Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 14.373
Filter
1.
Viruses ; 16(6)2024 May 21.
Article in English | MEDLINE | ID: mdl-38932109

ABSTRACT

Climate change, unpredictable weather patterns, and droughts are depleting water resources in some parts of the globe, where recycling and reusing wastewater is a strategy for different purposes. To counteract this, the EU regulation for water reuse sets minimum requirements for the use of reclaimed water for agricultural irrigation, including a reduction in human enteric viruses. In the present study, the occurrence of several human enteric viruses, including the human norovirus genogroup I (HuNoV GI), HuNoV GII, and rotavirus (RV), along with viral fecal contamination indicator crAssphage was monitored by using (RT)-qPCR methods on influent wastewater and reclaimed water samples. Moreover, the level of somatic coliphages was also determined as a culturable viral indicator. To assess the potential viral infectivity, an optimization of a capsid integrity PMAxx-RT-qPCR method was performed on sewage samples. Somatic coliphages were present in 60% of the reclaimed water samples, indicating inefficient virus inactivation. Following PMAxx-RT-qPCR optimization, 66% of the samples tested positive for at least one of the analyzed enteric viruses, with concentrations ranging from 2.79 to 7.30 Log10 genome copies (gc)/L. Overall, most of the analyzed reclaimed water samples did not comply with current EU legislation and contained potential infectious viral particles.


Subject(s)
Sewage , Wastewater , Wastewater/virology , Sewage/virology , Humans , Capsid , Coliphages/isolation & purification , Coliphages/genetics , Coliphages/classification , Rotavirus/genetics , Rotavirus/isolation & purification , Norovirus/isolation & purification , Norovirus/genetics , Water Microbiology , Real-Time Polymerase Chain Reaction , Feces/virology , Enterovirus/isolation & purification , Enterovirus/genetics , Enterovirus/classification , Capsid Proteins/genetics
2.
Protein Sci ; 33(7): e5074, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38888268

ABSTRACT

Adeno-associated virus (AAV), a widely used gene therapy vector, is a small, nonenveloped virus that contains a single-stranded DNA genome with a maximum length of 4.7 kb. Despite extensive biophysical and structural characterization, many aspects of AAV functions remain elusive. This knowledge gap is primarily due to a lack of structurally resolved dynamic information and the absence of structural coverage of functionally critical segments on the AAV capsid. Here, we developed a protocol to study AAV structural dynamics by hydrogen-deuterium exchange mass spectrometry (HDX-MS), a powerful method for monitoring protein structure stability and dynamics in solution. We performed HDX-MS measurements on AAVs without or with different DNA payloads of different sizes, and obtained detailed dynamic information on the entire AAV sequence including the two functionally important segments not previously structurally characterized. The unique N terminus of the capsid protein VP1 (VP1u) was found to adopt a highly dynamic and unstable conformation with low HDX protection across the entire region, whereas the presence of a DNA payload increased its protection. The VP1 and VP2 shared region (VP1/2) showed no measurable protection, with or without DNA. Differential HDX between empty and full capsid samples allowed us to identify potential new DNA-capsid interaction sites located primarily around the five-fold channel, which differ from the three-fold pocket binding site previously identified. Our HDX-MS method for characterizing AAV structural dynamics opens a new way for future efforts to understand AAV structure-function relationships and engineer next-generation AAV vectors with improved gene delivery properties.


Subject(s)
Capsid Proteins , Capsid , Dependovirus , Genetic Therapy , Genetic Vectors , Dependovirus/genetics , Dependovirus/chemistry , Capsid Proteins/chemistry , Capsid Proteins/genetics , Capsid Proteins/metabolism , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Genetic Vectors/genetics , Genetic Therapy/methods , Capsid/chemistry , Capsid/metabolism , Hydrogen Deuterium Exchange-Mass Spectrometry , Protein Stability , Humans , Protein Conformation , Models, Molecular
3.
Structure ; 32(6): 652-653, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38848682

ABSTRACT

In a recent issue of Nature, Coshic et al. employ a computational multiscale approach to package the complete HK97 viral genome into its capsid. They find both good agreement with experimental observations and shed new light on the heterogeneity of genome structures and the mechanism by which they package.


Subject(s)
Capsid , Genome, Viral , Capsid/metabolism , Capsid/chemistry , Capsid Proteins/chemistry , Capsid Proteins/metabolism , Capsid Proteins/genetics , Virus Assembly , Molecular Dynamics Simulation , Models, Molecular
4.
Viruses ; 16(6)2024 May 21.
Article in English | MEDLINE | ID: mdl-38932107

ABSTRACT

Rotaviruses (RVs) are 11-segmented, double-stranded (ds) RNA viruses and important causes of acute gastroenteritis in humans and other animal species. Early RV particle assembly is a multi-step process that includes the assortment, packaging and replication of the 11 genome segments in close connection with capsid morphogenesis. This process occurs inside virally induced, cytosolic, membrane-less organelles called viroplasms. While many viral and cellular proteins play roles during early RV assembly, the octameric nonstructural protein 2 (NSP2) has emerged as a master orchestrator of this key stage of the viral replication cycle. NSP2 is critical for viroplasm biogenesis as well as for the selective RNA-RNA interactions that underpin the assortment of 11 viral genome segments. Moreover, NSP2's associated enzymatic activities might serve to maintain nucleotide pools for use during viral genome replication, a process that is concurrent with early particle assembly. The goal of this review article is to summarize the available data about the structures, functions and interactions of RV NSP2 while also drawing attention to important unanswered questions in the field.


Subject(s)
Genome, Viral , Rotavirus , Viral Nonstructural Proteins , Virus Assembly , Virus Replication , Rotavirus/genetics , Rotavirus/physiology , Viral Nonstructural Proteins/metabolism , Viral Nonstructural Proteins/genetics , Humans , Animals , RNA, Viral/genetics , RNA, Viral/metabolism , Capsid/metabolism , RNA-Binding Proteins
5.
Article in English | MEDLINE | ID: mdl-38908134

ABSTRACT

The field of recombinant adeno-associated virus (rAAV) gene therapy has attracted increasing attention over decades. Within the ongoing challenges of rAAV manufacturing, the co-production of impurities, such as empty and partial capsids containing no or truncated transgenes, poses a significant challenge. Due to their potential impact on drug efficacy and clinical safety, it is imperative to conduct comprehensive monitoring and characterization of these impurities prior to the release of the final gene therapy product. Nevertheless, existing analytical techniques encounter notable limitations, encompassing low throughput, long turnaround times, high sample consumption, and/or complicated data analysis. Chromatography-based analytical methods are recognized for their current Good Manufacturing Practice (cGMP) alignment, high repeatability, reproducibility, low limit of detection, and rapid turnaround times. Despite these advantages, current anion exchange high pressure liquid chromatography (AEX-HPLC) methods struggle with baseline separation of partial capsids from full and empty capsids, resulting in inaccurate full-to-empty capsid ratio, as partial capsids are obscured within peaks corresponding to empty and full capsids. In this study, we present a unique analytical AEX method designed to characterize not only empty and full capsids but also partial capsids. This method utilizes continuous N-Rich chromatography with recycling between two identical AEX columns for the accumulation and isolation of partial capsids. The development process is comprehensively discussed, covering the preparation of reference materials representing full (rAAV-LacZ), partial (rAAV-GFP), and empty (rAAV-empty) capsids, N-rich method development, fraction analysis, determination of fluorescence response factors between capsid variants, and validation through comparison with other comparative techniques.


Subject(s)
Capsid , Dependovirus , Dependovirus/genetics , Dependovirus/isolation & purification , Chromatography, Ion Exchange/methods , Capsid/chemistry , Chromatography, High Pressure Liquid/methods , Reproducibility of Results
6.
Nat Microbiol ; 9(7): 1842-1855, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38918469

ABSTRACT

The viral nuclear egress complex (NEC) allows herpesvirus capsids to escape from the nucleus without compromising the nuclear envelope integrity. The NEC lattice assembles on the inner nuclear membrane and mediates the budding of nascent nucleocapsids into the perinuclear space and their subsequent release into the cytosol. Its essential role makes it a potent antiviral target, necessitating structural information in the context of a cellular infection. Here we determined structures of NEC-capsid interfaces in situ using electron cryo-tomography, showing a substantial structural heterogeneity. In addition, while the capsid is associated with budding initiation, it is not required for curvature formation. By determining the NEC structure in several conformations, we show that curvature arises from an asymmetric assembly of disordered and hexagonally ordered lattice domains independent of pUL25 or other viral capsid vertex components. Our results advance our understanding of the mechanism of nuclear egress in the context of a living cell.


Subject(s)
Capsid , Cell Nucleus , Cryoelectron Microscopy , Nuclear Envelope , Virus Release , Cell Nucleus/metabolism , Cell Nucleus/virology , Humans , Nuclear Envelope/metabolism , Capsid/metabolism , Capsid Proteins/metabolism , Capsid Proteins/genetics , Nucleocapsid/metabolism , Electron Microscope Tomography , Viral Proteins/metabolism , Viral Proteins/genetics , Herpesviridae/physiology , Herpesviridae/genetics
7.
J Am Soc Mass Spectrom ; 35(7): 1567-1575, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38888112

ABSTRACT

Adeno-associated viruses (AAVs) have emerged as a leading platform for in vivo therapeutic gene delivery and offer tremendous potential in the treatment and prevention of human disease. The fast-paced development of this growing class of therapeutics, coupled with their intrinsic structural complexity, places a high demand on analytical methods capable of efficiently monitoring product quality to ensure safety and efficacy, as well as to support manufacturing and process optimization. Importantly, the presence and relative abundance of both empty and partially filled AAV capsid subpopulations are of principal concern, as these represent the most common product-related impurities in AAV manufacturing and have a direct impact on therapeutic potential. For this reason, the capsid content, or ratio of empty and partial capsids to those packaged with the full-length therapeutic genome, has been identified by regulatory agencies as a critical quality attribute (CQA) that must be carefully controlled to meet clinical specifications. Established analytical methods for the quantitation of capsid content ratios often suffer from long turnaround times, low throughput, and high sample demands that are not well-suited to the narrow timelines and limited sample availability typical of process development. In this study, we present an integrated online native mass spectrometry platform that aims to minimize sample handling and maximize throughput and robustness for rapid and sensitive quantitation of AAV capsid content ratios. The primary advantages of this platform for AAV analysis include the ability to perform online buffer exchange under low flow conditions to maintain sample stability with minimal sample dilution, as well as the ability to achieve online charge reduction via dopant-modified desolvation gas. By exploiting the latter, enhanced spectral resolution of signals arising from empty, partial, and full AAV capsids was accomplished in the m/z domain to facilitate improved spectral interpretation and quantitation that correlated well with the industry standard analytical ultracentrifugation (AUC) method for capsid content ratio determination. The utility of this approach was further demonstrated in several applications, including the rapid and universal screening of different AAV serotypes, evaluation of capsid content for in-process samples, and the monitoring of capsid stability when subjected to thermal stress conditions.


Subject(s)
Capsid Proteins , Capsid , Dependovirus , Dependovirus/chemistry , Capsid Proteins/analysis , Capsid Proteins/chemistry , Capsid/chemistry , Humans , Mass Spectrometry/methods
8.
Cell Host Microbe ; 32(6): 980-995.e9, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38729153

ABSTRACT

Mammalian orthoreovirus (reovirus) is a nonenveloped virus that establishes primary infection in the intestine and disseminates to sites of secondary infection, including the CNS. Reovirus entry involves multiple engagement factors, but how the virus disseminates systemically and targets neurons remains unclear. In this study, we identified murine neuropilin 1 (mNRP1) as a receptor for reovirus. mNRP1 binds reovirus with nanomolar affinity using a unique mechanism of virus-receptor interaction, which is coordinated by multiple interactions between distinct reovirus capsid subunits and multiple NRP1 extracellular domains. By exchanging essential capsid protein-encoding gene segments, we determined that the multivalent interaction is mediated by outer-capsid protein σ3 and capsid turret protein λ2. Using capsid mutants incapable of binding NRP1, we found that NRP1 contributes to reovirus dissemination and neurovirulence in mice. Collectively, our results demonstrate that NRP1 is an entry receptor for reovirus and uncover mechanisms by which NRPs promote viral entry and pathogenesis.


Subject(s)
Capsid Proteins , Neuropilin-1 , Orthoreovirus, Mammalian , Receptors, Virus , Reoviridae Infections , Virus Internalization , Animals , Mice , Capsid Proteins/metabolism , Capsid Proteins/genetics , Neuropilin-1/metabolism , Neuropilin-1/genetics , Orthoreovirus, Mammalian/genetics , Orthoreovirus, Mammalian/physiology , Orthoreovirus, Mammalian/metabolism , Reoviridae Infections/virology , Reoviridae Infections/metabolism , Receptors, Virus/metabolism , Humans , Capsid/metabolism , Cell Line , HEK293 Cells , Protein Binding , Mice, Inbred C57BL
9.
Nucleic Acids Res ; 52(11): 6518-6531, 2024 Jun 24.
Article in English | MEDLINE | ID: mdl-38783157

ABSTRACT

Precise genomic editing through the combination of CRISPR/Cas systems and recombinant adeno-associated virus (rAAV)-delivered homology directed repair (HDR) donor templates represents a powerful approach. However, the challenge of effectively suppressing leaky transcription from the rAAV vector, a phenomenon associated to cytotoxicity, persists. In this study, we demonstrated substantial promoter activities of various homology arms and inverted terminal repeats (ITR). To address this issue, we identified a novel rAAV variant, Y704T, which not only yields high-vector quantities but also effectively suppresses in cis mRNA transcription driven by a robust promoter. The Y704T variant maintains normal functionality in receptor interaction, intracellular trafficking, nuclear entry, uncoating, and second-strand synthesis, while specifically exhibiting defects in transcription. Importantly, this inhibitory effect is found to be independent of ITR, promoter types, and RNA polymerases. Mechanistic studies unveiled the involvement of Valosin Containing Protein (VCP/p97) in capsid-mediated transcription repression. Remarkably, the Y704T variant delivers HDR donor templates without compromising DNA replication ability and homologous recombination efficiency. In summary, our findings enhance the understanding of capsid-regulated transcription and introduce novel avenues for the application of the rAAV-CRISPR/Cas9 system in human gene therapy.


Subject(s)
Dependovirus , Gene Editing , Homologous Recombination , Promoter Regions, Genetic , Dependovirus/genetics , Humans , Promoter Regions, Genetic/genetics , Gene Editing/methods , Homologous Recombination/genetics , HEK293 Cells , Capsid Proteins/genetics , Capsid Proteins/metabolism , Capsid/metabolism , Mutation , Genetic Vectors/genetics , Transcription, Genetic , CRISPR-Cas Systems , Recombinational DNA Repair , Terminal Repeat Sequences/genetics , DNA Replication/genetics
10.
Hum Gene Ther ; 35(11-12): 401-411, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38717948

ABSTRACT

Currently, adeno-associated virus (AAV) is one of the primary gene delivery vectors in gene therapy, facilitating long-term in vivo gene expression. Despite being imperative, it is incredibly challenging to precisely assess AAV particle distribution according to the sedimentation coefficient and identify impurities related to capsid structures. This study performed the systematic methodological validation of quantifying the AAV empty and full capsid ratio. This includes specificity, accuracy, precision, linearity, and parameter variables involving the sedimentation velocity analytical ultracentrifugation (SV-AUC) method. Specifically, SV-AUC differentiated among the empty, partial, full, and high sedimentation coefficient substance (HSCS) AAV particles while evaluating their sedimentation heterogeneity. The intermediate precision analysis of HE (high percentage of empty capsid) and HF (high percentage of full capsid) samples revealed that the specific species percentage, such as empty or full, was more significant than 50%. Moreover, the relative standard deviation (RSD) could be within 5%. Even for empty or partially less than 15%, the RSD could be within 10%. The accuracy recovery rates of empty capsid were between 103.9% and 108.7% across three different mixtures. When the measured percentage of specific species was more significant than 14%, the recovery rate was between 77.9% and 106.6%. Linearity analysis revealed an excellent linear correlation between the empty, partial, and full in the HE samples. The AAV samples with as low as 7.4 × 1011 cp/mL AAV could be accurately quantified with SV-AUC. The parameter variable analyses revealed that variations in cell alignment significantly affected the overall results. Still, the detection wavelength of 235 nm slightly influenced the empty, partial, and full percentages. Minor detection wavelength changes showed no impact on the sedimentation coefficient of these species. However, the temperature affected the measured sedimentation coefficient. These results validated the SV-AUC method to quantify AAV. This study provides solutions to AAV empty and full capsid ratio quantification challenges and the subsequent basis for calibrating the AAV empty capsid system suitability substance. Because of the AAV structure and potential variability complexity in detection, we jointly calibrated empty capsid system suitability substance with three laboratories to accurately detect the quantitative AAV empty and full capsid ratio. The empty capsid system suitability substance could be used as an external reference to measure the performance of the instrument. The results could be compared with multiple QC (quality control) laboratories based on the AAV vector and calibration accuracy. This is crucial for AUC to be used for QC release and promote gene therapy research worldwide.


Subject(s)
Dependovirus , Genetic Vectors , Ultracentrifugation , Dependovirus/genetics , Ultracentrifugation/methods , Humans , Genetic Vectors/genetics , Genetic Vectors/chemistry , Calibration , Genetic Therapy/methods , Capsid/chemistry , HEK293 Cells
11.
ACS Synth Biol ; 13(6): 1842-1850, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38729919

ABSTRACT

In-cell self-assembly of natural viral capsids is an event that can be visualized under transmission electron microscopy (TEM) observations. By mimicking the self-assembly of natural viral capsids, various artificial protein- and peptide-based nanocages were developed; however, few studies have reported the in-cell self-assembly of such nanocages. Our group developed a ß-Annulus peptide that can form a nanocage called artificial viral capsid in vitro, but in-cell self-assembly of the capsid has not been achieved. Here, we designed an artificial viral capsid decorated with a fluorescent protein, StayGold, to visualize in-cell self-assembly. Fluorescence anisotropy measurements and fluorescence resonance energy transfer imaging, in addition to TEM observations of the cells and super-resolution microscopy, revealed that StayGold-conjugated ß-Annulus peptides self-assembled into the StayGold-decorated artificial viral capsid in a cell. Using these techniques, we achieved the in-cell self-assembly of an artificial viral capsid.


Subject(s)
Capsid Proteins , Capsid , Fluorescence Resonance Energy Transfer , Peptides , Peptides/chemistry , Capsid/chemistry , Capsid/metabolism , Capsid Proteins/chemistry , Capsid Proteins/metabolism , Capsid Proteins/genetics , Luminescent Proteins/chemistry , Luminescent Proteins/metabolism , Luminescent Proteins/genetics , Microscopy, Electron, Transmission , Fluorescence Polarization , Virus Assembly
12.
Science ; 384(6701): 1220-1227, 2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38753766

ABSTRACT

Developing vehicles that efficiently deliver genes throughout the human central nervous system (CNS) will broaden the range of treatable genetic diseases. We engineered an adeno-associated virus (AAV) capsid, BI-hTFR1, that binds human transferrin receptor (TfR1), a protein expressed on the blood-brain barrier. BI-hTFR1 was actively transported across human brain endothelial cells and, relative to AAV9, provided 40 to 50 times greater reporter expression in the CNS of human TFRC knockin mice. The enhanced tropism was CNS-specific and absent in wild-type mice. When used to deliver GBA1, mutations of which cause Gaucher disease and are linked to Parkinson's disease, BI-hTFR1 substantially increased brain and cerebrospinal fluid glucocerebrosidase activity compared with AAV9. These findings establish BI-hTFR1 as a potential vector for human CNS gene therapy.


Subject(s)
Antigens, CD , Brain , Capsid , Gene Transfer Techniques , Genetic Vectors , Glucosylceramidase , Receptors, Transferrin , Animals , Humans , Mice , Antigens, CD/metabolism , Antigens, CD/genetics , Blood-Brain Barrier/metabolism , Brain/metabolism , Capsid/metabolism , Capsid Proteins/metabolism , Capsid Proteins/genetics , Dependovirus , Endothelial Cells/metabolism , Gene Knock-In Techniques , Genetic Therapy , Receptors, Transferrin/metabolism , Receptors, Transferrin/genetics , Glucosylceramidase/genetics , Gaucher Disease/genetics , Gaucher Disease/therapy , Parkinson Disease/genetics , Parkinson Disease/therapy
13.
Biol Pharm Bull ; 47(5): 912-916, 2024.
Article in English | MEDLINE | ID: mdl-38692868

ABSTRACT

The human herpesviruses (HHVs) are classified into the following three subfamilies: Alphaherpesvirinae, Betaherpesvirinae, and Gammaherpesvirinae. These HHVs have distinct pathological features, while containing a highly conserved viral replication pathway. Among HHVs, the basic viral particle structure and the sequential processes of viral replication are nearly identical. In particular, the capsid formation mechanism has been proposed to be highly similar among herpesviruses, because the viral capsid-organizing proteins are highly conserved at the structural and functional levels. Herpesviruses form capsids containing the viral genome in the nucleus of infected cells during the lytic phase, and release infectious virus (i.e., virions) to the cell exterior. In the capsid formation process, a single-unit-length viral genome is encapsidated into a preformed capsid. The single-unit-length viral genome is produced by cleavage from a viral genome precursor in which multiple unit-length viral genomes are tandemly linked. This encapsidation and cleavage is carried out by the terminase complex, which is composed of viral proteins. Since the terminase complex-mediated encapsidation and cleavage is a virus-specific mechanism that does not exist in humans, it may be an excellent inhibitory target for anti-viral drugs with high virus specificity. This review provides an overview of the functions of the terminase complexes of HHVs.


Subject(s)
Herpesviridae , Humans , Herpesviridae/physiology , Endodeoxyribonucleases/metabolism , Endodeoxyribonucleases/genetics , Viral Proteins/metabolism , Viral Proteins/genetics , Animals , Genome, Viral , Capsid/metabolism , Virus Replication
14.
Methods Mol Biol ; 2807: 153-161, 2024.
Article in English | MEDLINE | ID: mdl-38743227

ABSTRACT

Recent evidence has shown that uncoating and reverse transcription precede nuclear import. These recent breakthroughs have been made possible through the development of innovative biochemical and imaging techniques. This method outlines the biochemical assay used for detecting the presence of the HIV-1 core in the nuclear compartment. In this procedure, human cells are infected with HIV-1NL4-3, with or without the inclusion of PF74, a small molecule that inhibits core entry into the nuclear compartment. Subsequently, cells are separated into cytosolic and nuclear fractions. To assess whether the capsid protein has reached the nuclear compartment, cytosolic and nuclear fractions are subjected to Western blot analysis, utilizing antibodies specific to the HIV-1 capsid protein p24. To validate the true origin of these fractions, Western blot analysis employing antibodies against cytosolic and nuclear markers are also performed. In summary, this assay provides a reliable and efficient means to detect the presence of the HIV-1 capsid protein in the nucleus during infection under various conditions.


Subject(s)
Capsid , HIV Infections , HIV-1 , Humans , Blotting, Western/methods , Capsid/metabolism , Capsid Proteins/metabolism , Cell Line , Cell Nucleus/metabolism , HIV Core Protein p24/metabolism , HIV Core Protein p24/analysis , HIV Infections/virology , HIV Infections/metabolism , Phenylalanine/metabolism , Phenylalanine/analogs & derivatives
15.
J Virol ; 98(6): e0030524, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38771042

ABSTRACT

Many functions of viral attachment proteins are established, but less is known about the biological importance of viral attachment protein encapsidation efficiency. The mammalian orthoreovirus (reovirus) σ1 attachment protein forms filamentous trimers that incorporate into pentamers of the λ2 capsid protein. Reovirus strains vary in the efficiency of σ1 encapsidation onto progeny virions, which influences viral stability during entry into cells and the efficacy of tumor cell lysis. While the role of σ1 encapsidation has been evaluated in studies using cultured cells, the contribution of attachment protein encapsidation efficiency to viral infection in animals is less clear. Polymorphisms in reovirus σ1 at residues 22 and 249 have been implicated in viral dissemination in mice and susceptibility to proteolysis in the murine intestine, respectively. To determine whether these residues contribute to σ1 encapsidation efficiency, we engineered σ1 mutant viruses with single- and double-residue substitutions at sites 22 and 249. We found that substitutions at these sites alter the encapsidation of σ1 and that reoviruses encapsidating higher amounts of σ1 bind cells more avidly and have a modest replication advantage in a cell-type-specific manner relative to low σ1-encapsidating reoviruses. Furthermore, we found that a high σ1-encapsidating reovirus replicates and disseminates more efficiently in mice relative to a low σ1-encapsidating reovirus. These findings provide evidence of a relationship between viral attachment protein encapsidation efficiency and viral replication in cell culture and animal hosts. IMPORTANCE: Viral attachment proteins can serve multiple functions during viral replication, including attachment to host cells, cell entry and disassembly, and modulation of host immune responses. The relationship between viral attachment protein encapsidation efficiency and viral replication in cells and animals is poorly understood. We engineered and characterized a panel of reoviruses that differ in the capacity to encapsidate the σ1 attachment protein. We found that strains encapsidating σ1 with higher efficiency bind cells more avidly and replicate and spread more efficiently in mice relative to those encapsidating σ1 with lower efficiency. These results highlight a function for σ1 attachment protein capsid abundance in viral replication in cells and animals, which may inform future use of reovirus as an oncolytic therapeutic.


Subject(s)
Capsid Proteins , Reoviridae Infections , Virus Replication , Animals , Mice , Capsid Proteins/metabolism , Capsid Proteins/genetics , Reoviridae Infections/virology , Reoviridae Infections/metabolism , Virus Attachment , Polymorphism, Genetic , Orthoreovirus, Mammalian/genetics , Orthoreovirus, Mammalian/physiology , Orthoreovirus, Mammalian/metabolism , Virus Assembly , Cell Line , Capsid/metabolism , Humans
16.
J Virol ; 98(6): e0063324, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38775479

ABSTRACT

Adeno-associated viruses (AAVs) package a single-stranded (ss) DNA genome of 4.7 kb in their capsid of ~20 nm in diameter. AAV replication requires co-infection of a helper virus, such as adenovirus. During the optimization of recombinant AAV production, a small viral nonstructural protein, membrane-associated accessory protein (MAAP), was identified. However, the function of the MAAP in the context of AAV infection remains unknown. Here, we investigated the expression strategy and function of the MAAP during infection of both AAV2 and AAV5 in human embryonic kidney (HEK)293 cells. We found that AAV2 MAAP2 and AAV5 MAAP5 are expressed from the capsid gene (cap)-transcribing mRNA spliced from the donor to the second splice site that encodes VP2 and VP3. Thus, this AAV cap gene transcribes a multicistronic mRNA that can be translated to four viral proteins, MAAP, VP2, AAP, and VP3 in order. In AAV2 infection, MAAP2 predominantly localized in the cytoplasm, alongside the capsid, near the nuclear and plasma membranes, but a fraction of MAAP2 exhibited nuclear localization. In AAV5 infection, MAAP5 revealed a distinct pattern, predominantly localizing within the nucleus. In the cells infected with an MAAP knockout mutant of AAV2 or AAV5, both viral DNA replication and virus replication increased, whereas virus egress decreased, and the decrease in virus egress can be restored by providing MAAP in trans. In summary, MAAP, a novel AAV nonstructural protein translated from a multicistronic viral cap mRNA, not only facilitates cellular egress of AAV but also likely negatively affects viral DNA replication during infection. IMPORTANCE: Recombinant adeno-associated virus (rAAV) has been used as a gene delivery vector in clinical gene therapy. In current gene therapies employing rAAV, a high dose of the vector is required. Consequently, there is a high demand for efficient and high-purity vector production systems. In this study, we demonstrated that membrane-associated accessory protein (MAAP), a small viral nonstructural protein, is translated from the same viral mRNA transcript encoding VP2 and VP3. In AAV-infected cells, apart from its prevalent expression in the cytoplasm with localization near the plasma and nuclear membranes, the MAAP also exhibits notable localization within the nucleus. During AAV infection, MAAP expression increases the cellular egress of progeny virions and decreases viral DNA replication and progeny virion production. Thus, the choice of MAAP expression has pros and cons during AAV infection, which could provide a guide to rAAV production.


Subject(s)
Capsid Proteins , Dependovirus , Virus Replication , Humans , Dependovirus/genetics , Dependovirus/metabolism , HEK293 Cells , Capsid Proteins/metabolism , Capsid Proteins/genetics , Viral Nonstructural Proteins/metabolism , Viral Nonstructural Proteins/genetics , Parvoviridae Infections/virology , Parvoviridae Infections/metabolism , Capsid/metabolism
17.
Antimicrob Agents Chemother ; 68(7): e0042024, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38780261

ABSTRACT

Capsid assembly mediated by hepatitis B virus (HBV) core protein (HBc) is an essential part of the HBV replication cycle, which is the target for different classes of capsid assembly modulators (CAMs). While both CAM-A ("aberrant") and CAM-E ("empty") disrupt nucleocapsid assembly and reduce extracellular HBV DNA, CAM-As can also reduce extracellular HBV surface antigen (HBsAg) by triggering apoptosis of HBV-infected cells in preclinical mouse models. However, there have not been substantial HBsAg declines in chronic hepatitis B (CHB) patients treated with CAM-As to date. To investigate this disconnect, we characterized the antiviral activity of tool CAM compounds in HBV-infected primary human hepatocytes (PHHs), as well as in HBV-infected human liver chimeric mice and mice transduced with adeno-associated virus-HBV. Mechanistic studies in HBV-infected PHH revealed that CAM-A, but not CAM-E, induced a dose-dependent aggregation of HBc in the nucleus which is negatively regulated by the ubiquitin-binding protein p62. We confirmed that CAM-A, but not CAM-E, induced HBc-positive cell death in both mouse models via induction of apoptotic and inflammatory pathways and demonstrated that the degree of HBV-positive cell loss was positively correlated with intrahepatic HBc levels. Importantly, we determined that there is a significantly lower level of HBc per hepatocyte in CHB patient liver biopsies than in either of the HBV mouse models. Taken together, these data confirm that CAM-As have a unique secondary mechanism with the potential to kill HBc-positive hepatocytes. However, this secondary mechanism appears to require higher intrahepatic HBc levels than is typically observed in CHB patients, thereby limiting the therapeutic potential.


Subject(s)
Hepatitis B virus , Hepatitis B, Chronic , Hepatocytes , Humans , Hepatocytes/virology , Hepatocytes/drug effects , Animals , Hepatitis B virus/drug effects , Hepatitis B virus/physiology , Mice , Hepatitis B, Chronic/drug therapy , Hepatitis B, Chronic/virology , Viral Core Proteins/metabolism , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Hepatitis B Core Antigens/metabolism , Capsid/metabolism , Capsid/drug effects , Liver/virology , Liver/drug effects , Liver/metabolism , Hepatitis B Surface Antigens/metabolism , Virus Assembly/drug effects , Apoptosis/drug effects , Virus Replication/drug effects
18.
ACS Nano ; 18(21): 13755-13767, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38752610

ABSTRACT

The ability to manipulate the self-assembly of proteins is essential to understanding the mechanisms of life and beneficial to fabricating advanced nanomaterials. Here, we report the transformation of the MS2 phage capsid from nanocages to nanotubes and then to nanotube hydrogels through simple point mutations guided by interfacial interaction redesign. We demonstrate that site 70, which lies in the flexible FG loop of the capsid protein (CP), is a "magic" site that can largely dictate the final morphology of assemblies. By varying the amino acid at site 70, with the aid of a cysteine-to-alanine mutation at site 46, we achieved the assembly of double-helical or single-helical nanotubes in addition to nanocages. Furthermore, an additional cysteine substitution on the surface of nanotubes mediated their cross-linking to form hydrogels with reducing agent responsiveness. The hierarchical self-assembly system allowed for the investigation of morphology-related immunogenicity of MS2 CPs, which revealed dramatic differences among nanocages, nanotubes, and nanotube hydrogels in terms of immune response types, antibody levels and T cell functions. This study provides insights into the assembly manipulation of protein nanomaterials and the customized design of nanovaccines and drug delivery systems.


Subject(s)
Capsid Proteins , Capsid , Hydrogels , Nanotubes , Hydrogels/chemistry , Nanotubes/chemistry , Capsid Proteins/chemistry , Capsid Proteins/immunology , Capsid Proteins/genetics , Capsid/chemistry , Capsid/immunology , Levivirus/chemistry , Levivirus/immunology , Levivirus/genetics , Animals , Nanostructures/chemistry , Mice , Models, Molecular
19.
Viruses ; 16(5)2024 04 25.
Article in English | MEDLINE | ID: mdl-38793552

ABSTRACT

The HIV-1 capsid (CA) protein forms the outer shell of the viral core that is released into the cytoplasm upon infection. CA binds various cellular proteins, including CPSF6, that direct HIV-1 integration into speckle-associated domains in host chromatin. Upon HIV-1 infection, CPSF6 forms puncta in the nucleus. Here, we characterised these CPSF6 puncta further in HeLa cells, T-cells and macrophages and confirmed that integration and reverse transcription are not required for puncta formation. Indeed, we found that puncta formed very rapidly after infection, correlating with the time that CA entered the nucleus. In aphidicolin-treated HeLa cells and macrophages, puncta were detected for the length of the experiment, suggesting that puncta are only lost upon cell division. CA still co-localised with CPSF6 puncta at the latest time points, considerably after the peak of reverse transcription and integration. Intriguingly, the number of puncta induced in macrophages did not correlate with the MOI or the total number of nuclear speckles present in each cell, suggesting that CA/CPSF6 is only directed to a few nuclear speckles. Furthermore, we found that CPSF6 already co-localised with nuclear speckles in uninfected T-cells, suggesting that HIV-1 promotes a natural behaviour of CPSF6.


Subject(s)
HIV-1 , Macrophages , T-Lymphocytes , mRNA Cleavage and Polyadenylation Factors , HIV-1/physiology , Humans , mRNA Cleavage and Polyadenylation Factors/metabolism , mRNA Cleavage and Polyadenylation Factors/genetics , T-Lymphocytes/virology , T-Lymphocytes/metabolism , HeLa Cells , Macrophages/virology , Macrophages/metabolism , Virus Integration , Cell Nucleus/metabolism , Capsid Proteins/metabolism , Capsid Proteins/genetics , HIV Infections/virology , HIV Infections/metabolism , Capsid/metabolism
20.
Sci Rep ; 14(1): 10253, 2024 05 04.
Article in English | MEDLINE | ID: mdl-38704431

ABSTRACT

The tegument protein pp150 of Human Cytomegalovirus (HCMV) is known to be essential for the final stages of virus maturation and mediates its functions by interacting with capsid proteins. Our laboratory has previously identified the critical regions in pp150 important for pp150-capsid interactions and designed peptides similar in sequence to these regions, with a goal to competitively inhibit capsid maturation. Treatment with a specific peptide (PepCR2 or P10) targeted to pp150 conserved region 2 led to a significant reduction in murine CMV (MCMV) growth in cell culture, paving the way for in vivo testing in a mouse model of CMV infection. However, the general pharmacokinetic parameters of peptides, including rapid degradation and limited tissue and cell membrane permeability, pose a challenge to their successful use in vivo. Therefore, we designed a biopolymer-stabilized elastin-like polypeptide (ELP) fusion construct (ELP-P10) to enhance the bioavailability of P10. Antiviral efficacy and cytotoxic effects of ELP-P10 were studied in cell culture, and pharmacokinetics, biodistribution, and antiviral efficacy were studied in a mouse model of CMV infection. ELP-P10 maintained significant antiviral activity in cell culture, and this conjugation significantly enhanced P10 bioavailability in mouse tissues. The fluorescently labeled ELP-P10 accumulated to higher levels in mouse liver and kidneys as compared to the unconjugated P10. Moreover, viral titers from vital organs of MCMV-infected mice indicated a significant reduction of virus load upon ELP-P10 treatment. Therefore, ELP-P10 has the potential to be developed into an effective antiviral against CMV infection.


Subject(s)
Antiviral Agents , Cytomegalovirus Infections , Elastin , Muromegalovirus , Peptides , Phosphoproteins , Viral Matrix Proteins , Animals , Elastin/chemistry , Elastin/metabolism , Cytomegalovirus Infections/drug therapy , Cytomegalovirus Infections/virology , Mice , Antiviral Agents/pharmacology , Antiviral Agents/pharmacokinetics , Antiviral Agents/chemistry , Peptides/pharmacology , Peptides/chemistry , Muromegalovirus/drug effects , Humans , Capsid Proteins/metabolism , Capsid Proteins/chemistry , Cytomegalovirus/drug effects , Capsid/metabolism , Capsid/drug effects , Recombinant Fusion Proteins/pharmacology , Recombinant Fusion Proteins/pharmacokinetics , Disease Models, Animal , Elastin-Like Polypeptides
SELECTION OF CITATIONS
SEARCH DETAIL