Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Mol Ther Oncol ; 32(2): 200804, 2024 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-38694569

RESUMO

Despite decades of research, the prognosis of high-grade pediatric brain tumors (PBTs) remains dismal; however, recent cases of favorable clinical responses were documented in clinical trials using oncolytic viruses (OVs). In the current study, we employed four different species of OVs: adenovirus Delta24-RGD, herpes simplex virus rQNestin34.5v1, reovirus R124, and the non-virulent Newcastle disease virus rNDV-F0-GFP against three entities of PBTs (high-grade gliomas, atypical teratoid/rhabdoid tumors, and ependymomas) to determine their in vitro efficacy. These four OVs were screened on 14 patient-derived PBT cell cultures and the degree of oncolysis was assessed using an ATP-based assay. Subsequently, the observed viral efficacies were correlated to whole transcriptome data and Gene Ontology analysis was performed. Although no significant tumor type-specific OV efficacy was observed, the analysis revealed the intrinsic biological processes that associated with OV efficacy. The predictive power of the identified expression profiles was further validated in vitro by screening additional PBTs. In summary, our results demonstrate OV susceptibility of multiple patient-derived PBT entities and the ability to predict in vitro responses to OVs using unique expression profiles. Such profiles may hold promise for future OV preselection with effective oncolytic potency in a specific tumor, therewith potentially improving OV responses.

2.
mSphere ; 9(2): e0074323, 2024 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-38265200

RESUMO

Human metapneumovirus (HMPV), a member of the Pneumoviridae family, causes upper and lower respiratory tract infections in humans. In vitro studies with HMPV have mostly been performed in monolayers of undifferentiated epithelial cells. In vivo studies in cynomolgus macaques and cotton rats have shown that ciliated epithelial cells are the main target of HMPV infection, but these observations cannot be studied in monolayer systems. Here, we established an organoid-derived bronchial culture model that allows physiologically relevant studies on HMPV. Inoculation with multiple prototype HMPV viruses and recent clinical virus isolates led to differences in replication among HMPV isolates. Prolific HMPV replication in this model caused damage to the ciliary layer, including cilia loss at advanced stages post-infection. These cytopathic effects correlated with those observed in previous in vivo studies with cynomolgus macaques. The assessment of the innate immune responses in three donors upon HMPV and RSV inoculation highlighted the importance of incorporating multiple donors to account for donor-dependent variation. In conclusion, these data indicate that the organoid-derived bronchial cell culture model resembles in vivo findings and is therefore a suitable and robust model for future HMPV studies. IMPORTANCE: Human metapneumovirus (HMPV) is one of the leading causative agents of respiratory disease in humans, with no treatment or vaccine available yet. The use of primary epithelial cultures that recapitulate the tissue morphology and biochemistry of the human airways could aid in defining more relevant targets to prevent HMPV infection. For this purpose, this study established the first primary organoid-derived bronchial culture model suitable for a broad range of HMPV isolates. These bronchial cultures were assessed for HMPV replication, cellular tropism, cytopathology, and innate immune responses, where the observations were linked to previous in vivo studies with HMPV. This study exposed an important gap in the HMPV field since extensively cell-passaged prototype HMPV B viruses did not replicate in the bronchial cultures, underpinning the need to use recently isolated viruses with a controlled passage history. These results were reproducible in three different donors, supporting this model to be suitable to study HMPV infection.


Assuntos
Metapneumovirus , Infecções por Paramyxoviridae , Humanos , Animais , Metapneumovirus/fisiologia , Citologia , Replicação Viral , Infecções por Paramyxoviridae/patologia , Epitélio , Macaca , Tropismo
3.
J Virol ; 97(10): e0132523, 2023 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-37823646

RESUMO

IMPORTANCE: Itaconate derivates, as well as the naturally produced metabolite, have been proposed as antivirals against influenza virus. Here, the mechanism behind the antiviral effects of exogenous 4-octyl itaconate (4-OI), a derivative of itaconate, against the influenza A virus replication is demonstrated. The data indicate that 4-OI targets the cysteine at position 528 of the CRM1 protein, resulting in inhibition of the nuclear export of viral ribonucleoprotein complexes in a similar manner as previously described for other selective inhibitors of nuclear export. These results postulate a mechanism not observed before for this immuno-metabolite derivative. This knowledge is helpful for the development of derivatives of 4-OI as potential antiviral and anti-inflammatory therapeutics.


Assuntos
Antivirais , Proteína Exportina 1 , Influenza Humana , Succinatos , Replicação Viral , Humanos , Transporte Ativo do Núcleo Celular , Antivirais/farmacologia , Proteínas Nucleares/metabolismo , Replicação Viral/efeitos dos fármacos , Succinatos/farmacologia , Proteína Exportina 1/metabolismo
4.
iScience ; 26(9): 107698, 2023 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-37680489

RESUMO

Viral sensing in myeloid cells involves inflammasome activation leading to gasdermin pore formation, cytokine release, and cell death. However, less is known about viral sensing in barrier epithelial cells, which are critical to the innate immune response to RNA viruses. Here, we show that poly(I:C), a mimic of viral dsRNA, is sensed by NLRP1 in human bronchial epithelial cells, leading to inflammasome-dependent gasdermin D (GSDMD) pore formation via caspase-1. DsRNA also stimulated a parallel sensing pathway via PKR which activated caspase-3 to cleave gasdermin E (GSDME) to form active pores. Influenza A virus (IAV) infection of cells caused GSDME activation, cytokine release, and cell death, in a PKR-dependent but NLRP1-independent manner, involving caspase-8 and caspase-3. Suppression of GSDMD and GSDME expression increased IAV replication. These data clarify mechanisms of gasdermin cleavage in response to viral sensing and reveal that gasdermin pore formation is intrinsically antiviral in human epithelial cells.

5.
mBio ; 14(1): e0228022, 2023 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-36507832

RESUMO

Human metapneumovirus (HMPV) is one of the leading causes of respiratory illness (RI), primarily in infants. Worldwide, two genetic lineages (A and B) of HMPV are circulating that are antigenically distinct and can each be further divided into genetic sublineages. Surveillance combined with large-scale whole-genome sequencing studies of HMPV are scarce but would help to identify viral evolutionary dynamics. Here, we analyzed 130 whole HMPV genome sequences obtained from samples collected from individuals hospitalized with RI and partial fusion (n = 144) and attachment (n = 123) protein gene sequences obtained from samples collected from patients with RI visiting general practitioners between 2005 and 2021 in the Netherlands. Phylogenetic analyses demonstrated that HMPV continued to group in the four sublineages described in 2004 (A1, A2, B1, and B2). However, one sublineage (A1) was no longer detected in the Netherlands after 2006, while the others continued to evolve. No differences were observed in dominant (sub)lineages between samples obtained from patients with RI being hospitalized and those consulting general practitioners. In both populations, viruses of lineage A2 carrying a 180-nucleotide or 111-nucleotide duplication in the attachment protein gene became the most frequently detected genotypes. In the past, different names for the newly energing lineages have been proposed, demonstrating the need for a consistent naming convention. Here, criteria are proposed for the designation of new genetic lineages to aid in moving toward a systematic HMPV classification. IMPORTANCE Human metapneumovirus (HMPV) is one of the major causative agents of human respiratory tract infections. Monitoring of virus evolution could aid toward the development of new antiviral treatments or vaccine designs. Here, we studied HMPV evolution between 2005 and 2021, with viruses obtained from samples collected from hospitalized individuals and patients with respiratory infections consulting general practitioners. Phylogenetic analyses demonstrated that HMPV continued to group in the four previously described sublineages (A1, A2, B1, and B2). However, one sublineage (A1) was no longer detected after 2006, while the others continued to evolve. No differences were observed in dominant (sub)lineages between patients being hospitalized and those consulting general practitioners. In both populations, viruses of lineage A2 carrying a 180-nucleotide or 111-nucleotide duplication in the attachment protein gene became the most frequently detected genotypes. These data were used to propose criteria for the designation of new genetic lineages to aid toward a systematic HMPV classification.


Assuntos
Metapneumovirus , Infecções por Paramyxoviridae , Infecções Respiratórias , Lactente , Humanos , Metapneumovirus/genética , Infecções por Paramyxoviridae/epidemiologia , Filogenia , Variação Genética , Genótipo , Nucleotídeos
6.
Heliyon ; 8(7): e09915, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-35874055

RESUMO

Pancreatic ductal adenocarcinoma (PDAC) is characterized by a poor clinical prognosis and is usually a metastatic disease. In the last decades, oncolytic viro-immunotherapy has shown a promise as treatment strategy with encouraging results for a variety of tumors. Newcastle Disease Virus (NDV) is an oncolytic virus which selectively infects and damages tumors either by directly killing tumor cells or by promoting an anti-tumor immune response. Several studies have demonstrated that NDV strains with a multi-basic cleavage site (MBCS) in the fusion protein (F) have increased anti-tumor efficacy upon intratumoral injection in murine tumor models. However, intravenous injections, in which the oncolytic virus spreads systemically, could be more beneficial to treat metastasized PDAC in addition to the primary tumor. In this study, we compared the oncolytic efficacy and safety of intratumoral and intravenous injections with NDV containing an MBCS in F (NDV F3aa) in an immune deficient murine xenograft (BxPC3) model for PDAC. In this model, both intratumoral and intravenous injections with NDV F3aa induced anti-tumor efficacy as measured at 10 days after the first injection. Upon intravenous injection virus was detected in some of the tumors, indicating the systemic spread of the virus. Upon both treatments, mice did not display weight loss or abnormalities and treated mice did not secrete virus to the environment. These data demonstrate that intravenous injections of NDV F3aa can be applicable to treat metastasized cancers in immune deficient hosts without inflicting adverse effects.

7.
J Pediatric Infect Dis Soc ; 11(7): 341-344, 2022 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-35390156

RESUMO

Transplacental hMPV-neutralizing antibody transfer was reduced from mothers living with HIV-1. However, a comparison of antibody titers at birth between hMPV hospitalization cases at <6 months and matched controls suggested that reduced maternal antibody might not be the primary cause of the previously reported elevated hMPV risk in HIV-1-exposed infants.


Assuntos
HIV-1 , Metapneumovirus , Infecções por Paramyxoviridae , Anticorpos Neutralizantes , Anticorpos Antivirais , Humanos , Recém-Nascido
8.
PLoS One ; 17(2): e0263707, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35139115

RESUMO

Newcastle Disease Virus (NDV) is an avian RNA virus, which was shown to be effective and safe for use in oncolytic viral therapy for several tumour malignancies. The presence of a multi basic cleavage site (MBCS) in the fusion protein improved its oncolytic efficacy in vitro and in vivo. However, NDV with a MBCS can be virulent in poultry. We aimed to develop an NDV with a MBCS but with reduced virulence for poultry while remaining effective in killing human tumour cells. To this end, the open reading frame of the V protein, an avian specific type I interferon antagonist, was disrupted by introducing multiple mutations. NDV with a mutated V gene was attenuated in avian cells and chicken and duck eggs. Although this virus still killed tumour cells, the efficacy was reduced compared to the virulent NDV. Introduction of various mutations in the fusion (F) and hemagglutinin-neuraminidase (HN) genes slightly improved this efficacy. Taken together, these data demonstrated that NDV with a MBCS but with abrogation of the V protein ORF and mutations in the F and HN genes can be safe for evaluation in oncolytic viral therapy.


Assuntos
Neoplasias/terapia , Vírus da Doença de Newcastle/genética , Terapia Viral Oncolítica , Vírus Oncolíticos , Proteínas Estruturais Virais/genética , Células A549 , Animais , Apoptose/genética , Calibragem , Proteínas do Capsídeo/genética , Células Cultivadas , Embrião de Galinha , Chlorocebus aethiops , Patos/embriologia , Proteína HN/genética , Humanos , Mutagênese Sítio-Dirigida/métodos , Neoplasias/patologia , Vírus da Doença de Newcastle/patogenicidade , Vírus da Doença de Newcastle/fisiologia , Terapia Viral Oncolítica/efeitos adversos , Terapia Viral Oncolítica/métodos , Terapia Viral Oncolítica/normas , Vírus Oncolíticos/genética , Vírus Oncolíticos/patogenicidade , Vírus Oncolíticos/fisiologia , Fases de Leitura Aberta/genética , Segurança do Paciente , Microambiente Tumoral/genética , Células Vero , Proteínas Virais de Fusão/efeitos adversos , Proteínas Virais de Fusão/genética , Virulência/genética , Replicação Viral/genética
9.
Virus Res ; 302: 198490, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34146613

RESUMO

Human metapneumovirus (HMPV), a member of the Pneumoviridae family, is a causative agent of respiratory illness in young children, the elderly, and immunocompromised individuals. Globally, viruses belonging to two main genetic lineages circulate, A and B, which are further divided into four genetic sublineages (A1, A2, B1, B2). Classical genotyping of HMPV is based on the sequence of the fusion (F) and attachment (G) glycoprotein genes, which are under direct antibody-mediated immune pressure. Whole genome sequencing provides more information than sequencing of subgenomic fragments and is therefore a powerful tool for studying virus evolution and disease epidemiology and for identifying transmission events and nosocomial outbreaks. Here, we report a robust method to obtain whole genome sequences for HMPV using MinION Nanopore technology. This assay is able to generate HMPV whole genome sequences from clinical specimens with good coverage of the highly variable G gene and is equally sensitive for strains of all four genetic HMPV sublineages. This method can be used for studying HMPV genetics, epidemiology, and evolutionary dynamics.


Assuntos
Metapneumovirus , Nanoporos , Infecções por Paramyxoviridae , Infecções Respiratórias , Idoso , Criança , Pré-Escolar , Variação Genética , Humanos , Lactente , Metapneumovirus/genética , Filogenia , Tecnologia , Sequenciamento Completo do Genoma
10.
Infect Dis (Lond) ; 53(7): 488-497, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33612055

RESUMO

BACKGROUND: The first outbreak of coronavirus disease 2019 (COVID-19) occurred in March 2020 in Europe, which is normally the peak incidence period of human metapneumovirus (HMPV) infections, implying cocirculation and potentially causing competition between them. METHODS: We investigated differences in clinical characteristics and outcomes of HMPV infections in hospitalized patients before (January 2016-28 February, 2020) and HMPV and COVID-19 during part of the COVID-19 pandemic (28 February, 2020-1 April, 2020). RESULTS: A total of 239 HMPV patients and 303 COVID-19 patients were included. Incidence of HMPV peaked in March. Despite a 324% increase in HMPV testing during the COVID-19 outbreak, incidence of HMPV remained stable. Clinical characteristics showed 25 (11%) ICU admissions and 14 (6%) deaths. History of myocardial infarction, higher age and lower BMI were independently associated with increased 30-day mortality. Clinical characteristics of HMPV-infected patients did not differ between the non-COVID-19 period and the examined COVID-19 period except for length of hospital stay (7 vs. 5 days). HMPV infection and COVID-19 shared many clinical features but HMPV was associated with female gender, elderly patients and chronic conditions (COPD and chronic heart failure). Clinical outcomes did not differ between the viruses during the COVID-19 period. CONCLUSIONS: The clinical impact of HMPV infection did not change during the COVID-19 outbreak in terms of incidence and/or disease severity; hence, HMPV and SARS-CoV-2 are probably co-circulating independently. Despite the current clinical focus on the COVID-19 pandemic, clinicians should keep in mind that HMPV-infection may mimic COVID-19 and is also associated with serious adverse outcomes.


Assuntos
COVID-19 , Metapneumovirus , Infecções por Paramyxoviridae , Infecções Respiratórias , Idoso , Europa (Continente) , Feminino , Humanos , Lactente , Pandemias , Infecções por Paramyxoviridae/epidemiologia , Infecções Respiratórias/epidemiologia , SARS-CoV-2
11.
Cytokine Growth Factor Rev ; 56: 133-140, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32553482

RESUMO

The use of oncolytic viruses forms an appealing approach for cancer treatment. On the one hand the viruses replicate in, and kill, tumor cells, leading to their intra-tumoral amplification. On the other hand the viral infection will activate virus-directed immune responses, and may trigger immune responses directed against tumor cells and tumor antigens. To date, a wide variety of oncolytic viruses is being developed for use in cancer treatment. While the development of oncolytic viruses has often been initiated by researchers in academia and other public institutions, a large majority of the final product development and the testing of these products in clinical trials is industry led. As a consequence relatively few pre-clinical and clinical studies evaluated different oncolytic viruses in competitive side-by-side preclinical or clinical studies. In this review we will summarize the steps and considerations essential in the development and characterization of oncolytic viruses, and describe our multidisciplinary academic consortium, which involves a dozen departments in three different Dutch universities, collaborating in the development of oncolytic viruses. This consortium has the ambition to develop a small series of oncolytic viruses and to evaluate these in various cancers.


Assuntos
Neoplasias , Terapia Viral Oncolítica , Vírus Oncolíticos , Humanos , Neoplasias/terapia
12.
Front Immunol ; 10: 1763, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31404141

RESUMO

Specialized receptors that recognize molecular patterns such as double stranded RNA duplexes-indicative of viral replication-are potent triggers of the innate immune system. Although their activation is beneficial during viral infection, RNA transcribed from endogenous mobile genetic elements may also act as ligands potentially causing autoimmunity. Recent advances indicate that the adenosine deaminase ADAR1 through RNA editing is involved in dampening the canonical antiviral RIG-I-like receptor-, PKR-, and OAS-RNAse L pathways to prevent autoimmunity. However, this inhibitory effect must be overcome during viral infections. In this review we discuss ADAR1's critical role in balancing immune activation and self-tolerance.


Assuntos
Adenosina Desaminase/genética , Adenosina Desaminase/metabolismo , Imunidade Inata , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Animais , Pontos de Checagem do Ciclo Celular , Citoplasma , Proteína DEAD-box 58/metabolismo , Suscetibilidade a Doenças , Endorribonucleases/metabolismo , Humanos , Interferons/metabolismo , Edição de RNA , Transdução de Sinais , eIF-2 Quinase/metabolismo
14.
Hum Vaccin Immunother ; 11(7): 1573-84, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25996182

RESUMO

Oncolytic viruses are a relatively new class of anti-cancer immunotherapy agents. Several viruses have undergone evaluation in clinical trials in the last decades, and the first agent is about to be approved to be used as a novel cancer therapy modality. In the current review, an overview is presented on recent (pre)clinical developments in the field of oncolytic viruses that have previously been or currently are being evaluated in clinical trials. Special attention is given to possible safety issues like toxicity, environmental shedding, mutation and reversion to wildtype virus.


Assuntos
Terapia Viral Oncolítica , Vírus Oncolíticos , Ensaios Clínicos como Assunto , Humanos , Pesquisa Translacional Biomédica , Eliminação de Partículas Virais
15.
Antimicrob Agents Chemother ; 58(8): 4875-84, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24841269

RESUMO

Coronaviruses can cause respiratory and enteric disease in a wide variety of human and animal hosts. The 2003 outbreak of severe acute respiratory syndrome (SARS) first demonstrated the potentially lethal consequences of zoonotic coronavirus infections in humans. In 2012, a similar previously unknown coronavirus emerged, Middle East respiratory syndrome coronavirus (MERS-CoV), thus far causing over 650 laboratory-confirmed infections, with an unexplained steep rise in the number of cases being recorded over recent months. The human MERS fatality rate of ∼ 30% is alarmingly high, even though many deaths were associated with underlying medical conditions. Registered therapeutics for the treatment of coronavirus infections are not available. Moreover, the pace of drug development and registration for human use is generally incompatible with strategies to combat emerging infectious diseases. Therefore, we have screened a library of 348 FDA-approved drugs for anti-MERS-CoV activity in cell culture. If such compounds proved sufficiently potent, their efficacy might be directly assessed in MERS patients. We identified four compounds (chloroquine, chlorpromazine, loperamide, and lopinavir) inhibiting MERS-CoV replication in the low-micromolar range (50% effective concentrations [EC(50)s], 3 to 8 µM). Moreover, these compounds also inhibit the replication of SARS coronavirus and human coronavirus 229E. Although their protective activity (alone or in combination) remains to be assessed in animal models, our findings may offer a starting point for treatment of patients infected with zoonotic coronaviruses like MERS-CoV. Although they may not necessarily reduce viral replication to very low levels, a moderate viral load reduction may create a window during which to mount a protective immune response.


Assuntos
Antivirais/farmacologia , Coronavirus Humano 229E/efeitos dos fármacos , Coronavírus da Síndrome Respiratória do Oriente Médio/efeitos dos fármacos , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/farmacologia , Animais , Linhagem Celular , Chlorocebus aethiops , Cloroquina/farmacologia , Clorpromazina/farmacologia , Coronavirus Humano 229E/fisiologia , Aprovação de Drogas , Hepatócitos/efeitos dos fármacos , Hepatócitos/patologia , Hepatócitos/virologia , Ensaios de Triagem em Larga Escala , Humanos , Concentração Inibidora 50 , Loperamida/farmacologia , Lopinavir/farmacologia , Coronavírus da Síndrome Respiratória do Oriente Médio/fisiologia , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/fisiologia , Células Vero , Replicação Viral/efeitos dos fármacos
16.
J Gen Virol ; 95(Pt 8): 1625-1633, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24760760

RESUMO

Type I IFN production is one of the hallmarks of host innate immune responses upon virus infection. Whilst most respiratory viruses carry IFN antagonists, reports on human metapneumovirus (HMPV) have been conflicting. Using deep sequencing, we have demonstrated that HMPV particles accumulate excessive amounts of defective interfering RNA (DIs) rapidly upon in vitro passage, and that these are associated with IFN induction. Importantly, the DIs were edited extensively; up to 70% of the original A and T residues had mutated to G or C, respectively. Such high editing rates of viral RNA have not, to our knowledge, been reported before. Bioinformatics and PCR assays indicated that adenosine deaminase acting on RNA (ADAR) was the most likely editing enzyme. HMPV thus has an unusually high propensity to generate DIs, which are edited at an unprecedented high frequency. The conflicting published data on HMPV IFN induction and antagonism are probably explained by DIs in virus stocks. The interaction of HMPV DIs with the RNA-editing machinery and IFN responses warrants further investigation.


Assuntos
Interferon Tipo I/imunologia , Metapneumovirus/genética , Metapneumovirus/imunologia , RNA Interferente Pequeno/genética , RNA Viral/metabolismo , Adenosina Desaminase , Animais , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Dados de Sequência Molecular , RNA Interferente Pequeno/metabolismo , RNA Viral/genética , Proteínas de Ligação a RNA
17.
J Virol Methods ; 193(1): 159-65, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23777750

RESUMO

Paramyxoviruses, including measles virus (MV), human metapneumovirus (HMPV), human respiratory syncytial virus (HRSV) and canine distemper virus (CDV), are transmitted via the respiratory route. Despite their close phylogenetic relationship, the pathogenesis of these viruses is very different. To study viral tropism and replication ex vivo, a protocol for the inflation of lungs with low-melting-point agarose mixed with culture medium was established. Lung slices were prepared and remained viable in culture at 37°C for at least 7 days. Lung slices obtained from different animal species were infected with recombinant paramyxoviruses expressing enhanced green fluorescent protein (EGFP). Progression of infection was monitored in real time by detection of EGFP fluorescence. Flow cytometric analysis of cells emigrating into the culture medium proved to be a valuable tool for qualitative and quantitative assessment of infection over time. MV replicated optimally in lung slices of macaques, HMPV and HRSV in lung slices of cotton rats, and CDV in lung slices obtained from dogs and ferrets. In conclusion, these ex vivo lung slice cultures are discriminatory models for the study of respiratory virus infections and can be used to inform the design of future in vivo experiments, thereby reducing numbers of animals required.


Assuntos
Pulmão/virologia , Paramyxovirinae/fisiologia , Replicação Viral , Animais , Cães , Macaca , Técnicas de Cultura de Órgãos/métodos , Paramyxovirinae/isolamento & purificação , Sigmodontinae , Cultura de Vírus/métodos
18.
J Gen Virol ; 94(Pt 8): 1749-1760, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23620378

RESUMO

Coronavirus (CoV) infections are commonly associated with respiratory and enteric disease in humans and animals. The 2003 outbreak of severe acute respiratory syndrome (SARS) highlighted the potentially lethal consequences of CoV-induced disease in humans. In 2012, a novel CoV (Middle East Respiratory Syndrome coronavirus; MERS-CoV) emerged, causing 49 human cases thus far, of which 23 had a fatal outcome. In this study, we characterized MERS-CoV replication and cytotoxicity in human and monkey cell lines. Electron microscopy of infected Vero cells revealed extensive membrane rearrangements, including the formation of double-membrane vesicles and convoluted membranes, which have been implicated previously in the RNA synthesis of SARS-CoV and other CoVs. Following infection, we observed rapidly increasing viral RNA synthesis and release of high titres of infectious progeny, followed by a pronounced cytopathology. These characteristics were used to develop an assay for antiviral compound screening in 96-well format, which was used to identify cyclosporin A as an inhibitor of MERS-CoV replication in cell culture. Furthermore, MERS-CoV was found to be 50-100 times more sensitive to alpha interferon (IFN-α) treatment than SARS-CoV, an observation that may have important implications for the treatment of MERS-CoV-infected patients. MERS-CoV infection did not prevent the IFN-induced nuclear translocation of phosphorylated STAT1, in contrast to infection with SARS-CoV where this block inhibits the expression of antiviral genes. These findings highlight relevant differences between these distantly related zoonotic CoVs in terms of their interaction with and evasion of the cellular innate immune response.


Assuntos
Antivirais/farmacologia , Coronavirus/efeitos dos fármacos , Coronavirus/fisiologia , Ciclosporina/farmacologia , Efeito Citopatogênico Viral , Interferon-alfa/farmacologia , Replicação Viral , Animais , Linhagem Celular , Membrana Celular/ultraestrutura , Chlorocebus aethiops , Coronavirus/patogenicidade , Avaliação Pré-Clínica de Medicamentos , Humanos , Testes de Sensibilidade Microbiana , Microscopia Eletrônica de Transmissão
19.
Vaccine ; 26(33): 4224-30, 2008 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-18585830

RESUMO

Human metapneumovirus (HMPV) is an important cause of acute respiratory tract disease for which the development of vaccine candidates is warranted. We have previously described the generation of an iscom matrix-adjuvanted HMPV fusion protein subunit vaccine (Fsol) and a live-attenuated vaccine (HMPVM11). Here, we evaluate the immunogenicity and efficacy of these vaccines in cynomolgus macaques. Immunization with Fsol induced HMPV F-specific antibody responses, virus neutralizing antibody titers, and cellular immune responses, but the induced humoral immune response waned rapidly over time. HMPVM11 was strongly attenuated and displayed limited immunogenicity, although immunization with this virus primed for a good secondary HMPV-specific lymphoproliferative response after challenge infection. The duration of virus shedding in HMPVM11-immunized animals was reduced compared to sham-immunized animals. Both vaccines induced HMPV-specific immune responses, but the rapid waning of immunity is a challenging obstacle for vaccine development.


Assuntos
Metapneumovirus/imunologia , Infecções por Paramyxoviridae/prevenção & controle , Vacinas Virais/imunologia , Animais , Anticorpos Antivirais/sangue , Proliferação de Células , Humanos , Imunidade Celular , Linfócitos/imunologia , Macaca fascicularis , Testes de Neutralização , Infecções Respiratórias/prevenção & controle , Fatores de Tempo , Vacinas Atenuadas/imunologia , Vacinas de Subunidades Antigênicas/imunologia , Eliminação de Partículas Virais
20.
J Gen Virol ; 89(Pt 7): 1553-1562, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18559924

RESUMO

Human metapneumovirus (HMPV) causes acute respiratory tract illness primarily in young children, immunocompromised individuals and the elderly. Vaccines would be desirable to prevent severe illnesses in these risk groups. Here, we describe the generation and evaluation of cold-passage (cp) temperature-sensitive (ts) HMPV strains as vaccine candidates. Repeated passage of HMPV at low temperatures in Vero cells resulted in the accumulation of mutations in the viral genome. Introduction of these mutations in a recombinant HMPV by reverse genetics resulted in a ts-phenotype, judged on the decreased shut-off temperature for virus replication in vitro. As an alternative approach, three previously described cp-respiratory syncytial virus (cp-HRSV) mutations were introduced in a recombinant HMPV, which also resulted in a low shut-off temperature in vitro. Replication of these ts-viruses containing either the cp-HMPV or cp-HRSV mutations was reduced in the upper respiratory tract (URT) and undetectable in the lower respiratory tract (LRT) of hamsters. Nevertheless, high titres of HMPV-specific antibodies were induced by both ts-viruses. Upon immunization with the ts-viruses, the LRT of hamsters were completely protected against challenge infection with a heterologous HMPV strain, and URT viral titres were reduced by 10 000-fold. In conclusion, we provide proof-of-principle for two candidate live-attenuated HMPV vaccines that induce cross-protective immunity to prevent infection of the LRT in Syrian golden hamsters. Further mapping of the molecular determinants of attenuation of HMPV should be the subject of future studies.


Assuntos
Temperatura Alta , Metapneumovirus/crescimento & desenvolvimento , Metapneumovirus/imunologia , Infecções por Paramyxoviridae/imunologia , Infecções por Paramyxoviridae/prevenção & controle , Vacinas Virais/imunologia , Animais , Anticorpos Antivirais/sangue , Chlorocebus aethiops , Cricetinae , Feminino , Pulmão/virologia , Metapneumovirus/genética , Mutação , Pneumonia Viral/prevenção & controle , Vírus Sinciciais Respiratórios/genética , Sistema Respiratório/virologia , Infecções Respiratórias/prevenção & controle , Infecções Respiratórias/virologia , Células Vero , Vacinas Virais/genética , Replicação Viral/efeitos da radiação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA