Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
PLoS Pathog ; 19(1): e1011128, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36689483

RESUMO

Coronavirus disease 2019 is a respiratory infectious disease caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Evidence on the pathogenesis of SARS-CoV-2 is accumulating rapidly. In addition to structural proteins such as Spike and Envelope, the functional roles of non-structural and accessory proteins in regulating viral life cycle and host immune responses remain to be understood. Here, we show that open reading frame 8 (ORF8) acts as messenger for inter-cellular communication between alveolar epithelial cells and macrophages during SARS-CoV-2 infection. Mechanistically, ORF8 is a secretory protein that can be secreted by infected epithelial cells via both conventional and unconventional secretory pathways. Conventionally secreted ORF8 is glycosylated and loses the ability to recognize interleukin 17 receptor A of macrophages, possibly due to the steric hindrance imposed by N-glycosylation at Asn78. However, unconventionally secreted ORF8 does not undergo glycosylation without experiencing the ER-Golgi trafficking, thereby activating the downstream NF-κB signaling pathway and facilitating a burst of cytokine release. Furthermore, we show that ORF8 deletion in SARS-CoV-2 attenuates inflammation and yields less lung lesions in hamsters. Our data collectively highlights a role of ORF8 protein in the development of cytokine storms during SARS-CoV-2 infection.


Assuntos
COVID-19 , Síndrome da Liberação de Citocina , SARS-CoV-2 , Proteínas Virais , Humanos , COVID-19/patologia , Síndrome da Liberação de Citocina/patologia , Inflamação , Fases de Leitura Aberta , SARS-CoV-2/fisiologia , Proteínas Virais/metabolismo
2.
Mol Ther ; 31(8): 2391-2407, 2023 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-37263272

RESUMO

Live attenuated vaccines (LAVs) administered via the mucosal route may offer better control of the COVID-19 pandemic than non-replicating vaccines injected intramuscularly. Conceptionally, LAVs have several advantages, including presentation of the entire antigenic repertoire of the virus, and the induction of strong mucosal immunity. Thus, immunity induced by LAV could offer superior protection against future surges of COVID-19 cases caused by emerging SARS-CoV-2 variants. However, LAVs carry the risk of unintentional transmission. To address this issue, we investigated whether transmission of a SARS-CoV-2 LAV candidate can be blocked by removing the furin cleavage site (FCS) from the spike protein. The level of protection and immunity induced by the attenuated virus with the intact FCS was virtually identical to the one induced by the attenuated virus lacking the FCS. Most importantly, removal of the FCS completely abolished horizontal transmission of vaccine virus between cohoused hamsters. Furthermore, the vaccine was safe in immunosuppressed animals and showed no tendency to recombine in vitro or in vivo with a SARS-CoV-2 field strain. These results indicate that removal of the FCS from SARS-CoV-2 LAV is a promising strategy to increase vaccine safety and prevent vaccine transmission without compromising vaccine efficacy.


Assuntos
Vacinas contra COVID-19 , COVID-19 , Animais , Cricetinae , Humanos , COVID-19/prevenção & controle , Pandemias , SARS-CoV-2 , Vacinas Atenuadas , Anticorpos Antivirais , Anticorpos Neutralizantes
3.
J Virol ; 96(14): e0065322, 2022 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-35862676

RESUMO

Infectious bronchitis virus (IBV) is an avian coronavirus that causes infectious bronchitis, an acute and highly contagious respiratory disease of chickens. IBV evolution under the pressure of comprehensive and widespread vaccination requires surveillance for vaccine resistance, as well as periodic vaccine updates. Reverse genetics systems are very valuable tools in virology, as they facilitate rapid genetic manipulation of viral genomes, thereby advancing basic and applied research. We report here the construction of an infectious clone of IBV strain Beaudette as a bacterial artificial chromosome (BAC). The engineered full-length IBV clone allowed the rescue of an infectious virus that was phenotypically indistinguishable from the parental virus. We used the infectious IBV clone and examined whether an enhanced green fluorescent protein (EGFP) can be produced by the replicase gene ORF1 and autocatalytically released from the replicase polyprotein through cleavage by the main coronavirus protease. We show that IBV tolerates insertion of the EGFP ORF at the 3' end of the replicase gene, between the sequences encoding nsp13 and nsp16 (helicase, RNA exonuclease, RNA endonuclease, and RNA methyltransferase). We further show that EGFP is efficiently cleaved from the replicase polyprotein and can be localized in double-membrane vesicles along with viral RNA polymerase and double-stranded RNA, an intermediate of IBV genome replication. One of the engineered reporter EGFP viruses were genetically stable during passage in cultured cells. We demonstrate that the reporter EGFP viruses can be used to study virus replication in host cells and for antiviral drug discovery and development of diagnostic assays. IMPORTANCE Reverse genetics systems based on bacterial artificial chromosomes (BACs) are the most valuable systems in coronavirus research. Here, we describe the establishment of a reverse genetics system for the avian coronavirus strain Beaudette, the most intensively studied strain. We cloned a copy of the avian coronavirus genome into a BAC vector and recovered infectious virus in permissive cells. We used the new system to construct reporter viruses that produce enhanced green fluorescent protein (EGFP). The EGFP coding sequence was inserted into 11 known cleavage sites of the major coronavirus protease in the replicase gene ORF1. Avian coronavirus tolerated the insertion of the EGFP coding sequence at three sites. The engineered reporter viruses replicated with parental efficiency in cultured cells and were sufficiently genetically stable. The new system facilitates functional genomics of the avian coronavirus genome but can also be used for the development of novel vaccines and anticoronaviral drugs.


Assuntos
Infecções por Coronavirus , Vírus da Bronquite Infecciosa , Genética Reversa , Animais , Galinhas , Infecções por Coronavirus/veterinária , Genes Reporter , Proteínas de Fluorescência Verde , Vírus da Bronquite Infecciosa/genética , Peptídeo Hidrolases , Poliproteínas , RNA Viral/genética
4.
PLoS Pathog ; 14(1): e1006857, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29377958

RESUMO

Codon pair bias deoptimization (CPBD) has enabled highly efficient and rapid attenuation of RNA viruses. The technique relies on recoding of viral genes by increasing the number of codon pairs that are statistically underrepresented in protein coding genes of the viral host without changing the amino acid sequence of the encoded proteins. Utilization of naturally underrepresented codon pairs reduces protein production of recoded genes and directly causes virus attenuation. As a result, the mutant virus is antigenically identical with the parental virus, but virulence is reduced or absent. Our goal was to determine if a virus with a large double-stranded DNA genome, highly oncogenic Marek's disease virus (MDV), can be attenuated by CPBD. We recoded UL30 that encodes the catalytic subunit of the viral DNA polymerase to minimize (deoptimization), maximize (optimization), or preserve (randomization) the level of overrepresented codon pairs of the MDV host, the chicken. A fully codon pair-deoptimized UL30 mutant could not be recovered in cell culture. The sequence of UL30 was divided into three segments of equal length and we generated a series of mutants with different segments of the UL30 recoded. The codon pair-deoptimized genes, in which two segments of UL30 had been recoded, showed reduced rates of protein production. In cultured cells, the corresponding viruses formed smaller plaques and grew to lower titers compared with parental virus. In contrast, codon pair-optimized and -randomized viruses replicated in vitro with kinetics that were similar to those of the parental virus. Animals that were infected with the partially codon pair-deoptimized virus showed delayed progression of disease and lower mortality rates than codon pair-optimized and parental viruses. These results demonstrate that CPBD of a herpesvirus gene causes attenuation of the recoded virus and that CPBD may be an applicable strategy for attenuation of other large DNA viruses.


Assuntos
Pareamento Incorreto de Bases , Códon/genética , Herpesvirus Galináceo 2/genética , Doença de Marek/virologia , Vacinas Atenuadas/genética , Virulência , Algoritmos , Animais , Pareamento Incorreto de Bases/fisiologia , Células Cultivadas , Embrião de Galinha , Galinhas , Chlorocebus aethiops , Biologia Computacional/métodos , Genes Virais , Células HEK293 , Células HeLa , Herpesvirus Galináceo 2/imunologia , Humanos , Doença de Marek/imunologia , Vacinas Atenuadas/metabolismo , Células Vero , Proteínas Virais/genética , Virulência/genética
5.
Virus Genes ; 56(4): 448-460, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32328924

RESUMO

Orthohantaviruses are re-emerging rodent-borne pathogens distributed all over the world. Here, we report the isolation of a Puumala orthohantavirus (PUUV) strain from bank voles caught in a highly endemic region around the city Osnabrück, north-west Germany. Coding and non-coding sequences of all three segments (S, M, and L) were determined from original lung tissue, after isolation and after additional passaging in VeroE6 cells and a bank vole-derived kidney cell line. Different single amino acid substitutions were observed in the RNA-dependent RNA polymerase (RdRP) of the two stable PUUV isolates. The PUUV strain from VeroE6 cells showed a lower titer when propagated on bank vole cells compared to VeroE6 cells. Additionally, glycoprotein precursor (GPC)-derived virus-like particles of a German PUUV sequence allowed the generation of monoclonal antibodies that allowed the reliable detection of the isolated PUUV strain in the immunofluorescence assay. In conclusion, this is the first isolation of a PUUV strain from Central Europe and the generation of glycoprotein-specific monoclonal antibodies for this PUUV isolate. The obtained virus isolate and GPC-specific antibodies are instrumental tools for future reservoir host studies.


Assuntos
Anticorpos Monoclonais/imunologia , Anticorpos Antivirais/imunologia , Orthohepadnavirus/genética , Virus Puumala/genética , Animais , Anticorpos Antivirais/genética , Alemanha , Humanos , Orthohepadnavirus/imunologia , Orthohepadnavirus/isolamento & purificação , Virus Puumala/imunologia , Virus Puumala/isolamento & purificação
6.
J Gen Virol ; 99(12): 1705-1716, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30113295

RESUMO

Codon pair bias deoptimization (CPBD) has been successfully used to attenuate several RNA viruses. CPBD involves recoding a viral protein-coding sequence to maximize the number of codon pairs that are statistically underrepresented in the host, which presumably slows protein translation and, hence, causes virus attenuation. However, since recoding preserves the amino acid composition and codon bias, attenuated and parental viruses are antigenically identical. To determine if Marek's disease virus (MDV), a highly oncogenic herpesvirus of the chicken with a large double-stranded DNA genome, can be attenuated by CPBD of its major oncogene meq, we recoded the gene to minimize (meq-D), maximize (meq-O), or preserve (meq-R) the level of codon pairs that are overrepresented in the chicken protein-coding sequences. Unexpectedly, mutants carrying recoded genes produced comparable or increased levels of Meq in the context of viral infection in cultured cells. In addition, parental virus and mutant viruses carrying recoded meq genes replicated with comparable kinetics in vitro and in vivo, and were equally virulent in susceptible chickens. In summary, CPBD of meq failed to produce any quantifiable attenuation of MDV and confirms differences in the complexity of applying CPBD to large DNA viruses versus RNA viruses.


Assuntos
Códon , Herpesvirus Meleagrídeo 1/crescimento & desenvolvimento , Herpesvirus Meleagrídeo 1/genética , Proteínas Oncogênicas Virais/genética , Proteínas Recombinantes/genética , Replicação Viral , Animais , Linhagem Celular , Galinhas , Células Epiteliais/virologia , Proteínas Oncogênicas Virais/metabolismo , Proteínas Recombinantes/metabolismo , Virulência , Cultura de Vírus
7.
J Biol Chem ; 288(49): 35396-405, 2013 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-24142700

RESUMO

Signal peptide cleavage and N-glycosylation of proteins are co-translational processes, but little is known about their interplay if they compete for adjacent sites. Here we report two unique findings for processing of glycoprotein 3 of equine arteritis virus. Glycoprotein 3 (Gp3) contains an N-terminal signal peptide, which is not removed, although bioinformatics predicts cleavage with high probability. There is an overlapping sequon, NNTT, adjacent to the signal peptide that we show to be glycosylated at both asparagines. Exchanging the overlapping sequon and blocking glycosylation allows signal peptide cleavage, indicating that carbohydrate attachment inhibits processing of a potentially cleavable signal peptide. Bioinformatics analyses suggest that a similar processing scheme may exist for some cellular proteins. Membrane fractionation and secretion experiments revealed that the signal peptide of Gp3 does not act as a membrane anchor, indicating that it is completely translocated into the lumen of the endoplasmic reticulum. Membrane attachment is caused by the hydrophobic C terminus of Gp3, which, however, does not span the membrane but rather attaches the protein peripherally to endoplasmic reticulum membranes.


Assuntos
Equartevirus/genética , Equartevirus/metabolismo , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Asparagina/química , Sítios de Ligação/genética , Células CHO , Biologia Computacional , Cricetulus , Retículo Endoplasmático/metabolismo , Glicosilação , Cavalos , Humanos , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Modificação Traducional de Proteínas , Sinais Direcionadores de Proteínas/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas do Envelope Viral/química
8.
Nat Commun ; 15(1): 995, 2024 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-38307868

RESUMO

The development of effective SARS-CoV-2 vaccines has been essential to control COVID-19, but significant challenges remain. One problem is intramuscular administration, which does not induce robust mucosal immune responses in the upper airways-the primary site of infection and virus shedding. Here we compare the efficacy of a mucosal, replication-competent yet fully attenuated virus vaccine, sCPD9-ΔFCS, and the monovalent mRNA vaccine BNT162b2 in preventing transmission of SARS-CoV-2 variants B.1 and Omicron BA.5 in two scenarios. Firstly, we assessed the protective efficacy of the vaccines by exposing vaccinated male Syrian hamsters to infected counterparts. Secondly, we evaluated transmission of the challenge virus from vaccinated and subsequently challenged male hamsters to naïve contacts. Our findings demonstrate that the live-attenuated vaccine (LAV) sCPD9-ΔFCS significantly outperformed the mRNA vaccine in preventing virus transmission in both scenarios. Our results provide evidence for the advantages of locally administered LAVs over intramuscularly administered mRNA vaccines in preventing infection and reducing virus transmission.


Assuntos
Vacinas contra COVID-19 , COVID-19 , Animais , Cricetinae , Masculino , Humanos , Vacina BNT162 , COVID-19/prevenção & controle , Vacinas de mRNA , SARS-CoV-2 , Mesocricetus , Anticorpos Antivirais , Anticorpos Neutralizantes
9.
Avian Dis ; 57(2 Suppl): 351-9, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23901746

RESUMO

Proteomics is the application of rapidly evolving high-throughput technologies that enable analysis of proteins on a large scale. Recent advances in instrumentation have allowed detection, identification, and quantification of proteins with unparalleled precision and reproducibility, and this, in combination with novel bioinformatics tools, has helped to move proteomics from the simple cataloging of expressed proteins toward discovery of operating mechanisms in the biological systems. Proteomics holds great promise for advancing the understanding of viral pathogenesis, immunity, and the dynamics of virus-host protein interactions. Nevertheless, only a small number of proteomic studies have been done on animal viruses and avian herpesviruses in particular. This review summarizes the basic concepts and technologies used in proteomics and highlights the most successful applications of different proteomic approaches that resulted in identification of new virus-host protein interactions, mechanisms of genetic resistance and susceptibility to Marek's disease in chickens, and profiling and analysis of proteomes of Gallid herpesvirus 2 (GaHV-2), GaHV-3, and Meleagrid herpesvirus 1 infected or transformed cells. This review also discusses current limitations and potential future applications of proteomic methods in avian herpesvirus research.


Assuntos
Galinhas , Mardivirus/genética , Doença de Marek/genética , Doenças das Aves Domésticas/genética , Proteoma , Proteômica/métodos , Proteínas Virais/genética , Animais , Resistência à Doença , Herpesviridae/genética , Herpesviridae/metabolismo , Mardivirus/metabolismo , Proteínas Virais/metabolismo
10.
Cell Host Microbe ; 31(7): 1170-1184.e7, 2023 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-37402373

RESUMO

The historically dominant SARS-CoV-2 Delta variant and the currently dominant Omicron variants carry a T492I substitution within the non-structural protein 4 (NSP4). Based on in silico analyses, we hypothesized that the T492I mutation increases viral transmissibility and adaptability, which we confirmed with competition experiments in hamster and human airway tissue culture models. Furthermore, we showed that the T492I mutation increases the replication capacity and infectiveness of the virus and improves its ability to evade host immune responses. Mechanistically, the T492I mutation increases the cleavage efficiency of the viral main protease NSP5 by enhancing enzyme-substrate binding, which increases production of nearly all non-structural proteins processed by NSP5. Importantly, the T492I mutation suppresses viral-RNA-associated chemokine production in monocytic macrophages, which may contribute to the attenuated pathogenicity of Omicron variants. Our results highlight the importance of NSP4 adaptation in the evolutionary dynamics of SARS-CoV-2.


Assuntos
COVID-19 , Animais , Cricetinae , Humanos , SARS-CoV-2/genética , Evolução Biológica , Mutação , Glicoproteína da Espícula de Coronavírus
11.
Viruses ; 15(2)2023 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-36851681

RESUMO

Seoul orthohantavirus (SEOV) is a rat-associated zoonotic pathogen with an almost worldwide distribution. In 2019, the first autochthonous human case of SEOV-induced hemorrhagic fever with renal syndrome was reported in Germany, and a pet rat was identified as the source of the zoonotic infection. To further investigate the SEOV reservoir, additional rats from the patient and another owner, all of which were purchased from the same vendor, were tested. SEOV RNA and anti-SEOV antibodies were found in both of the patient's rats and in two of the three rats belonging to the other owner. The complete coding sequences of the small (S), medium (M), and large (L) segments obtained from one rat per owner exhibited a high sequence similarity to SEOV strains of breeder rat or human origin from the Netherlands, France, the USA, and Great Britain. Serological screening of 490 rats from breeding facilities and 563 wild rats from Germany (2007-2020) as well as 594 wild rats from the Netherlands (2013-2021) revealed 1 and 6 seropositive individuals, respectively. However, SEOV RNA was not detected in any of these animals. Increased surveillance of pet, breeder, and wild rats is needed to identify the origin of the SEOV strain in Europe and to develop measures to prevent transmission to the human population.


Assuntos
Vírus Seoul , Zoonoses , Humanos , Animais , Ratos , Europa (Continente) , Cruzamento , Éxons , França , RNA , Vírus Seoul/genética
12.
Nat Microbiol ; 8(5): 860-874, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-37012419

RESUMO

Vaccines play a critical role in combating the COVID-19 pandemic. Future control of the pandemic requires improved vaccines with high efficacy against newly emerging SARS-CoV-2 variants and the ability to reduce virus transmission. Here we compare immune responses and preclinical efficacy of the mRNA vaccine BNT162b2, the adenovirus-vectored spike vaccine Ad2-spike and the live-attenuated virus vaccine candidate sCPD9 in Syrian hamsters, using both homogeneous and heterologous vaccination regimens. Comparative vaccine efficacy was assessed by employing readouts from virus titrations to single-cell RNA sequencing. Our results show that sCPD9 vaccination elicited the most robust immunity, including rapid viral clearance, reduced tissue damage, fast differentiation of pre-plasmablasts, strong systemic and mucosal humoral responses, and rapid recall of memory T cells from lung tissue after challenge with heterologous SARS-CoV-2. Overall, our results demonstrate that live-attenuated vaccines offer advantages over currently available COVID-19 vaccines.


Assuntos
COVID-19 , SARS-CoV-2 , Animais , Cricetinae , Humanos , Vacinas Atenuadas , COVID-19/prevenção & controle , Vacinas contra COVID-19 , Vacina BNT162 , Pandemias , Mesocricetus
13.
Comput Struct Biotechnol J ; 20: 4376-4380, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35992535

RESUMO

Research with infectious SARS-CoV-2 is complicated because it must be conducted under biosafety level 3 (BSL-3) conditions. Recently, we constructed a live attenuated SARS-CoV-2 virus by rational design through partial recoding of the SARS-CoV-2 genome and showed that the attenuated virus, designated sCPD9, was highly attenuated in preclinical animal models. The recoded sequence was designed by codon pair deoptimization and is located at the distal end of gene ORF1ab. Codon pair deoptimization involves recoding of the viral sequence with underrepresented codon pairs but without altering the amino acid sequence of the encoded proteins. Thus, parental and attenuated viruses produce exactly the same proteins. In Germany, the live attenuated SARS-CoV-2 mutant sCPD9 was recently classified as a BSL-2 pathogen based on its genetic stability and strong attenuation in preclinical animal models. Despite its high attenuation in vivo, sCPD9 grows to high titers in common cell lines, making it suitable as substitute for virulent SARS-CoV-2 in many experimental setups. Consequently, sCPD9 can ease and accelerate SARS-CoV-2 research under BSL-2 conditions, particularly in experiments requiring replicating virus, such as diagnostics and development of antiviral drugs.

14.
Transbound Emerg Dis ; 69(2): 886-890, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33534959

RESUMO

The striped field mouse (Apodemus agrarius) is known to carry several zoonotic pathogens, including Leptospira spp. and Dobrava-Belgrade orthohantavirus (DOBV). Since its first detection in 1996 in south-east Austria, the striped field mouse has further expanded its range in Austria. Here, we screened 35 striped field mice collected in an Austrian region near the Hungarian border for DOBV, Leptospira spp. and seven vector-borne pathogens. Hantavirus RT-PCR screening and DOBV IgG ELISA analysis led to the detection of two DOBV-positive striped field mice. The complete coding sequences of all three genome segments of both strains were determined by a combination of target enrichment and next-generation sequencing. Both complete coding S segment sequences clustered within the DOBV genotype Kurkino clade with the highest similarity to a sequence from Hungary. In one of 35 striped field mice, Leptospira borgpetersenii sequence type (ST) 146 was detected. Bartonella spp., Borrelia miyamotoi and Neoehrlichia mikurensis DNA was detected in four, one and two of 32 mice, respectively. Babesia, Anaplasma, Ehrlichia and Rickettsia specific DNA was not detected. Future investigations will have to determine the prevalence and invasion of these pathogens with the ongoing range expansion of the striped field mouse in Austria.


Assuntos
Anaplasmataceae , Infecções por Hantavirus , Orthohantavírus , Doenças dos Roedores , Animais , Áustria/epidemiologia , Orthohantavírus/genética , Infecções por Hantavirus/epidemiologia , Infecções por Hantavirus/veterinária , Camundongos , Murinae/microbiologia , Doenças dos Roedores/diagnóstico , Doenças dos Roedores/epidemiologia , Doenças dos Roedores/microbiologia
15.
Virus Evol ; 8(2): veac099, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36405341

RESUMO

Evolution relies on the availability of genetic diversity for fitness-based selection. However, most deoxyribonucleic acid (DNA) viruses employ DNA polymerases (Pol) capable of exonucleolytic proofreading to limit mutation rates during DNA replication. The relative genetic stability produced by high-fidelity genome replication can make studying DNA virus adaptation and evolution an intensive endeavor, especially in slowly replicating viruses. Here, we present a proofreading-impaired Pol mutant (Y547S) of Marek's disease virus that exhibits a hypermutator phenotype while maintaining unimpaired growth in vitro and wild-type (WT)-like pathogenicity in vivo. At the same time, mutation frequencies observed in Y547S virus populations are 2-5-fold higher compared to the parental WT virus. We find that Y547S adapts faster to growth in originally non-permissive cells, evades pressure conferred by antiviral inhibitors more efficiently, and is more easily attenuated by serial passage in cultured cells compared to WT. Our results suggest that hypermutator viruses can serve as a tool to accelerate evolutionary processes and help identify key genetic changes required for adaptation to novel host cells and resistance to antiviral therapy. Similarly, the rapid attenuation achieved through adaptation of hypermutators to growth in cell culture enables identification of genetic changes underlying attenuation and virulence, knowledge that could practically exploited, e.g. in the rational design of vaccines.

16.
Mol Inform ; 40(9): e2100031, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34378348

RESUMO

Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) continues to be a global threat, causing millions of deaths worldwide. SARS-CoV-2 is an enveloped virus with spike (S) glycoproteins conferring binding to the host cell's angiotensin-converting enzyme 2 (ACE2), which is critical for cellular entry. The host range of the virus extends well beyond humans and non-human primates. Natural and experimental infections have confirmed the high susceptibility of cats, ferrets, and Syrian hamsters, whereas dogs, mice, rats, pigs, and chickens are refractory to SARS-CoV-2 infection. To investigate the underlying reason for the variable susceptibility observed in different species, we have developed molecular descriptors to efficiently analyse dynamic simulation models of complexes between SARS-CoV-2 S and ACE2. Our extensive analyses represent the first systematic structure-based approach that allows predictions of species susceptibility to SARS-CoV-2 infection.


Assuntos
SARS-CoV-2/química , Enzima de Conversão de Angiotensina 2/química , Enzima de Conversão de Angiotensina 2/metabolismo , Animais , COVID-19/metabolismo , Gatos , Cães , Furões , Haplorrinos , Humanos , Mesocricetus , Camundongos , Simulação de Dinâmica Molecular , Ratos , SARS-CoV-2/metabolismo , Especificidade da Espécie , Suínos
17.
Sci Adv ; 7(49): eabk0172, 2021 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-34851677

RESUMO

Vaccines are instrumental and indispensable in the fight against the COVID-19 pandemic. Several recent SARS-CoV-2 variants are more transmissible and evade infection- or vaccine-induced protection. We constructed live attenuated vaccine candidates by large-scale recoding of the SARS-CoV-2 genome and showed that the lead candidate, designated sCPD9, protects Syrian hamsters from a challenge with ancestral virus. Here, we assessed immunogenicity and protective efficacy of sCPD9 in the Roborovski dwarf hamster, a nontransgenic rodent species that is highly susceptible to SARS-CoV-2 and severe COVID-19­like disease. We show that a single intranasal vaccination with sCPD9 elicited strong cross-neutralizing antibody responses against four current SARS-CoV-2 variants of concern, B.1.1.7 (Alpha), B.1.351 (Beta), B.1.1.28.1 (Gamma), and B.1.617.2 (Delta). The sCPD9 vaccine offered complete protection from COVID-19­like disease caused by the ancestral SARS-CoV-2 variant B.1 and the two variants of concern B.1.1.7 and B.1.351.

18.
Cell Rep ; 36(5): 109493, 2021 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-34320400

RESUMO

Safe and effective vaccines are urgently needed to stop the pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). We construct a series of live attenuated vaccine candidates by large-scale recoding of the SARS-CoV-2 genome and assess their safety and efficacy in Syrian hamsters. Animals were vaccinated with a single dose of the respective recoded virus and challenged 21 days later. Two of the tested viruses do not cause clinical symptoms but are highly immunogenic and induce strong protective immunity. Attenuated viruses replicate efficiently in the upper but not in the lower airways, causing only mild pulmonary histopathology. After challenge, hamsters develop no signs of disease and rapidly clear challenge virus: at no time could infectious virus be recovered from the lungs of infected animals. The ease with which attenuated virus candidates can be produced and administered favors their further development as vaccines to combat the ongoing pandemic.


Assuntos
Vacinas contra COVID-19 , COVID-19/imunologia , COVID-19/prevenção & controle , Sistema Respiratório/patologia , Sistema Respiratório/virologia , SARS-CoV-2/genética , SARS-CoV-2/imunologia , Animais , Chlorocebus aethiops , Edição de Genes , Genoma Viral , Humanos , Imunidade , Mesocricetus , Mutação , Pandemias/prevenção & controle , Vacinas Atenuadas , Células Vero , Replicação Viral
19.
Viruses ; 12(7)2020 07 20.
Artigo em Inglês | MEDLINE | ID: mdl-32698441

RESUMO

In late 2019, an outbreak of a severe respiratory disease caused by an emerging coronavirus, SARS-CoV-2, resulted in high morbidity and mortality in infected humans. Complete understanding of COVID-19, the multi-faceted disease caused by SARS-CoV-2, requires suitable small animal models, as does the development and evaluation of vaccines and antivirals. Since age-dependent differences of COVID-19 were identified in humans, we compared the course of SARS-CoV-2 infection in young and aged Syrian hamsters. We show that virus replication in the upper and lower respiratory tract was independent of the age of the animals. However, older hamsters exhibited more pronounced and consistent weight loss. In situ hybridization in the lungs identified viral RNA in bronchial epithelium, alveolar epithelial cells type I and II, and macrophages. Histopathology revealed clear age-dependent differences, with young hamsters launching earlier and stronger immune cell influx than aged hamsters. The latter developed conspicuous alveolar and perivascular edema, indicating vascular leakage. In contrast, we observed rapid lung recovery at day 14 after infection only in young hamsters. We propose that comparative assessment in young versus aged hamsters of SARS-CoV-2 vaccines and treatments may yield valuable information, as this small-animal model appears to mirror age-dependent differences in human patients.


Assuntos
Betacoronavirus , Infecções por Coronavirus/etiologia , Modelos Animais de Doenças , Pneumonia Viral/etiologia , Fatores Etários , Animais , Betacoronavirus/imunologia , Betacoronavirus/isolamento & purificação , COVID-19 , Cricetinae , Progressão da Doença , Feminino , Pulmão/virologia , Masculino , Mesocricetus , Pandemias , RNA Viral/análise , SARS-CoV-2 , Vacinas Virais/imunologia
20.
Cell Rep ; 33(10): 108488, 2020 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-33271063

RESUMO

The coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has precipitated an unprecedented and yet-unresolved health crisis worldwide. Different mammals are susceptible to SARS-CoV-2; however, few species examined so far develop robust clinical disease that mirrors severe human cases or allows testing of vaccines and drugs under conditions of severe disease. Here, we compare the susceptibilities of three dwarf hamster species (Phodopus spp.) to SARS-CoV-2 and introduce the Roborovski dwarf hamster (P. roborovskii) as a highly susceptible COVID-19 model with consistent and fulminant clinical signs. Particularly, only this species shows SARS-CoV-2-induced severe acute diffuse alveolar damage and hyaline microthrombi in the lungs, changes described in patients who succumbed to the infection but not reproduced in any experimentally infected animal. Based on our findings, we propose the Roborovski dwarf hamster as a valuable model to examine the efficacy and safety of vaccine candidates and therapeutics, particularly for use in highly susceptible individuals.


Assuntos
COVID-19/virologia , Modelos Animais de Doenças , Pulmão/virologia , Phodopus/virologia , SARS-CoV-2 , Enzima de Conversão de Angiotensina 2/genética , Animais , COVID-19/patologia , COVID-19/fisiopatologia , Pulmão/patologia , Pulmão/fisiopatologia , Alvéolos Pulmonares/fisiopatologia , Alvéolos Pulmonares/virologia , SARS-CoV-2/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA