Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 16.132
Filtrar
1.
Methods Mol Biol ; 2808: 35-56, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38743361

RESUMEN

Mononegaviruses are promising tools as oncolytic and transgene vectors for gene therapy and regenerative medicine. However, when mononegaviruses are used for therapeutic applications, the viral activity must be strictly controlled due to concerns about toxicity and severe side effects. With this technology, mononegavirus vectors can be grown where they are intended and can be easily removed when they are no longer needed. In particular, a photoswitch protein called Magnet (consisting of two magnet domains) is incorporated into the hinge region between the connector and methyltransferase domains of the mononegavirus polymerase protein (L protein) to disrupt the L protein functions. Blue light (470 ± 20 nm) irradiation causes the dimerization of the two magnet domains, and the L protein is restored to activity, allowing viral gene expression and virus replication. Since the magnet domains' dimerization is reversible, viral gene expression and replication cease when blue light irradiation is stopped.


Asunto(s)
Regulación Viral de la Expresión Génica , Replicación Viral , Replicación Viral/genética , Humanos , Proteínas Virales/genética , Proteínas Virales/metabolismo , Luz , Animales , Vectores Genéticos/genética
2.
Molecules ; 29(9)2024 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-38731543

RESUMEN

Ribonuclease P (RNase P) complexed with an external guide sequence (EGS) represents a promising nucleic acid-based gene targeting approach for gene expression knock-down and modulation. The RNase P-EGS strategy is unique as an EGS can be designed to basepair any mRNA sequence and recruit intracellular RNase P for hydrolysis of the target mRNA. In this study, we provide the first direct evidence that the RNase P-based approach effectively blocks the gene expression and replication of herpes simplex virus 2 (HSV-2), the causative agent of genital herpes. We constructed EGSs to target the mRNA encoding HSV-2 single-stranded DNA binding protein ICP8, which is essential for viral DNA genome replication and growth. In HSV-2 infected cells expressing a functional EGS, ICP8 levels were reduced by 85%, and viral growth decreased by 3000 folds. On the contrary, ICP8 expression and viral growth exhibited no substantial differences between cells expressing no EGS and those expressing a disabled EGS with mutations precluding RNase P recognition. The anti-ICP8 EGS is specific in targeting ICP8 because it only affects ICP8 expression but does not affect the expression of the other viral immediate-early and early genes examined. This study shows the effective and specific anti-HSV-2 activity of the RNase P-EGS approach and demonstrates the potential of EGS RNAs for anti-HSV-2 applications.


Asunto(s)
Regulación Viral de la Expresión Génica , Herpesvirus Humano 2 , Replicación Viral , Herpesvirus Humano 2/genética , Herpesvirus Humano 2/fisiología , Humanos , Ribonucleasa P/metabolismo , Ribonucleasa P/genética , Animales , Proteínas Virales/genética , Proteínas Virales/metabolismo , Chlorocebus aethiops , ARN Mensajero/genética , ARN Mensajero/metabolismo , Células Vero , Proteínas Inmediatas-Precoces/genética , Proteínas Inmediatas-Precoces/metabolismo , Proteínas de Unión al ADN
3.
Nat Commun ; 15(1): 3729, 2024 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-38702330

RESUMEN

The unique virus-cell interaction in Epstein-Barr virus (EBV)-associated malignancies implies targeting the viral latent-lytic switch is a promising therapeutic strategy. However, the lack of specific and efficient therapeutic agents to induce lytic cycle in these cancers is a major challenge facing clinical implementation. We develop a synthetic transcriptional activator that specifically activates endogenous BZLF1 and efficiently induces lytic reactivation in EBV-positive cancer cells. A lipid nanoparticle encapsulating nucleoside-modified mRNA which encodes a BZLF1-specific transcriptional activator (mTZ3-LNP) is synthesized for EBV-targeted therapy. Compared with conventional chemical inducers, mTZ3-LNP more efficiently activates EBV lytic gene expression in EBV-associated epithelial cancers. Here we show the potency and safety of treatment with mTZ3-LNP to suppress tumor growth in EBV-positive cancer models. The combination of mTZ3-LNP and ganciclovir yields highly selective cytotoxic effects of mRNA-based lytic induction therapy against EBV-positive tumor cells, indicating the potential of mRNA nanomedicine in the treatment of EBV-associated epithelial cancers.


Asunto(s)
Infecciones por Virus de Epstein-Barr , Herpesvirus Humano 4 , Liposomas , Nanopartículas , Transactivadores , Humanos , Herpesvirus Humano 4/genética , Transactivadores/metabolismo , Transactivadores/genética , Infecciones por Virus de Epstein-Barr/virología , Infecciones por Virus de Epstein-Barr/tratamiento farmacológico , Animales , Nanopartículas/química , Línea Celular Tumoral , Ratones , ARN Mensajero/genética , ARN Mensajero/metabolismo , Activación Viral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto , Regulación Viral de la Expresión Génica/efectos de los fármacos , Ratones Desnudos , Femenino
4.
Nat Commun ; 15(1): 4156, 2024 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-38755141

RESUMEN

Epstein-Barr virus (EBV) uses a biphasic lifecycle of latency and lytic reactivation to infect >95% of adults worldwide. Despite its central role in EBV persistence and oncogenesis, much remains unknown about how EBV latency is maintained. We used a human genome-wide CRISPR/Cas9 screen to identify that the nuclear protein SFPQ was critical for latency. SFPQ supported expression of linker histone H1, which stabilizes nucleosomes and regulates nuclear architecture, but has not been previously implicated in EBV gene regulation. H1 occupied latent EBV genomes, including the immediate early gene BZLF1 promoter. Upon reactivation, SFPQ was sequestered into sub-nuclear puncta, and EBV genomic H1 occupancy diminished. Enforced H1 expression blocked EBV reactivation upon SFPQ knockout, confirming it as necessary downstream of SFPQ. SFPQ knockout triggered reactivation of EBV in B and epithelial cells, as well as of Kaposi's sarcoma-associated herpesvirus in B cells, suggesting a conserved gamma-herpesvirus role. These findings highlight SFPQ as a major regulator of H1 expression and EBV latency.


Asunto(s)
Herpesvirus Humano 4 , Histonas , Factor de Empalme Asociado a PTB , Activación Viral , Latencia del Virus , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/fisiología , Humanos , Histonas/metabolismo , Activación Viral/genética , Latencia del Virus/genética , Factor de Empalme Asociado a PTB/metabolismo , Factor de Empalme Asociado a PTB/genética , Regulación Viral de la Expresión Génica , Linfocitos B/virología , Linfocitos B/metabolismo , Infecciones por Virus de Epstein-Barr/virología , Infecciones por Virus de Epstein-Barr/genética , Infecciones por Virus de Epstein-Barr/metabolismo , Sistemas CRISPR-Cas , Regiones Promotoras Genéticas/genética , Transactivadores/metabolismo , Transactivadores/genética , Genoma Viral
5.
Methods Mol Biol ; 2807: 175-194, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38743229

RESUMEN

Retroviruses must overcome cellular restrictions to the nucleocytoplasmic export of viral mRNAs that retain introns in order to complete their replication cycle. HIV accomplishes this using a system comprised of a trans-acting viral protein, Rev, and a cis-acting RNA secondary structure in the viral genome, the Rev-Response Element (RRE). HIV primary isolates differ with respect to the sequence and functional activity of the Rev-RRE system. Here, we describe a high throughput assay system for analyzing Rev-RRE functional activity using packageable viral vectors.


Asunto(s)
ARN Viral , Elementos de Respuesta , Productos del Gen rev del Virus de la Inmunodeficiencia Humana , Humanos , Productos del Gen rev del Virus de la Inmunodeficiencia Humana/genética , Productos del Gen rev del Virus de la Inmunodeficiencia Humana/metabolismo , Elementos de Respuesta/genética , ARN Viral/genética , VIH-1/genética , VIH-1/fisiología , Regulación Viral de la Expresión Génica , Replicación Viral/genética , Vectores Genéticos/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo
6.
J Virol ; 98(5): e0003224, 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38651900

RESUMEN

Critical stages of lytic herpes simplex virus type 1 (HSV-1) replication are marked by the sequential expression of immediate early (IE) to early (E), then late (L) viral genes. HSV-1 can also persist in neuronal cells via a non-replicative, transcriptionally repressed infection called latency. The regulation of lytic and latent transcriptional profiles is critical to HSV-1 pathogenesis and persistence. We sought a fluorescence-based approach to observe the outcome of neuronal HSV-1 infection at the single-cell level. To achieve this goal, we constructed and characterized a novel HSV-1 recombinant that enables discrimination between lytic and latent infection. The dual reporter HSV-1 encodes a human cytomegalovirus-immediate early (hCMV-IE) promoter-driven enhanced yellow fluorescent protein (eYFP) to visualize the establishment of infection and an endogenous mCherry-VP26 fusion to report lytic replication. We confirmed that viral gene expression, replication, and spread of infection are not altered by the incorporation of the fluorescent reporters, and fluorescent protein (FP) detection virtuously reports the progression of lytic replication. We demonstrate that the outcome of HSV-1 infection of compartmentalized primary neurons is determined by viral inoculating dose: high-dose axonal inoculation proceeds to lytic replication, whereas low-dose axonal inoculation establishes a latent HSV-1 infection. Interfering with low-dose axonal inoculation via small molecule drugs reports divergent phenotypes of eYFP and mCherry reporter detection, correlating with altered states of viral gene expression. We report that the transcriptional state of neuronal HSV-1 infection is variable in response to changes in the intracellular neuronal environment.IMPORTANCEHerpes simplex virus type 1 (HSV-1) is a prevalent human pathogen that infects approximately 67% of the global human population. HSV-1 invades the peripheral nervous system, where latent HSV-1 infection persists within the host for life. Immunological evasion, viral persistence, and herpetic pathologies are determined by the regulation of HSV-1 gene expression. Studying HSV-1 gene expression during neuronal infection is challenging but essential for the development of antiviral therapeutics and interventions. We used a recombinant HSV-1 to evaluate viral gene expression during infection of primary neurons. Manipulation of cell signaling pathways impacts the establishment and transcriptional state of HSV-1 latency in neurons. The work here provides critical insight into the cellular and viral factors contributing to the establishment of latent HSV-1 infection.


Asunto(s)
Herpes Simple , Herpesvirus Humano 1 , Proteínas Luminiscentes , Neuronas , Replicación Viral , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/fisiología , Neuronas/virología , Neuronas/metabolismo , Humanos , Animales , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Herpes Simple/virología , Genes Reporteros , Latencia del Virus/genética , Regulación Viral de la Expresión Génica , Chlorocebus aethiops , Células Vero , Citomegalovirus/genética , Citomegalovirus/fisiología
7.
PLoS Pathog ; 20(4): e1011939, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38683861

RESUMEN

Epstein-Barr virus (EBV) persistently infects 95% of adults worldwide and is associated with multiple human lymphomas that express characteristic EBV latency programs used by the virus to navigate the B-cell compartment. Upon primary infection, the EBV latency III program, comprised of six Epstein-Barr Nuclear Antigens (EBNA) and two Latent Membrane Protein (LMP) antigens, drives infected B-cells into germinal center (GC). By incompletely understood mechanisms, GC microenvironmental cues trigger the EBV genome to switch to the latency II program, comprised of EBNA1, LMP1 and LMP2A and observed in GC-derived Hodgkin lymphoma. To gain insights into pathways and epigenetic mechanisms that control EBV latency reprogramming as EBV-infected B-cells encounter microenvironmental cues, we characterized GC cytokine effects on EBV latency protein expression and on the EBV epigenome. We confirmed and extended prior studies highlighting GC cytokine effects in support of the latency II transition. The T-follicular helper cytokine interleukin 21 (IL-21), which is a major regulator of GC responses, and to a lesser extent IL-4 and IL-10, hyper-induced LMP1 expression, while repressing EBNA expression. However, follicular dendritic cell cytokines including IL-15 and IL-27 downmodulate EBNA but not LMP1 expression. CRISPR editing highlighted that STAT3 and STAT5 were necessary for cytokine mediated EBNA silencing via epigenetic effects at the EBV genomic C promoter. By contrast, STAT3 was instead necessary for LMP1 promoter epigenetic remodeling, including gain of activating histone chromatin marks and loss of repressive polycomb repressive complex silencing marks. Thus, EBV has evolved to coopt STAT signaling to oppositely regulate the epigenetic status of key viral genomic promoters in response to GC cytokine cues.


Asunto(s)
Epigénesis Genética , Infecciones por Virus de Epstein-Barr , Regulación Viral de la Expresión Génica , Centro Germinal , Herpesvirus Humano 4 , Latencia del Virus , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/fisiología , Humanos , Centro Germinal/inmunología , Centro Germinal/virología , Infecciones por Virus de Epstein-Barr/virología , Infecciones por Virus de Epstein-Barr/genética , Infecciones por Virus de Epstein-Barr/inmunología , Citocinas/metabolismo , Linfocitos B/virología , Linfocitos B/metabolismo
8.
Viruses ; 16(4)2024 03 25.
Artículo en Inglés | MEDLINE | ID: mdl-38675845

RESUMEN

Schlafen (SLFN) is a family of proteins upregulated by type I interferons with a regulatory role in translation. Intriguingly, SLFN14 associates with the ribosome and can degrade rRNA, tRNA, and mRNA in vitro, but a role in translation is still unknown. Ribosomes are important regulatory hubs during translation elongation of mRNAs rich in rare codons. Therefore, we evaluated the potential role of SLFN14 in the expression of mRNAs enriched in rare codons, using HIV-1 genes as a model. We found that, in a variety of cell types, including primary immune cells, SLFN14 regulates the expression of HIV-1 and non-viral genes based on their codon adaptation index, a measurement of the synonymous codon usage bias; consequently, SLFN14 inhibits the replication of HIV-1. The potent inhibitory effect of SLFN14 on the expression of the rare codon-rich transcript HIV-1 Gag was minimized by codon optimization. Mechanistically, we found that the endoribonuclease activity of SLFN14 is required, and that ribosomal RNA degradation is involved. Therefore, we propose that SLFN14 impairs the expression of HIV-1 transcripts rich in rare codons, in a catalytic-dependent manner.


Asunto(s)
Uso de Codones , VIH-1 , Replicación Viral , Humanos , Codón/genética , Regulación Viral de la Expresión Génica , Células HEK293 , Infecciones por VIH/virología , Infecciones por VIH/genética , VIH-1/genética , VIH-1/fisiología , Biosíntesis de Proteínas , ARN Mensajero/genética , ARN Mensajero/metabolismo , Línea Celular Tumoral
9.
Viruses ; 16(4)2024 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-38675871

RESUMEN

The post-transcriptional regulatory element (PRE) is present in all HBV mRNAs and plays a major role in their stability, nuclear export, and enhancement of viral gene expression. Understanding PRE's structure, function, and mode of action is essential to leverage its potential as a therapeutic target. A wide range of PRE-based reagents and tools have been developed and assessed in preclinical and clinical settings for therapeutic and biotechnology applications. This manuscript aims to provide a systematic review of the characteristics and mechanism of action of PRE, as well as elucidating its current applications in basic and clinical research. Finally, we discuss the promising opportunities that PRE may provide to antiviral development, viral biology, and potentially beyond.


Asunto(s)
Virus de la Hepatitis B , Hepatitis B , ARN Viral , Virus de la Hepatitis B/genética , Virus de la Hepatitis B/fisiología , Humanos , Hepatitis B/virología , Hepatitis B/tratamiento farmacológico , ARN Viral/genética , ARN Viral/metabolismo , Antivirales/uso terapéutico , Antivirales/farmacología , Regulación Viral de la Expresión Génica , ARN Mensajero/genética , Procesamiento Postranscripcional del ARN , Animales
10.
PLoS Pathog ; 20(4): e1012172, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38662769

RESUMEN

The implementation of antiretroviral therapy (ART) has effectively restricted the transmission of Human Immunodeficiency Virus (HIV) and improved overall clinical outcomes. However, a complete cure for HIV remains out of reach, as the virus persists in a stable pool of infected cell reservoir that is resistant to therapy and thus a main barrier towards complete elimination of viral infection. While the mechanisms by which host proteins govern viral gene expression and latency are well-studied, the emerging regulatory functions of non-coding RNAs (ncRNA) in the context of T cell activation, HIV gene expression and viral latency have not yet been thoroughly explored. Here, we report the identification of the Cytoskeleton Regulator (CYTOR) long non-coding RNA (lncRNA) as an activator of HIV gene expression that is upregulated following T cell stimulation. Functional studies show that CYTOR suppresses viral latency by directly binding to the HIV promoter and associating with the cellular positive transcription elongation factor (P-TEFb) to activate viral gene expression. CYTOR also plays a global role in regulating cellular gene expression, including those involved in controlling actin dynamics. Depletion of CYTOR expression reduces cytoplasmic actin polymerization in response to T cell activation. In addition, treating HIV-infected cells with pharmacological inhibitors of actin polymerization reduces HIV gene expression. We conclude that both direct and indirect effects of CYTOR regulate HIV gene expression.


Asunto(s)
Regulación Viral de la Expresión Génica , Infecciones por VIH , VIH-1 , ARN Largo no Codificante , Latencia del Virus , Humanos , ARN Largo no Codificante/genética , Infecciones por VIH/virología , Infecciones por VIH/genética , VIH-1/genética , VIH-1/fisiología , Regiones Promotoras Genéticas , Activación de Linfocitos , Células Jurkat
11.
Virology ; 595: 110065, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38569227

RESUMEN

Nucleot(s)ide analogues, the current antiviral treatments against chronic hepatitis B (CHB) infection, are non-curative due to their inability to eliminate covalently closed circular DNA (cccDNA) from the infected hepatocytes. Preclinical studies have shown that coumarin derivatives can effectively reduce the HBV DNA replication. We evaluated the antiviral efficacy of thirty new coumarin derivatives in cell culture models for studying HBV. Furanocoumarins Fc-20 and Fc-31 suppressed the levels of pre-genomic RNA as well as cccDNA, and reduced the secretion of virions, HBsAg and HBeAg. The antiviral efficacies of Fc-20 and Fc31 improved further when used in combination with the hepatitis B antiviral drug Entecavir. There was a marked reduction in the intracellular HBx level in the presence of these furanocoumarins due to proteasomal degradation resulting in the down-regulation of HBx-dependent viral genes. Importantly, both Fc-20 and Fc-31 were non-cytotoxic to cells even at high concentrations. Further, our molecular docking studies confirmed a moderate to high affinity interaction between furanocoumarins and viral HBx via residues Ala3, Arg26 and Lys140. These data suggest that furanocoumarins could be developed as a new therapeutic for CHB infection.


Asunto(s)
Antivirales , ADN Circular , Furocumarinas , Virus de la Hepatitis B , Complejo de la Endopetidasa Proteasomal , Transactivadores , Proteínas Reguladoras y Accesorias Virales , Replicación Viral , Virus de la Hepatitis B/efectos de los fármacos , Virus de la Hepatitis B/genética , Virus de la Hepatitis B/fisiología , Virus de la Hepatitis B/metabolismo , Replicación Viral/efectos de los fármacos , Humanos , Transactivadores/metabolismo , Transactivadores/genética , ADN Circular/metabolismo , ADN Circular/genética , Proteínas Reguladoras y Accesorias Virales/metabolismo , Proteínas Reguladoras y Accesorias Virales/genética , Furocumarinas/farmacología , Antivirales/farmacología , Complejo de la Endopetidasa Proteasomal/metabolismo , ADN Viral/metabolismo , ADN Viral/genética , Regulación hacia Abajo/efectos de los fármacos , Transcripción Genética/efectos de los fármacos , Proteolisis/efectos de los fármacos , Regulación Viral de la Expresión Génica/efectos de los fármacos , Células Hep G2
12.
Viruses ; 16(4)2024 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-38675925

RESUMEN

The interferon-induced host cell protein Shiftless (SFL) inhibits -1 programmed ribosomal frameshifting (-1PRF) required for the expression of HIV-1 Gal-Pol and the formation of infectious HIV-1 particles. However, the specific regions in SFL required for antiviral activity and the mechanism by which SFL inhibits -1PRF remain unclear. Employing alanine scanning mutagenesis, we found that basic amino acids in the predicted zinc ribbon motif of SFL are essential for the suppression of Gag-Pol expression but dispensable for anti-HIV-1 activity. We have shown that SFL inhibits the expression of the murine leukemia virus (MLV) Gag-Pol polyprotein and the formation of infectious MLV particles, although Gag-Pol expression of MLV is independent of -1PRF but requires readthrough of a stop codon. These findings indicate that SFL might inhibit HIV-1 infection by more than one mechanism and that SFL might target programmed translational readthrough as well as -1PRF signals, both of which are regulated by mRNA secondary structure elements.


Asunto(s)
Proteínas de Fusión gag-pol , Infecciones por VIH , VIH-1 , Proteínas de Unión al ARN , Humanos , Sistema de Lectura Ribosómico , Proteínas de Fusión gag-pol/genética , Proteínas de Fusión gag-pol/metabolismo , Regulación Viral de la Expresión Génica , Células HEK293 , Infecciones por VIH/virología , Infecciones por VIH/genética , Infecciones por VIH/metabolismo , VIH-1/genética , VIH-1/fisiología , Virus de la Leucemia Murina/genética , Virus de la Leucemia Murina/fisiología , Replicación Viral , Proteínas de Unión al ARN/metabolismo
13.
Viruses ; 16(4)2024 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-38675926

RESUMEN

The transcription and replication of the Newcastle disease virus (NDV) strictly rely on the viral ribonucleoprotein (RNP) complex, which is composed of viral NP, P, L and RNA. However, it is not known whether other viral non-RNP proteins participate in this process for viral self-regulation. In this study, we used a minigenome (MG) system to identify the regulatory role of the viral non-RNP proteins V, M, W, F and HN. Among them, V significantly reduced MG-encoded reporter activity compared with the other proteins and inhibited the synthesis of viral mRNA and cRNA. Further, V interacted with NP. A mutation in residue W195 of V diminished V-NP interaction and inhibited inclusion body (IB) formation in NP-P-L-cotransfected cells. Furthermore, a reverse-genetics system for the highly virulent strain F48E9 was established. The mutant rF48E9-VW195R increased viral replication and apparently enhanced IB formation. In vivo experiments demonstrated that rF48E9-VW195R decreased virulence and retarded time of death. Overall, the results indicate that the V-NP interaction of the W195 mutant V decreased, which regulated viral RNA synthesis, IB formation, viral replication and pathogenicity. This study provides insight into the self-regulation of non-RNP proteins in paramyxoviruses.


Asunto(s)
Virus de la Enfermedad de Newcastle , Proteínas Virales , Replicación Viral , Virus de la Enfermedad de Newcastle/genética , Virus de la Enfermedad de Newcastle/fisiología , Virus de la Enfermedad de Newcastle/metabolismo , Animales , Proteínas Virales/metabolismo , Proteínas Virales/genética , Nucleoproteínas/metabolismo , Nucleoproteínas/genética , Enfermedad de Newcastle/virología , Enfermedad de Newcastle/metabolismo , Línea Celular , Regulación Viral de la Expresión Génica , ARN Viral/genética , ARN Viral/metabolismo , Pollos , Virulencia , Unión Proteica , Mutación
14.
Viruses ; 16(4)2024 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-38675950

RESUMEN

Hepatitis B virus (HBV) is the etiologic agent of chronic hepatitis B, which puts at least 300 million patients at risk of developing fibrosis, cirrhosis, and hepatocellular carcinoma. HBV is a partially double-stranded DNA virus of the Hepadnaviridae family. While HBV was discovered more than 50 years ago, many aspects of its replicative cycle remain incompletely understood. Central to HBV persistence is the formation of covalently closed circular DNA (cccDNA) from the incoming relaxed circular DNA (rcDNA) genome. cccDNA persists as a chromatinized minichromosome and is the major template for HBV gene transcription. Here, we review how cccDNA and the viral minichromosome are formed and how viral gene transcription is regulated and highlight open questions in this area of research.


Asunto(s)
ADN Circular , ADN Viral , Virus de la Hepatitis B , Replicación Viral , Virus de la Hepatitis B/genética , Virus de la Hepatitis B/fisiología , ADN Circular/genética , Humanos , ADN Viral/genética , Transcripción Viral/genética , Regulación Viral de la Expresión Génica , Transcripción Genética , Genoma Viral , Hepatitis B Crónica/virología , Hepatitis B/virología , Replicación del ADN
15.
Viruses ; 16(4)2024 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-38675956

RESUMEN

Chronic hepatitis B (CHB) virus infection is a major public health burden and the leading cause of hepatocellular carcinoma. Despite the efficacy of current treatments, hepatitis B virus (HBV) cannot be fully eradicated due to the persistence of its minichromosome, or covalently closed circular DNA (cccDNA). The HBV community is investing large human and financial resources to develop new therapeutic strategies that either silence or ideally degrade cccDNA, to cure HBV completely or functionally. cccDNA transcription is considered to be the key step for HBV replication. Transcription not only influences the levels of viral RNA produced, but also directly impacts their quality, generating multiple variants. Growing evidence advocates for the role of the co-transcriptional regulation of HBV RNAs during CHB and viral replication, paving the way for the development of novel therapies targeting these processes. This review focuses on the mechanisms controlling the different co-transcriptional processes that HBV RNAs undergo, and their contribution to both viral replication and HBV-induced liver pathogenesis.


Asunto(s)
Regulación Viral de la Expresión Génica , Virus de la Hepatitis B , ARN Viral , Replicación Viral , Virus de la Hepatitis B/genética , Virus de la Hepatitis B/fisiología , Humanos , ARN Viral/genética , Hepatitis B Crónica/virología , ADN Circular/genética , ADN Circular/metabolismo , Transcripción Genética , Animales , ADN Viral/genética
16.
J Virol ; 98(4): e0024224, 2024 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-38446633

RESUMEN

Viral genomes frequently harbor overlapping genes, complicating the development of virus-vectored vaccines and gene therapies. This study introduces a novel conditional splicing system to precisely control the expression of such overlapping genes through recombinase-mediated conditional splicing. We refined site-specific recombinase (SSR) conditional splicing systems and explored their mechanisms. The systems demonstrated exceptional inducibility (116,700-fold increase) with negligible background expression, facilitating the conditional expression of overlapping genes in adenovirus-associated virus (AAV) and human immunodeficiency virus type 1. Notably, this approach enabled the establishment of stable AAV producer cell lines, encapsulating all necessary packaging genes. Our findings underscore the potential of the SSR-conditional splicing system to significantly advance vector engineering, enhancing the efficacy and scalability of viral-vector-based therapies and vaccines. IMPORTANCE: Regulating overlapping genes is vital for gene therapy and vaccine development using viral vectors. The regulation of overlapping genes presents challenges, including cytotoxicity and impacts on vector capacity and genome stability, which restrict stable packaging cell line development and broad application. To address these challenges, we present a "loxp-splice-loxp"-based conditional splicing system, offering a novel solution for conditional expression of overlapping genes and stable cell line establishment. This system may also regulate other cytotoxic genes, representing a significant advancement in cell engineering and gene therapy as well as biomass production.


Asunto(s)
Dependovirus , Genes Sobrepuestos , Genes Virales , Ingeniería Genética , VIH-1 , Empalme del ARN , Humanos , Línea Celular , Dependovirus/genética , ADN Nucleotidiltransferasas/genética , ADN Nucleotidiltransferasas/metabolismo , Regulación Viral de la Expresión Génica , Genes Sobrepuestos/genética , Genes Virales/genética , Ingeniería Genética/métodos , Terapia Genética/métodos , Vectores Genéticos/genética , VIH-1/genética , Empalme del ARN/genética , Vacunas/biosíntesis , Vacunas/genética , Empaquetamiento del Genoma Viral/genética
17.
PLoS Pathog ; 20(3): e1011998, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38530845

RESUMEN

Epstein-Barr virus (EBV) contributes to ~1% of all human cancers including several B-cell neoplasms. A characteristic feature of EBV life cycle is its ability to transform metabolically quiescent B-lymphocytes into hyperproliferating B-cell blasts with the establishment of viral latency, while intermittent lytic cycle induction is necessary for the production of progeny virus. Our RNA-Seq analyses of both latently infected naïve B-lymphocytes and transformed B-lymphocytes upon lytic cycle replication indicate a contrasting expression pattern of a membrane-associated carbonic anhydrase isoform CA9, an essential component for maintaining cell acid-base homeostasis. We show that while CA9 expression is transcriptionally activated during latent infection model, lytic cycle replication restrains its expression. Pharmacological inhibition of CA-activity using specific inhibitors retards EBV induced B-cell transformation, inhibits B-cells outgrowth and colony formation ability of transformed B-lymphocytes through lowering the intracellular pH, induction of cell apoptosis and facilitating degradation of CA9 transcripts. Reanalyses of ChIP-Seq data along with utilization of EBNA2 knockout virus, ectopic expression of EBNA2 and sh-RNA mediated knockdown of CA9 expression we further demonstrate that EBNA2 mediated CA9 transcriptional activation is essential for EBV latently infected B-cell survival. In contrast, during lytic cycle reactivation CA9 expression is transcriptionally suppressed by the key EBV lytic cycle transactivator, BZLF1 through its transactivation domain. Overall, our study highlights the dynamic alterations of CA9 expression and its activity in regulating pH homeostasis act as one of the major drivers for EBV induced B-cell transformation and subsequent B-cell lymphomagenesis.


Asunto(s)
Infecciones por Virus de Epstein-Barr , Herpesvirus Humano 4 , Humanos , Herpesvirus Humano 4/fisiología , Infecciones por Virus de Epstein-Barr/genética , Linfocitos B , Latencia del Virus , Transactivadores/genética , Activación Viral , Regulación Viral de la Expresión Génica
18.
J Virol ; 98(4): e0201523, 2024 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-38451083

RESUMEN

Herpes simplex virus 1 (HSV-1) transcription is restricted in latently infected neurons and the genomes are in mostly silenced chromatin, whereas all viral genes are transcribed in lytically infected cells, in which the genomes are dynamically chromatinized. Epigenetic regulation modulates HSV-1 transcription during lytic, latent, and reactivating infections but the precise mechanisms are not fully defined. Nucleosomes are dynamic: they slide, breathe, assemble, and disassemble. We and others have proposed that the most dynamic HSV-1 chromatin is transcriptionally competent, whereas the least dynamic is silenced. However, the mechanisms yielding the unusually dynamic viral chromatin remain unknown. Histone variants affect nucleosome dynamics. The dynamics of H2A, H2A.X, and macroH2A were enhanced in infected cells, whereas those of H2A.B were uniquely decreased. We constructed stably transduced cells expressing tagged histone H2A, H2A.B, macroH2A, or H2B, which assembles the H2A/H2B nucleosome dimers with all H2A variants. All H2A variants, as well as ectopic and endogenous H2B were assembled into HSV-1 chromatin evenly throughout the genome but canonical H2A was relatively depleted whereas H2A.B was enriched, particularly in the most dynamic viral chromatin. When viral transcription and DNA replication were restricted, H2A.B became as depleted from the viral chromatin through the entire genome as H2A. We propose that lytic HSV-1 nucleosomes are enriched in the dynamic variant H2A.B/H2B dimers to promote HSV-1 chromatin dynamics and transcriptional competency and conclude that the dynamics of HSV-1 chromatin are determined in part by the H2A variants. IMPORTANCE: Herpes simplex virus 1 (HSV-1) transcription is epigenetically regulated during latent and lytic infections, and epigenetic inhibitors have been proposed as potential antiviral drugs to modulate latency and reactivation. However, the detailed epigenetic mechanisms of regulation of HSV-1 transcription have not been fully characterized and may differ from those regulating cellular transcription. Whereas lytic HSV-1 chromatin is unusually dynamic, latent silenced HSV-1 chromatin is not. The mechanisms resulting in the unique dynamics of the lytic chromatin remain unknown. Here we identify the enrichment of the highly dynamic histone 2A variant H2A in the most dynamic viral chromatin, which provides a mechanistic understanding of its unique dynamics. Future work to identify the mechanisms of enrichment in H2A.B on the viral chromatin may identify novel druggable epigenetic regulators that modulate HSV-1 latency and reactivation.


Asunto(s)
Cromatina , Epigénesis Genética , Regulación Viral de la Expresión Génica , Herpesvirus Humano 1 , Histonas , Transcripción Viral , Replicación Viral , Cromatina/genética , Cromatina/metabolismo , Silenciador del Gen , Variación Genética , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/crecimiento & desarrollo , Herpesvirus Humano 1/fisiología , Histonas/genética , Histonas/metabolismo , Nucleosomas/genética , Nucleosomas/metabolismo , Activación Viral , Latencia del Virus , Humanos , Animales , Células Vero , Células HEK293
19.
J Virol ; 98(3): e0150223, 2024 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-38315015

RESUMEN

Capsid assembly is critical in the hepatitis B virus (HBV) life cycle, mediated by the viral core protein. Capsid assembly is the target for new anti-viral therapeutics known as capsid assembly modulators (CAMs) of which the CAM-aberrant (CAM-A) class induces aberrant shaped core protein structures and leads to hepatocyte cell death. This study aimed to identify the mechanism of action of CAM-A modulators leading to HBV-infected hepatocyte elimination where CAM-A-mediated hepatitis B surface antigen (HBsAg) reduction was evaluated in a stable HBV replicating cell line and in AAV-HBV-transduced C57BL/6, C57BL/6 SCID, and HBV-infected chimeric mice with humanized livers. Results showed that in vivo treatment with CAM-A modulators induced pronounced reductions in hepatitis B e antigen (HBeAg) and HBsAg, associated with a transient alanine amino transferase (ALT) increase. Both HBsAg and HBeAg reductions and ALT increase were delayed in C57BL/6 SCID and chimeric mice, suggesting that adaptive immune responses may indirectly contribute. However, CD8+ T cell depletion in transduced wild-type mice did not impact antigen reduction, indicating that CD8+ T cell responses are not essential. Transient ALT elevation in AAV-HBV-transduced mice coincided with a transient increase in endoplasmic reticulum stress and apoptosis markers, followed by detection of a proliferation marker. Microarray data revealed antigen presentation pathway (major histocompatibility complex class I molecules) upregulation, overlapping with the apoptosis. Combination treatment with HBV-specific siRNA demonstrated that CAM-A-mediated HBsAg reduction is dependent on de novo core protein translation. To conclude, CAM-A treatment eradicates HBV-infected hepatocytes with high core protein levels through the induction of apoptosis, which can be a promising approach as part of a regimen to achieve functional cure. IMPORTANCE: Treatment with hepatitis B virus (HBV) capsid assembly modulators that induce the formation of aberrant HBV core protein structures (CAM-A) leads to programmed cell death, apoptosis, of HBV-infected hepatocytes and subsequent reduction of HBV antigens, which differentiates CAM-A from other CAMs. The effect is dependent on the de novo synthesis and high levels of core protein.


Asunto(s)
Antivirales , Apoptosis , Regulación Viral de la Expresión Génica , Antígenos del Núcleo de la Hepatitis B , Virus de la Hepatitis B , Hepatocitos , Biosíntesis de Proteínas , Animales , Ratones , Antivirales/farmacología , Antivirales/uso terapéutico , Apoptosis/efectos de los fármacos , Cápside/química , Cápside/clasificación , Cápside/efectos de los fármacos , Cápside/metabolismo , Proteínas de la Cápside/metabolismo , Hepatitis B/tratamiento farmacológico , Hepatitis B/inmunología , Hepatitis B/metabolismo , Hepatitis B/virología , Antígenos del Núcleo de la Hepatitis B/biosíntesis , Antígenos del Núcleo de la Hepatitis B/metabolismo , Antígenos e de la Hepatitis B/metabolismo , Antígenos de Superficie de la Hepatitis B/metabolismo , Virus de la Hepatitis B/crecimiento & desarrollo , Virus de la Hepatitis B/inmunología , Virus de la Hepatitis B/metabolismo , Virus de la Hepatitis B/patogenicidad , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Hepatocitos/patología , Hepatocitos/virología , Ratones Endogámicos C57BL , Ratones SCID , Replicación Viral , Línea Celular , Linfocitos T CD8-positivos/inmunología , Presentación de Antígeno
20.
J Virol ; 98(3): e0151523, 2024 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-38323812

RESUMEN

Adeno-associated virus (AAV) requires co-infection with helper virus for efficient replication. We previously reported that Human Bocavirus 1 (HBoV1) genes, including NP1, NS2, and BocaSR, were critical for AAV2 replication. Here, we first demonstrate the essential roles of the NP1 protein in AAV2 DNA replication and protein expression. We show that NP1 binds to single-strand DNA (ssDNA) at least 30 nucleotides (nt) in length in a sequence-independent manner. Furthermore, NP1 colocalized with the BrdU-labeled AAV2 DNA replication center, and the loss of the ssDNA-binding ability of NP1 by site-directed mutation completely abolished AAV2 DNA replication. We used affinity-tagged NP1 protein to identify host cellular proteins associated with NP1 in cells cotransfected with the HBoV1 helper genes and AAV2 duplex genome. Of the identified proteins, we demonstrate that NP1 directly binds to the DBD-F domain of the RPA70 subunit with a high affinity through the residues 101-121. By reconstituting the heterotrimer protein RPA in vitro using gel filtration, we demonstrate that NP1 physically associates with RPA to form a heterologous complex characterized by typical fast-on/fast-off kinetics. Following a dominant-negative strategy, we found that NP1-RPA complex mainly plays a role in expressing AAV2 capsid protein by enhancing the transcriptional activity of the p40 promoter. Our study revealed a novel mechanism by which HBoV1 NP1 protein supports AAV2 DNA replication and capsid protein expression through its ssDNA-binding ability and direct interaction with RPA, respectively.IMPORTANCERecombinant adeno-associated virus (rAAV) vectors have been extensively used in clinical gene therapy strategies. However, a limitation of these gene therapy strategies is the efficient production of the required vectors, as AAV alone is replication-deficient in the host cells. HBoV1 provides the simplest AAV2 helper genes consisting of NP1, NS2, and BocaSR. An important question regarding the helper function of HBoV1 is whether it provides any direct function that supports AAV2 DNA replication and protein expression. Also of interest is how HBoV1 interplays with potential host factors to constitute a permissive environment for AAV2 replication. Our studies revealed that the multifunctional protein NP1 plays important roles in AAV2 DNA replication via its sequence-independent ssDNA-binding ability and in regulating AAV2 capsid protein expression by physically interacting with host protein RPA. Our findings present theoretical guidance for the future application of the HBoV1 helper genes in the rAAV vector production.


Asunto(s)
Proteínas de la Cápside , Cápside , ADN de Cadena Simple , ADN Viral , Proteínas de Unión al ADN , Dependovirus , Bocavirus Humano , Proteínas Virales , Humanos , Cápside/metabolismo , Proteínas de la Cápside/biosíntesis , Proteínas de la Cápside/química , Proteínas de la Cápside/genética , Proteínas de la Cápside/metabolismo , Dependovirus/genética , Dependovirus/crecimiento & desarrollo , Dependovirus/metabolismo , ADN de Cadena Simple/biosíntesis , ADN de Cadena Simple/metabolismo , ADN Viral/biosíntesis , ADN Viral/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Regulación Viral de la Expresión Génica , Bocavirus Humano/genética , Bocavirus Humano/metabolismo , Cinética , Mutagénesis Sitio-Dirigida , Mutación , Regiones Promotoras Genéticas , Unión Proteica , Dominios Proteicos , Proteínas Virales/genética , Proteínas Virales/metabolismo , Replicación Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA