future.


Subject(s)
Antiviral Agents/pharmacology , Ion Channels/chemistry , Ion Channels/pharmacology , Peptides/pharmacology , Virus Internalization/drug effects , Viruses/drug effects , Antiviral Agents/chemistry , Antiviral Agents/metabolism , Bacteria/drug effects , Cell Line , Drug Design , Hepacivirus/drug effects , Hepacivirus/physiology , Herpesvirus 1, Human/drug effects , Herpesvirus 1, Human/physiology , Humans , Ion Channels/metabolism , Lentivirus/drug effects , Lentivirus/physiology , Microbial Sensitivity Tests , Norovirus/drug effects , Norovirus/physiology , Peptides/chemistry , Peptides/metabolism , Retroviridae/drug effects , Retroviridae/physiology , Virus Physiological Phenomena
18.
Viruses ; 13(5)2021 05 02.
Article in English | MEDLINE | ID: mdl-34063186

ABSTRACT

Immunotherapy has been shown to be highly effective in some types of cancer caused by viruses. Gene therapy involves insertion or modification of a therapeutic gene, to correct for inappropriate gene products that cause/may cause diseases. Both these types of therapy have been used as alternative ways to avoid cancers caused by oncoviruses. In this review, we summarize recent studies on immunotherapy and gene therapy including the topics of oncolytic immunotherapy, immune checkpoint inhibitors, gene replacement, antisense oligonucleotides, RNA interference, clustered regularly interspaced short palindromic repeats Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)-based gene editing, transcription activator-like effector nucleases (TALENs) and custom treatment for Epstein-Barr virus, human T-lymphotropic virus 1, hepatitis B virus, human papillomavirus, hepatitis C virus, herpesvirus associated with Kaposi's sarcoma, Merkel cell polyomavirus, and cytomegalovirus.


Subject(s)
Genetic Therapy , Immunotherapy , Retroviridae Infections/therapy , Retroviridae/physiology , Animals , Clustered Regularly Interspaced Short Palindromic Repeats , Gene Editing , Humans , Retroviridae/genetics , Retroviridae Infections/genetics , Retroviridae Infections/immunology , Retroviridae Infections/virology
19.
OMICS ; 25(6): 358-371, 2021 06.
Article in English | MEDLINE | ID: mdl-34037476

ABSTRACT

About a tenth of all cancers are caused by viruses or associated with viral infection. Recent global events including the coronavirus disease-2019 (COVID-19) pandemic means that human encounter with viruses is increased. Cancer development in individuals with viral infection can take many years after infection, demonstrating that the involvement of viruses in cancer development is a long and complex process. This complexity emanates from individual genetic heterogeneity and the many steps involved in cancer development owing to viruses. The process of tumorigenesis is driven by the complex interaction between several viral factors and host factors leading to the creation of a tumor microenvironment (TME) that is ideal and promotes tumor formation. Viruses associated with human cancers ensure their survival and proliferation through activation of several cellular processes including inflammation, migration, and invasion, resistance to apoptosis and growth suppressors. In addition, most human oncoviruses evade immune detection and can activate signaling cascades including the PI3K-Akt-mTOR, Notch and Wnt pathways associated with enhanced proliferation and angiogenesis. This expert review examines and synthesizes the multiple biological factors related to oncoviruses, and the signaling cascades activated by these viruses contributing to viral oncogenesis. In particular, I examine and review the Epstein-Barr virus, human papillomaviruses, and Kaposi's sarcoma herpes virus in a context of cancer pathogenesis. I conclude with a future outlook on therapeutic targeting of the viruses and their associated oncogenic pathways within the TME. These anticancer strategies can be in the form of, but not limited to, antibodies and inhibitors.


Subject(s)
Epstein-Barr Virus Infections/virology , Neoplasms/virology , Papillomavirus Infections/virology , Retroviridae Infections/virology , Retroviridae/physiology , Sarcoma, Kaposi/virology , Tumor Virus Infections/virology , Alphapapillomavirus/physiology , Carcinogenesis , Cell Transformation, Viral , Epstein-Barr Virus Infections/pathology , Herpesvirus 4, Human/physiology , Herpesvirus 8, Human/physiology , Humans , Molecular Targeted Therapy , Neoplasms/pathology , Neoplasms/therapy , Papillomavirus Infections/pathology , Retroviridae Infections/pathology , Sarcoma, Kaposi/pathology , Signal Transduction , Tumor Microenvironment , Tumor Virus Infections/pathology
20.
Viruses ; 13(3)2021 03 16.
Article in English | MEDLINE | ID: mdl-33809689

ABSTRACT

I was fortunate to be associated with the lab of Stephen Oroszlan at the US National Cancer Institute from ~1982 until his conversion to Emeritus status in 1995. His lab made groundbreaking discoveries on retroviral proteins during that time, including many features that could not have been inferred or anticipated from straightforward sequence information. Building on the Oroszlan lab results, my colleagues and I demonstrated that the zinc fingers in nucleocapsid proteins play a crucial role in genomic RNA encapsidation; that the N-terminal myristylation of the Gag proteins of many retroviruses is important for their association with the plasma membrane before particle assembly is completed; and that gammaretroviruses initially synthesize their Env protein as an inactive precursor and then truncate the cytoplasmic tail of the transmembrane protein, activating Env fusogenicity, during virus maturation. We also elucidated several aspects of the mechanism of translational suppression in pol gene expression in gammaretroviruses; amazingly, this is a fundamentally different mechanism of suppression from that in most other retroviral genera.


Subject(s)
Retroviridae , Cell Membrane/metabolism , History, 21st Century , Retroviridae/genetics , Retroviridae/physiology , Viral Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...