Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 14.464
Filtrer
1.
Nat Commun ; 15(1): 6602, 2024 Aug 04.
Article de Anglais | MEDLINE | ID: mdl-39097583

RÉSUMÉ

Broadening gene therapy applications requires manufacturable vectors that efficiently transduce target cells in humans and preclinical models. Conventional selections of adeno-associated virus (AAV) capsid libraries are inefficient at searching the vast sequence space for the small fraction of vectors possessing multiple traits essential for clinical translation. Here, we present Fit4Function, a generalizable machine learning (ML) approach for systematically engineering multi-trait AAV capsids. By leveraging a capsid library that uniformly samples the manufacturable sequence space, reproducible screening data are generated to train accurate sequence-to-function models. Combining six models, we designed a multi-trait (liver-targeted, manufacturable) capsid library and validated 88% of library variants on all six predetermined criteria. Furthermore, the models, trained only on mouse in vivo and human in vitro Fit4Function data, accurately predicted AAV capsid variant biodistribution in macaque. Top candidates exhibited production yields comparable to AAV9, efficient murine liver transduction, up to 1000-fold greater human hepatocyte transduction, and increased enrichment relative to AAV9 in a screen for liver transduction in macaques. The Fit4Function strategy ultimately makes it possible to predict cross-species traits of peptide-modified AAV capsids and is a critical step toward assembling an ML atlas that predicts AAV capsid performance across dozens of traits.


Sujet(s)
Protéines de capside , Capside , Dependovirus , Vecteurs génétiques , Foie , Dependovirus/génétique , Animaux , Humains , Souris , Vecteurs génétiques/génétique , Capside/métabolisme , Protéines de capside/génétique , Protéines de capside/métabolisme , Foie/métabolisme , Transduction génétique , Techniques de transfert de gènes , Apprentissage machine , Thérapie génétique/méthodes , Macaca , Hépatocytes/métabolisme , Cellules HEK293 , Génie génétique/méthodes
2.
Nat Commun ; 15(1): 6551, 2024 Aug 02.
Article de Anglais | MEDLINE | ID: mdl-39095371

RÉSUMÉ

Jumbo phages are a group of tailed bacteriophages with large genomes and capsids. As a prototype of jumbo phage, ΦKZ infects Pseudomonas aeruginosa, a multi-drug-resistant (MDR) opportunistic pathogen leading to acute or chronic infection in immunocompromised individuals. It holds potential to be used as an antimicrobial agent and as a model for uncovering basic phage biology. Although previous low-resolution structural studies have indicated that jumbo phages may have more complicated capsid structures than smaller phages such as HK97, the detailed structures and the assembly mechanism of their capsids remain largely unknown. Here, we report a 3.5-Å-resolution cryo-EM structure of the ΦKZ capsid. The structure unveiled ten minor capsid proteins, with some decorating the outer surface of the capsid and the others forming a complex network attached to the capsid's inner surface. This network seems to play roles in driving capsid assembly and capsid stabilization. Similar mechanisms of capsid assembly and stabilization are probably employed by many other jumbo viruses.


Sujet(s)
Protéines de capside , Capside , Cryomicroscopie électronique , Pseudomonas aeruginosa , Capside/ultrastructure , Capside/composition chimique , Capside/métabolisme , Protéines de capside/composition chimique , Protéines de capside/métabolisme , Pseudomonas aeruginosa/virologie , Assemblage viral , Phages de Pseudomonas/ultrastructure , Phages de Pseudomonas/composition chimique , Bactériophages/physiologie , Bactériophages/composition chimique , Bactériophages/ultrastructure , Modèles moléculaires , Génome viral
3.
Viruses ; 16(7)2024 Jun 27.
Article de Anglais | MEDLINE | ID: mdl-39066201

RÉSUMÉ

In the fullerene cone HIV-1 capsid, the central channels of the hexameric and pentameric capsomers each contain a ring of arginine (Arg18) residues that perform essential roles in capsid assembly and function. In both the hexamer and pentamer, the Arg18 rings coordinate inositol hexakisphosphate, an assembly and stability factor for the capsid. Previously, it was shown that amino-acid substitutions of Arg18 can promote pentamer incorporation into capsid-like particles (CLPs) that spontaneously assemble in vitro under high-salt conditions. Here, we show that these Arg18 mutant CLPs contain a non-canonical pentamer conformation and distinct lattice characteristics that do not follow the fullerene geometry of retroviral capsids. The Arg18 mutant pentamers resemble the hexamer in intra-oligomeric contacts and form a unique tetramer-of-pentamers that allows for incorporation of an octahedral vertex with a cross-shaped opening in the hexagonal capsid lattice. Our findings highlight an unexpected degree of structural plasticity in HIV-1 capsid assembly.


Sujet(s)
Substitution d'acide aminé , Arginine , Protéines de capside , Capside , Fullerènes , VIH-1 (Virus de l'Immunodéficience Humaine de type 1) , Assemblage viral , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/génétique , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/physiologie , Protéines de capside/génétique , Protéines de capside/métabolisme , Protéines de capside/composition chimique , Fullerènes/composition chimique , Arginine/composition chimique , Capside/métabolisme , Capside/composition chimique , Humains , Multimérisation de protéines , Modèles moléculaires , Conformation des protéines
4.
Viruses ; 16(7)2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-39066243

RÉSUMÉ

Plus, minus, and double-strand RNA viruses are all found in nature. We use computational models to study the relative success of these strategies. We consider translation, replication, and virion assembly inside one cell, and transmission of virions between cells. For viruses which do not incorporate a polymerase in the capsid, transmission of only plus strands is the default strategy because virions containing minus strands are not infectious. Packaging only plus strands has a significant advantage if the number of RNA strands produced per cell is larger than the number of capsids. In this case, by not packaging minus strands, the virus produces more plus-strand virions. Therefore, plus-strand viruses are selected at low multiplicity of infection. However, at high multiplicity of infection, it is preferable to package both strands because the additional minus virions produced are helpful when there are multiple infections per cell. The fact that plus-strand viruses are widespread while viruses that package both strands are not seen in nature suggests that RNA strands are indeed produced in excess over capsids, and that the multiplicity of infection is not sufficiently high to favor the production of both kinds of virions. For double-strand viruses, we show that it is advantageous to produce only plus strands from the double strand within the cell, as is observed in real viruses. The reason for the success of minus-strand viruses is more puzzling initially. For viruses that incorporate a polymerase in the virion, minus virions are infectious. However, this is not sufficient to explain the success of minus-strand viruses, because in this case, viruses that package both strands outcompete those that package only minus or only plus. Real minus-strand viruses make use of replicable strands that are coated by a nucleoprotein, and separate translatable plus strands that are uncoated. Here we show that when there are distinct replicable and translatable strands, minus-strand viruses are selected.


Sujet(s)
Virus à ARN , ARN viral , Assemblage viral , Réplication virale , Virus à ARN/génétique , Virus à ARN/physiologie , ARN viral/génétique , ARN viral/métabolisme , Virion/génétique , Évolution moléculaire , Capside/métabolisme
5.
Viruses ; 16(7)2024 Jul 09.
Article de Anglais | MEDLINE | ID: mdl-39066266

RÉSUMÉ

Spiroplasma virus 4 (SpV4) is a bacteriophage of the Microviridae, which packages circular ssDNA within non-enveloped T = 1 icosahedral capsids. It infects spiroplasmas, which are known pathogens of honeybees. Here, the structure of the SpV4 virion is determined using cryo-electron microscopy to a resolution of 2.5 Å. A striking feature of the SpV4 capsid is the mushroom-like protrusions at the 3-fold axes, which is common among all members of the subfamily Gokushovirinae. While the function of the protrusion is currently unknown, this feature varies widely in this subfamily and is therefore possibly an adaptation for host recognition. Furthermore, on the interior of the SpV4 capsid, the location of DNA-binding protein VP8 was identified and shown to have low structural conservation to the capsids of other viruses in the family. The structural characterization of SpV4 will aid future studies analyzing the virus-host interaction, to understand disease mechanisms at a molecular level. Furthermore, the structural comparisons in this study, including a low-resolution structure of the chlamydia phage 2, provide an overview of the structural repertoire of the viruses in this family that infect various bacterial hosts, which in turn infect a wide range of animals and plants.


Sujet(s)
Protéines de capside , Capside , Cryomicroscopie électronique , Microviridae , Spiroplasma , Virion , Capside/ultrastructure , Capside/métabolisme , Capside/composition chimique , Protéines de capside/composition chimique , Protéines de capside/métabolisme , Protéines de capside/génétique , Spiroplasma/ultrastructure , Microviridae/génétique , Microviridae/ultrastructure , Microviridae/composition chimique , Virion/ultrastructure , Bactériophages/ultrastructure , Bactériophages/génétique , Bactériophages/classification , Bactériophages/composition chimique , Bactériophages/physiologie , Modèles moléculaires
6.
J Virol ; 98(7): e0070724, 2024 Jul 23.
Article de Anglais | MEDLINE | ID: mdl-38953655

RÉSUMÉ

Human norovirus was discovered more than five decades ago and is a widespread cause of outbreaks of acute gastroenteritis. There are no approved vaccines or antivirals currently available. However, norovirus inhibitors, including capsid-specific monoclonal antibodies (Mabs) and nanobodies, have recently shown promising results. Several Mabs and nanobodies were found to inhibit norovirus replication using a human intestinal enteroid (HIE) culture system and/or could block norovirus attachment to histo-blood group antigen (HBGA) co-factors. In our pursuit to develop a single broad-spectrum norovirus therapeutic, we continued our analysis and development of a cross-reactive and HBGA interfering nanobody (NB26). To improve NB26 binding capacity and therapeutic potential, we conjugated NB26 onto a human IgG Fc domain (Fc-NB26). We confirmed that Fc-NB26 cross-reacts with genetically diverse GII genotype capsid protruding (P) domains (GII.8, GII.14, GII.17, GII.24, GII.26, and GII.NA1) using a direct enzyme-linked immunosorbent assay. Furthermore, X-ray crystallography structures of these P domains and structures of other GII genotypes reveal that the NB26 binding site is largely conserved, validating its broad reactivity. We showed that Fc-NB26 has ~100-fold higher affinity toward the norovirus P domain compared to native NB26. We also found that both NB26 and Fc-NB26 neutralize human norovirus replication in the HIE culture system. Furthermore, the mode of inhibition confirmed that like NB26, Fc-NB26 caused norovirus particle disassembly and aggregation. Overall, these new findings demonstrate that structural modifications to nanobodies can improve their therapeutic potential.IMPORTANCEDeveloping vaccines and antivirals against norovirus remains a challenge, mainly due to the constant genetic and antigenic evolution. Moreover, re-infection with genetically related and/or antigenic variants is not uncommon. We further developed our leading norovirus nanobody (NB26) that indirectly interfered with norovirus binding to HBGAs, by converting NB26 into a dimeric Fc-linked Nanobody (Fc-NB26). We found that Fc-NB26 had improved binding affinity and neutralization capacity compared with native NB26. Using X-ray crystallography, we showed this nanobody engaged highly conserved capsid residues among genetically diverse noroviruses. Development of such broadly reactive potent therapeutic nanobodies delivered as a slow-releasing prophylactic could be of exceptional value for norovirus outbreaks, especially for the prevention or treatment of severe acute gastroenteritis in high-risk groups such as the young, elderly, and immunocompromised.


Sujet(s)
Infections à Caliciviridae , Protéines de capside , Norovirus , Anticorps à domaine unique , Norovirus/génétique , Norovirus/effets des médicaments et des substances chimiques , Norovirus/immunologie , Humains , Anticorps à domaine unique/immunologie , Anticorps à domaine unique/pharmacologie , Anticorps à domaine unique/composition chimique , Protéines de capside/immunologie , Protéines de capside/métabolisme , Protéines de capside/composition chimique , Protéines de capside/génétique , Infections à Caliciviridae/immunologie , Infections à Caliciviridae/virologie , Infections à Caliciviridae/thérapie , Antiviraux/pharmacologie , Fragments Fc des immunoglobulines/immunologie , Fragments Fc des immunoglobulines/composition chimique , Anticorps antiviraux/immunologie , Réactions croisées , Capside/métabolisme , Capside/immunologie , Antigènes de groupe sanguin/métabolisme , Réplication virale/effets des médicaments et des substances chimiques , Gastroentérite/virologie , Immunoglobuline G/immunologie , Anticorps monoclonaux/immunologie , Anticorps neutralisants/immunologie
7.
Int J Mol Sci ; 25(13)2024 Jun 28.
Article de Anglais | MEDLINE | ID: mdl-39000271

RÉSUMÉ

The human immunodeficiency virus type 1 (HIV-1) capsid is a protein core formed by multiple copies of the viral capsid (CA) protein. Inside the capsid, HIV-1 harbours all the viral components required for replication, including the genomic RNA and viral enzymes reverse transcriptase (RT) and integrase (IN). Upon infection, the RT transforms the genomic RNA into a double-stranded DNA molecule that is subsequently integrated into the host chromosome by IN. For this to happen, the viral capsid must open and release the viral DNA, in a process known as uncoating. Capsid plays a key role during the initial stages of HIV-1 replication; therefore, its stability is intimately related to infection efficiency, and untimely uncoating results in reverse transcription defects. How and where uncoating takes place and its relationship with reverse transcription is not fully understood, but the recent development of novel biochemical and cellular approaches has provided unprecedented detail on these processes. In this review, we present the latest findings on the intricate link between capsid stability, reverse transcription and uncoating, the different models proposed over the years for capsid uncoating, and the role played by other cellular factors on these processes.


Sujet(s)
Protéines de capside , Capside , VIH-1 (Virus de l'Immunodéficience Humaine de type 1) , Transcription inverse , Décapsidation virale , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/génétique , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/physiologie , Humains , Capside/métabolisme , Protéines de capside/métabolisme , Protéines de capside/génétique , Réplication virale , Infections à VIH/virologie , Infections à VIH/métabolisme , ARN viral/métabolisme , ARN viral/génétique , Transcriptase inverse du VIH/métabolisme , Transcriptase inverse du VIH/génétique
8.
Sci Rep ; 14(1): 16225, 2024 07 13.
Article de Anglais | MEDLINE | ID: mdl-39003336

RÉSUMÉ

In response to the escalating global threat of antimicrobial resistance, our laboratory has established a phagemid packaging system for the generation of CRISPR-Cas13a-antimicrobial capsids targeting methicillin-resistant Staphylococcus aureus (MRSA). However, a significant challenge arose during the packaging process: the unintentional production of wild-type phages alongside the antimicrobial capsids. To address this issue, the phagemid packaging system was optimized by strategically incorporated silent mutations. This approach effectively minimized contamination risks without compromising packaging efficiency. The study identified the indispensable role of phage packaging genes, particularly terL-terS, in efficient phagemid packaging. Additionally, the elimination of homologous sequences between the phagemid and wild-type phage genome was crucial in preventing wild-type phage contamination. The optimized phagemid-LSAB(mosaic) demonstrated sequence-specific killing, efficiently eliminating MRSA strains carrying target antibiotic-resistant genes. While acknowledging the need for further exploration across bacterial species and in vivo validation, this refined phagemid packaging system offers a valuable advancement in the development of CRISPR-Cas13a-based antimicrobials, shedding light on potential solutions in the ongoing battle against bacterial infections.


Sujet(s)
Systèmes CRISPR-Cas , Capside , Staphylococcus aureus résistant à la méticilline , Mutation , Staphylococcus aureus résistant à la méticilline/génétique , Staphylococcus aureus résistant à la méticilline/effets des médicaments et des substances chimiques , Capside/métabolisme , Antibactériens/pharmacologie , Bactériophages/génétique
9.
Nanoscale ; 16(29): 13820-13833, 2024 Jul 25.
Article de Anglais | MEDLINE | ID: mdl-38978480

RÉSUMÉ

Adeno-associated virus (AAV) has emerged as the most promising vector for in vivo human gene therapy, with several therapeutic approvals in the last few years and countless more under development. Underlying this remarkable success are several attractive features that AAV offers, including lack of pathogenicity, low immunogenicity, long-term gene expression without genomic integration, the ability to infect both dividing and non-dividing cells, etc. However, the commonly used wild-type AAV capsids in therapeutic development present significant challenges, including inadequate tissue specificity and the need for large doses to attain therapeutic effectiveness, raising safety concerns. Additionally, significant preexisting adaptive immunity against most natural capsids, and the development of such anti-capsid immunity after the first treatment, represent major challenges. Strategies to engineer the AAV capsid are critically needed to address these challenges and unlock the full promise of AAV gene therapy. Chemical modification of the AAV capsid has recently emerged as a powerful new approach to engineer its properties. Unlike genetic strategies, which can be more disruptive to the delicate capsid assembly and packaging processes, "late-stage" chemical modification of the assembled capsid-whether at natural amino acid residues or site-specifically installed noncanonical amino acid residues-often enables a versatile approach to introducing new properties to the capsid. This review summarizes the significant recent progress in AAV capsid engineering strategies, with a particular focus on chemical modifications in advancing the next generation of AAV-based gene therapies.


Sujet(s)
Capside , Dependovirus , Thérapie génétique , Vecteurs génétiques , Dependovirus/génétique , Humains , Vecteurs génétiques/composition chimique , Vecteurs génétiques/métabolisme , Capside/composition chimique , Capside/métabolisme , Protéines de capside/composition chimique , Protéines de capside/métabolisme , Protéines de capside/génétique , Animaux
10.
Biochemistry ; 63(15): 1913-1924, 2024 Aug 06.
Article de Anglais | MEDLINE | ID: mdl-39037053

RÉSUMÉ

Virus-like particles (VLPs) from bacteriophage MS2 provide a platform to study protein self-assembly and create engineered systems for drug delivery. Here, we aim to understand the impact of intersubunit interface mutations on the local and global structure and function of MS2-based VLPs. In previous work, our lab identified locally supercharged double mutants [T71K/G73R] that concentrate positive charge at capsid pores, enhancing uptake into mammalian cells. To study the effects of particle size on cellular internalization, we combined these double mutants with a single point mutation [S37P] that was previously reported to switch particle geometry from T = 3 to T = 1 icosahedral symmetry. These new variants retained their enhanced cellular uptake activity and could deliver small-molecule drugs with efficacy levels similar to our first-generation capsids. Surprisingly, these engineered triple mutants exhibit increased thermostability and unexpected geometry, producing T = 3 particles instead of the anticipated T = 1 assemblies. Transmission electron microscopy revealed various capsid assembly states, including wild-type (T = 3), T = 1, and rod-like particles, that could be accessed using different combinations of these point mutations. Molecular dynamics experiments recapitulated the structural rationale in silico for the single point mutation [S37P] forming a T = 1 virus-like particle and showed that this assembly state was not favored when combined with mutations that favor rod-like architectures. Through this work, we investigated how interdimer interface dynamics influence VLP size and morphology and how these properties affect particle function in applications such as drug delivery.


Sujet(s)
Capside , Levivirus , Levivirus/génétique , Levivirus/composition chimique , Levivirus/métabolisme , Capside/métabolisme , Capside/composition chimique , Capside/ultrastructure , Mutation , Protéines de capside/composition chimique , Protéines de capside/génétique , Protéines de capside/métabolisme , Virion/métabolisme , Virion/génétique , Virion/composition chimique , Mutation ponctuelle , Stabilité protéique , Humains , Modèles moléculaires
11.
Structure ; 32(6): 652-653, 2024 Jun 06.
Article de Anglais | MEDLINE | ID: mdl-38848682

RÉSUMÉ

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.


Sujet(s)
Capside , Génome viral , Capside/métabolisme , Capside/composition chimique , Protéines de capside/composition chimique , Protéines de capside/métabolisme , Protéines de capside/génétique , Assemblage viral , Simulation de dynamique moléculaire , Modèles moléculaires
12.
Protein Sci ; 33(7): e5074, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38888268

RÉSUMÉ

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.


Sujet(s)
Protéines de capside , Capside , Dependovirus , Thérapie génétique , Vecteurs génétiques , Dependovirus/génétique , Dependovirus/composition chimique , Protéines de capside/composition chimique , Protéines de capside/génétique , Protéines de capside/métabolisme , Vecteurs génétiques/composition chimique , Vecteurs génétiques/métabolisme , Vecteurs génétiques/génétique , Thérapie génétique/méthodes , Capside/composition chimique , Capside/métabolisme , Échange hydrogène/deutérium couplé à la spectrométrie de masse , Stabilité protéique , Humains , Conformation des protéines , Modèles moléculaires
13.
Article de Anglais | MEDLINE | ID: mdl-38908134

RÉSUMÉ

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.


Sujet(s)
Capside , Dependovirus , Dependovirus/génétique , Dependovirus/isolement et purification , Chromatographie d'échange d'ions/méthodes , Capside/composition chimique , Chromatographie en phase liquide à haute performance/méthodes , Reproductibilité des résultats
14.
Viruses ; 16(6)2024 May 21.
Article de Anglais | MEDLINE | ID: mdl-38932109

RÉSUMÉ

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.


Sujet(s)
Eaux d'égout , Eaux usées , Eaux usées/virologie , Eaux d'égout/virologie , Humains , Capside , Coliphages/isolement et purification , Coliphages/génétique , Coliphages/classification , Rotavirus/génétique , Rotavirus/isolement et purification , Norovirus/isolement et purification , Norovirus/génétique , Microbiologie de l'eau , Réaction de polymérisation en chaine en temps réel , Fèces/virologie , Enterovirus/isolement et purification , Enterovirus/génétique , Enterovirus/classification , Protéines de capside/génétique
15.
Viruses ; 16(6)2024 May 21.
Article de Anglais | MEDLINE | ID: mdl-38932107

RÉSUMÉ

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.


Sujet(s)
Génome viral , Rotavirus , Protéines virales non structurales , Assemblage viral , Réplication virale , Rotavirus/génétique , Rotavirus/physiologie , Protéines virales non structurales/métabolisme , Protéines virales non structurales/génétique , Humains , Animaux , ARN viral/génétique , ARN viral/métabolisme , Capside/métabolisme , Protéines de liaison à l'ARN
16.
Nat Microbiol ; 9(7): 1842-1855, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38918469

RÉSUMÉ

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.


Sujet(s)
Capside , Noyau de la cellule , Cryomicroscopie électronique , Enveloppe nucléaire , Libération de particules virales , Noyau de la cellule/métabolisme , Noyau de la cellule/virologie , Humains , Enveloppe nucléaire/métabolisme , Capside/métabolisme , Protéines de capside/métabolisme , Protéines de capside/génétique , Nucléocapside/métabolisme , Tomographie en microscopie électronique , Protéines virales/métabolisme , Protéines virales/génétique , Herpesviridae/physiologie , Herpesviridae/génétique
17.
J Am Soc Mass Spectrom ; 35(7): 1567-1575, 2024 Jul 03.
Article de Anglais | MEDLINE | ID: mdl-38888112

RÉSUMÉ

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.


Sujet(s)
Protéines de capside , Capside , Dependovirus , Dependovirus/composition chimique , Protéines de capside/analyse , Protéines de capside/composition chimique , Capside/composition chimique , Humains , Spectrométrie de masse/méthodes
18.
mBio ; 15(7): e0115824, 2024 Jul 17.
Article de Anglais | MEDLINE | ID: mdl-38912776

RÉSUMÉ

We have investigated the function of inositol hexakisphosphate (IP6) and inositol pentakisphosphate (IP5) in the replication of murine leukemia virus (MLV). While IP6 is known to be critical for the life cycle of HIV-1, its significance in MLV remains unexplored. We find that IP6 is indeed important for MLV replication. It significantly enhances endogenous reverse transcription (ERT) in MLV. Additionally, a pelleting-based assay reveals that IP6 can stabilize MLV cores, thereby facilitating ERT. We find that IP5 and IP6 are packaged in MLV particles. However, unlike HIV-1, MLV depends upon the presence of IP6 and IP5 in target cells for successful infection. This IP6/5 requirement for infection is reflected in impaired reverse transcription observed in IP6/5-deficient cell lines. In summary, our findings demonstrate the importance of capsid stabilization by IP6/5 in the replication of diverse retroviruses; we suggest possible reasons for the differences from HIV-1 that we observed in MLV.IMPORTANCEInositol hexakisphosphate (IP6) is crucial for the assembly and replication of HIV-1. IP6 is packaged in HIV-1 particles and stabilizes the viral core enabling it to synthesize viral DNA early in viral infection. While its importance for HIV-1 is well established, its significance for other retroviruses is unknown. Here we report the role of IP6 in the gammaretrovirus, murine leukemia virus (MLV). We found that like HIV-1, MLV packages IP6, and as in HIV-1, IP6 stabilizes the MLV core thus promoting reverse transcription. Interestingly, we discovered a key difference in the role of IP6 in MLV versus HIV-1: while HIV-1 is not dependent upon IP6 levels in target cells, MLV replication is significantly reduced in IP6-deficient cell lines. We suggest that this difference in IP6 requirements reflects key differences between HIV-1 and MLV replication.


Sujet(s)
Virus de la leucémie murine , Acide phytique , Réplication virale , Acide phytique/métabolisme , Virus de la leucémie murine/physiologie , Virus de la leucémie murine/génétique , Humains , Animaux , Transcription inverse , Souris , Inositol phosphates/métabolisme , Lignée cellulaire , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/physiologie , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/génétique , Cellules HEK293 , Capside/métabolisme , Assemblage viral
19.
Biochemistry ; 63(12): 1543-1552, 2024 Jun 18.
Article de Anglais | MEDLINE | ID: mdl-38787909

RÉSUMÉ

Hepatitis B virus (HBV) displays remarkable self-assembly capabilities that interest the scientific community and biotechnological industries as HBV is leading to an annual mortality of up to 1 million people worldwide (especially in Africa and Southeast Asia). When the ionic strength is increased, hepatitis B virus-like particles (VLPs) can assemble from dimers of the first 149 residues of the HBV capsid protein core assembly domain (Cp149). Using solution small-angle X-ray scattering, we investigated the disassembly of the VLPs by titrating guanidine hydrochloride (GuHCl). Measurements were performed with and without 1 M NaCl, added either before or after titrating GuHCl. Fitting the scattering curves to a linear combination of atomic models of Cp149 dimer (the subunit) and T = 3 and T = 4 icosahedral capsids revealed the mass fraction of the dimer in each structure in all the titration points. Based on the mass fractions, the variation in the dimer-dimer association standard free energy was calculated as a function of added GuHCl, showing a linear relation between the interaction strength and GuHCl concentration. Using the data, we estimated the energy barriers for assembly and disassembly and the critical nucleus size for all of the assembly reactions. Extrapolating the standard free energy to [GuHCl] = 0 showed an evident hysteresis in the assembly process, manifested by differences in the dimer-dimer association standard free energy obtained for the disassembly reactions compared with the equivalent assembly reactions. Similar hysteresis was observed in the energy barriers for assembly and disassembly and the critical nucleus size. The results suggest that above 1.5 M, GuHCl disassembled the capsids by attaching to the protein and adding steric repulsion, thereby weakening the hydrophobic attraction.


Sujet(s)
Capside , Guanidine , Virus de l'hépatite B , Guanidine/composition chimique , Guanidine/pharmacologie , Virus de l'hépatite B/composition chimique , Virus de l'hépatite B/physiologie , Virus de l'hépatite B/effets des médicaments et des substances chimiques , Capside/composition chimique , Capside/métabolisme , Protéines de capside/composition chimique , Protéines de capside/métabolisme , Diffusion aux petits angles , Multimérisation de protéines , Modèles moléculaires , Assemblage viral/effets des médicaments et des substances chimiques , Diffraction des rayons X
20.
ACS Synth Biol ; 13(6): 1842-1850, 2024 Jun 21.
Article de Anglais | MEDLINE | ID: mdl-38729919

RÉSUMÉ

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.


Sujet(s)
Protéines de capside , Capside , Transfert d'énergie par résonance de fluorescence , Peptides , Peptides/composition chimique , Capside/composition chimique , Capside/métabolisme , Protéines de capside/composition chimique , Protéines de capside/métabolisme , Protéines de capside/génétique , Protéines luminescentes/composition chimique , Protéines luminescentes/métabolisme , Protéines luminescentes/génétique , Microscopie électronique à transmission , Polarisation de fluorescence , Assemblage viral
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE