Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
PLoS Pathog ; 18(5): e1010485, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35587473

RESUMO

Crimean-Congo hemorrhagic fever virus (CCHFV) is an important human pathogen. In cell culture, CCHFV is sensed by the cytoplasmic RNA sensor retinoic acid-inducible gene I (RIG-I) molecule and its adaptor molecule mitochondrial antiviral signaling (MAVS) protein. MAVS initiates both type I interferon (IFN-I) and proinflammatory responses. Here, we studied the role MAVS plays in CCHFV infection in mice in both the presence and absence of IFN-I activity. MAVS-deficient mice were not susceptible to CCHFV infection when IFN-I signaling was active and showed no signs of disease. When IFN-I signaling was blocked by antibody, MAVS-deficient mice lost significant weight, but were uniformly protected from lethal disease, whereas all control mice succumbed to infection. Cytokine activity in the infected MAVS-deficient mice was markedly blunted. Subsequent investigation revealed that CCHFV infected mice lacking TNF-α receptor signaling (TNFA-R-deficient), but not IL-6 or IL-1 activity, had more limited liver injury and were largely protected from lethal outcomes. Treatment of mice with an anti-TNF-α neutralizing antibody also conferred partial protection in a post-virus exposure setting. Additionally, we found that a disease causing, but non-lethal strain of CCHFV produced more blunted inflammatory cytokine responses compared to a lethal strain in mice. Our work reveals that MAVS activation and cytokine production both contribute to CCHFV pathogenesis, potentially identifying new therapeutic targets to treat this disease.


Assuntos
Vírus da Febre Hemorrágica da Crimeia-Congo , Febre Hemorrágica da Crimeia , Animais , Citocinas , Modelos Animais de Doenças , Vírus da Febre Hemorrágica da Crimeia-Congo/genética , Camundongos , Camundongos Knockout , Índice de Gravidade de Doença , Inibidores do Fator de Necrose Tumoral
2.
J Virol ; 96(3): e0150421, 2022 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-34851148

RESUMO

In the age of COVID, nucleic acid vaccines have garnered much attention, at least in part, because of the simplicity of construction, production, and flexibility to adjust and adapt to an evolving outbreak. Orthopoxviruses remain a threat on multiple fronts, especially as emerging zoonoses. In response, we developed a DNA vaccine, termed 4pox, that protected nonhuman primates against monkeypox virus (MPXV)-induced severe disease. Here, we examined the protective efficacy of the 4pox DNA vaccine delivered by intramuscular (i.m.) electroporation (EP) in rabbits challenged with aerosolized rabbitpox virus (RPXV), a model that recapitulates the respiratory route of exposure and low dose associated with natural smallpox exposure in humans. We found that 4pox-vaccinated rabbits developed immunogen-specific antibodies, including neutralizing antibodies, and did not develop any clinical disease, indicating protection against aerosolized RPXV. In contrast, unvaccinated animals developed significant signs of disease, including lesions, and were euthanized. These findings demonstrate that an unformulated, nonadjuvanted DNA vaccine delivered i.m. can protect against an aerosol exposure. IMPORTANCE The eradication of smallpox and subsequent cessation of vaccination have left a majority of the population susceptible to variola virus or other emerging poxviruses. This is exemplified by human monkeypox, as evidenced by the increase in reported endemic and imported cases over the past decades. Therefore, a malleable vaccine technology that can be mass produced and does not require complex conditions for distribution and storage is sought. Herein, we show that a DNA vaccine, in the absence of a specialized formulation or adjuvant, can protect against a lethal aerosol insult of rabbitpox virus.


Assuntos
Vacinas Baseadas em Ácido Nucleico/imunologia , Orthopoxvirus/imunologia , Infecções por Poxviridae/prevenção & controle , Vaccinia virus/imunologia , Vacínia/prevenção & controle , Proteínas Virais/imunologia , Vacinas Virais/imunologia , Animais , Linfócitos B/imunologia , Linfócitos B/metabolismo , Relação Dose-Resposta Imunológica , Eletroporação , Feminino , Imunização/métodos , Imunogenicidade da Vacina , Ativação Linfocitária/imunologia , Vacinas Baseadas em Ácido Nucleico/administração & dosagem , Oligodesoxirribonucleotídeos/administração & dosagem , Oligodesoxirribonucleotídeos/imunologia , Coelhos , Vacinas de DNA/imunologia , Vaccinia virus/genética , Vacinas Virais/administração & dosagem
3.
J Gen Virol ; 102(5)2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33961540

RESUMO

SARS-CoV-2 is the causative agent of COVID-19 and human infections have resulted in a global health emergency. Small animal models that reproduce key elements of SARS-CoV-2 human infections are needed to rigorously screen candidate drugs to mitigate severe disease and prevent the spread of SARS-CoV-2. We and others have reported that transgenic mice expressing the human angiotensin-converting enzyme 2 (hACE2) viral receptor under the control of the Keratin 18 (K18) promoter develop severe and lethal respiratory disease subsequent to SARS-CoV-2 intranasal challenge. Here we report that some infected mice that survive challenge have residual pulmonary damages and persistent brain infection on day 28 post-infection despite the presence of anti-SARS-COV-2 neutralizing antibodies. Because of the hypersensitivity of K18-hACE2 mice to SARS-CoV-2 and the propensity of virus to infect the brain, we sought to determine if anti-infective biologics could protect against disease in this model system. We demonstrate that anti-SARS-CoV-2 human convalescent plasma protects K18-hACE2 against severe disease. All control mice succumbed to disease by day 7; however, all treated mice survived infection without observable signs of disease. In marked contrast to control mice, viral antigen and lesions were reduced or absent from lungs and absent in brains of antibody-treated mice. Our findings support the use of K18-hACE2 mice for protective efficacy studies of anti-SARS-CoV-2 medical countermeasures (MCMs). They also support the use of this system to study SARS-CoV-2 persistence and host recovery.


Assuntos
COVID-19/terapia , Lesão Pulmonar Aguda/prevenção & controle , Lesão Pulmonar Aguda/virologia , Enzima de Conversão de Angiotensina 2/genética , Enzima de Conversão de Angiotensina 2/metabolismo , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Encéfalo/patologia , Encéfalo/virologia , COVID-19/imunologia , COVID-19/patologia , COVID-19/virologia , Modelos Animais de Doenças , Feminino , Humanos , Imunização Passiva , Pulmão/patologia , Pulmão/virologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Receptores de Coronavírus/genética , Receptores de Coronavírus/metabolismo , SARS-CoV-2/imunologia , SARS-CoV-2/isolamento & purificação , SARS-CoV-2/fisiologia , Índice de Gravidade de Doença , Carga Viral , Replicação Viral , Soroterapia para COVID-19
4.
J Virol ; 94(22)2020 10 27.
Artigo em Inglês | MEDLINE | ID: mdl-32900822

RESUMO

Animal models recapitulating human COVID-19 disease, especially severe disease, are urgently needed to understand pathogenesis and to evaluate candidate vaccines and therapeutics. Here, we develop novel severe-disease animal models for COVID-19 involving disruption of adaptive immunity in Syrian hamsters. Cyclophosphamide (CyP) immunosuppressed or RAG2 knockout (KO) hamsters were exposed to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) by the respiratory route. Both the CyP-treated and RAG2 KO hamsters developed clinical signs of disease that were more severe than those in immunocompetent hamsters, notably weight loss, viral loads, and fatality (RAG2 KO only). Disease was prolonged in transiently immunosuppressed hamsters and was uniformly lethal in RAG2 KO hamsters. We evaluated the protective efficacy of a neutralizing monoclonal antibody and found that pretreatment, even in immunosuppressed animals, limited infection. Our results suggest that functional B and/or T cells are not only important for the clearance of SARS-CoV-2 but also play an early role in protection from acute disease.IMPORTANCE Syrian hamsters are in use as a model of disease caused by SARS-CoV-2. Pathology is pronounced in the upper and lower respiratory tract, and disease signs and endpoints include weight loss and viral RNA and/or infectious virus in swabs and organs (e.g., lungs). However, a high dose of virus is needed to produce disease, and the disease resolves rapidly. Here, we demonstrate that immunosuppressed hamsters are susceptible to low doses of virus and develop more severe and prolonged disease. We demonstrate the efficacy of a novel neutralizing monoclonal antibody using the cyclophosphamide transient suppression model. Furthermore, we demonstrate that RAG2 knockout hamsters develop severe/fatal disease when exposed to SARS-CoV-2. These immunosuppressed hamster models provide researchers with new tools for evaluating therapies and vaccines and understanding COVID-19 pathogenesis.


Assuntos
Infecções por Coronavirus/imunologia , Infecções por Coronavirus/patologia , Modelos Animais de Doenças , Mesocricetus , Pneumonia Viral/imunologia , Pneumonia Viral/patologia , Imunidade Adaptativa , Animais , Animais Geneticamente Modificados , Betacoronavirus/fisiologia , COVID-19 , Ciclofosfamida , Proteínas de Ligação a DNA/genética , Técnicas de Inativação de Genes , Imunossupressores , Pandemias , SARS-CoV-2 , Índice de Gravidade de Doença
5.
PLoS Pathog ; 15(9): e1008050, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31557262

RESUMO

Crimean-Congo hemorrhagic fever (CCHF) is the most medically important tick-borne viral disease of humans and tuberculosis is the leading cause of death worldwide by a bacterial pathogen. These two diseases overlap geographically, however, concurrent infection of CCHF virus (CCHFV) with mycobacterial infection has not been assessed nor has the ability of virus to persist and cause long-term sequela in a primate model. In this study, we compared the disease progression of two diverse strains of CCHFV in the recently described cynomolgus macaque model. All animals demonstrated signs of clinical illness, viremia, significant changes in clinical chemistry and hematology values, and serum cytokine profiles consistent with CCHF in humans. The European and Asian CCHFV strains caused very similar disease profiles in monkeys, which demonstrates that medical countermeasures can be evaluated in this animal model against multiple CCHFV strains. We identified evidence of CCHFV persistence in the testes of three male monkeys that survived infection. Furthermore, the histopathology unexpectedly revealed that six additional animals had evidence of a latent mycobacterial infection with granulomatous lesions. Interestingly, CCHFV persisted within the granulomas of two animals. This study is the first to demonstrate the persistence of CCHFV in the testes and within the granulomas of non-human primates with concurrent latent tuberculosis. Our results have important public health implications in overlapping endemic regions for these emerging pathogens.


Assuntos
Febre Hemorrágica da Crimeia/complicações , Tuberculose Latente/complicações , Testículo/patologia , Animais , Anticorpos Antivirais/sangue , Doenças Transmissíveis Emergentes/complicações , Doenças Transmissíveis Emergentes/patologia , Doenças Transmissíveis Emergentes/virologia , Citocinas/sangue , Modelos Animais de Doenças , Progressão da Doença , Granuloma/microbiologia , Granuloma/patologia , Granuloma/virologia , Vírus da Febre Hemorrágica da Crimeia-Congo/genética , Vírus da Febre Hemorrágica da Crimeia-Congo/imunologia , Vírus da Febre Hemorrágica da Crimeia-Congo/patogenicidade , Febre Hemorrágica da Crimeia/patologia , Febre Hemorrágica da Crimeia/virologia , Interações entre Hospedeiro e Microrganismos/imunologia , Humanos , Tuberculose Latente/microbiologia , Tuberculose Latente/patologia , Macaca fascicularis , Masculino , Testículo/microbiologia , Testículo/virologia
6.
J Virol ; 92(21)2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-30111561

RESUMO

Crimean-Congo hemorrhagic fever virus (CCHFV) can cause severe hepatic injury in humans. However, the mechanism(s) causing this damage is poorly characterized. CCHFV produces an acute disease, including liver damage, in mice lacking type I interferon (IFN-I) signaling due to either STAT-1 gene deletion or disruption of the IFN-I receptor 1 gene. Here, we explored CCHFV-induced liver pathogenesis in mice using an antibody to disrupt IFN-I signaling. When IFN-I blockade was induced within 24 h postexposure to CCHFV, mice developed severe disease with greater than 95% mortality by 6 days postexposure. In addition, we observed increased proinflammatory cytokines, chemoattractants, and liver enzymes in these mice. Extensive liver damage was evident by 4 days postexposure and was characterized by hepatocyte necrosis and the loss of CLEC4F-positive Kupffer cells. Similar experiments in CCHFV-exposed NOD-SCID-γ (NSG), Rag2-deficient, and perforin-deficient mice also demonstrated liver injury, suggesting that cytotoxic immune cells are dispensable for hepatic damage. Some apoptotic liver cells contained viral RNA, while other apoptotic liver cells were negative, suggesting that cell death occurred by both intrinsic and extrinsic mechanisms. Protein and transcriptional analysis of livers revealed that activation of tumor necrosis factor superfamily members occurred by day 4 postexposure, implicating these molecules as factors in liver cell death. These data provide insights into CCHFV-induced hepatic injury and demonstrate the utility of antibody-mediated IFN-I blockade in the study of CCHFV pathogenesis in mice.IMPORTANCE CCHFV is an important human pathogen that is both endemic and emerging throughout Asia, Africa, and Europe. A common feature of acute disease is liver injury ranging from mild to fulminant hepatic failure. The processes through which CCHFV induces severe liver injury are unclear, mostly due to the limitations of existing small-animal systems. The only small-animal model in which CCHFV consistently produces severe liver damage is mice lacking IFN-I signaling. In this study, we used antibody-mediated blockade of IFN-I signaling in mice to study CCHFV liver pathogenesis in various transgenic mouse systems. We found that liver injury did not depend on cytotoxic immune cells and observed extensive activation of death receptor signaling pathways in the liver during acute disease. Furthermore, acute CCHFV infection resulted in a nearly complete loss of Kupffer cells. Our model system provides insight into both the molecular and the cellular features of CCHFV hepatic injury.


Assuntos
Vírus da Febre Hemorrágica da Crimeia-Congo/patogenicidade , Febre Hemorrágica da Crimeia/patologia , Hepatócitos/patologia , Interferon Tipo I/antagonistas & inibidores , Células de Kupffer/citologia , Falência Hepática Aguda/patologia , Fígado/patologia , Animais , Anticorpos Bloqueadores/imunologia , Linhagem Celular , Chlorocebus aethiops , Citocinas/sangue , Modelos Animais de Doenças , Hepatócitos/virologia , Humanos , Interferon Tipo I/imunologia , Células de Kupffer/virologia , Fígado/lesões , Fígado/virologia , Falência Hepática Aguda/virologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Células Vero
7.
J Virol ; 90(7): 3515-29, 2016 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-26792737

RESUMO

UNLABELLED: Several members of the Arenaviridae can cause acute febrile diseases in humans, often resulting in lethality. The use of convalescent-phase human plasma is an effective treatment in humans infected with arenaviruses, particularly species found in South America. Despite this, little work has focused on developing potent and defined immunotherapeutics against arenaviruses. In the present study, we produced arenavirus neutralizing antibodies by DNA vaccination of rabbits with plasmids encoding the full-length glycoprotein precursors of Junín virus (JUNV), Machupo virus (MACV), and Guanarito virus (GTOV). Geometric mean neutralizing antibody titers, as measured by the 50% plaque reduction neutralization test (PRNT(50)), exceeded 5,000 against homologous viruses. Antisera against each targeted virus exhibited limited cross-species binding and, to a lesser extent, cross-neutralization. Anti-JUNV glycoprotein rabbit antiserum protected Hartley guinea pigs from lethal intraperitoneal infection with JUNV strain Romero when the antiserum was administered 2 days after challenge and provided some protection (∼30%) when administered 4 days after challenge. Treatment starting on day 6 did not protect animals. We further formulated an IgG antibody cocktail by combining anti-JUNV, -MACV, and -GTOV antibodies produced in DNA-vaccinated rabbits. This cocktail protected 100% of guinea pigs against JUNV and GTOV lethal disease. We then expanded on this cocktail approach by simultaneously vaccinating rabbits with a combination of plasmids encoding glycoproteins from JUNV, MACV, GTOV, and Sabia virus (SABV). Sera collected from rabbits vaccinated with the combination vaccine neutralized all four targets. These findings support the concept of using a DNA vaccine approach to generate a potent pan-arenavirus immunotherapeutic. IMPORTANCE: Arenaviruses are an important family of emerging viruses. In infected humans, convalescent-phase plasma containing neutralizing antibodies can mitigate the severity of disease caused by arenaviruses, particularly species found in South America. Because of variations in potency of the human-derived product, limited availability, and safety concerns, this treatment option has essentially been abandoned. Accordingly, despite this approach being an effective postinfection treatment option, research on novel approaches to produce potent polyclonal antibody-based therapies have been deficient. Here we show that DNA-based vaccine technology can be used to make potently neutralizing antibodies in rabbits that exclusively target the glycoproteins of several human-pathogenic arenaviruses found in South America, including JUNV, MACV, GTOV, and SABV. These antibodies protected guinea pigs from lethal disease when given post-virus challenge. We also generated a purified antibody cocktail with antibodies targeting three arenaviruses and demonstrated protective efficacy against all three targets. Our findings demonstrate that use of the DNA vaccine technology could be used to produce candidate antiarenavirus neutralizing antibody-based products.


Assuntos
Anticorpos Neutralizantes/administração & dosagem , Anticorpos Antivirais/administração & dosagem , Antígenos Virais/imunologia , Arenavirus do Novo Mundo/imunologia , Glicoproteínas/imunologia , Febre Hemorrágica Americana/prevenção & controle , Imunização Passiva/métodos , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Modelos Animais de Doenças , Feminino , Cobaias , Imunoglobulina G/administração & dosagem , Imunoglobulina G/imunologia , Testes de Neutralização , Coelhos , Análise de Sobrevida , Resultado do Tratamento , Vacinas de DNA/administração & dosagem , Vacinas de DNA/imunologia
8.
Vaccines (Basel) ; 12(6)2024 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-38932403

RESUMO

Convalescent plasma has been shown to be effective at protecting humans against severe diseases caused by New World (NW) arenaviruses, including Junin virus (JUNV) and Machupo virus (MACV). This plasma contains antibodies against the full complement of structural proteins including the nucleocapsid and envelope glycoproteins (GPcs) consisting of GP1 and GP2. To gain insights into the protective and cross-protective properties of anti-GPc-specific polyclonal antibodies, we evaluated the ability of a DNA vaccine-produced anti-GPc rabbit antisera targeting MACV strain Carvallo to provide heterologous protection against another MACV strain termed Chicava in the Hartley guinea pig model. The neutralizing activity of the rabbit antisera against the heterologous MACV strains Chicava and Mallale was found to be 54-fold and 23-fold lower, respectively, compared to the titer against the homologous MACV strain Carvallo in the PRNT50 assay. Despite lower neutralizing activity against the strain Chicava, the rabbit antisera protected 100% of the guinea pigs from this strain when administered up to four days post-infection, whereas all the control animals succumbed to the disease. Using vesicular stomatitis virus (VSV) particles pseudotyped with MACV GPc, we identified a single amino acid difference at position 122 between the strains Chicava and Carvallo GPc that significantly influenced the neutralization activity of the rabbit antisera. These findings indicate that polyclonal antibodies targeting the MACV glycoproteins can protect against lethal infection in a post-challenge setting. These data will help guide future antibody-based therapeutics development against NW arenaviruses.

9.
Nat Commun ; 15(1): 1722, 2024 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-38409240

RESUMO

Crimean-Congo hemorrhagic fever virus (CCHFV) is a WHO priority pathogen. Antibody-based medical countermeasures offer an important strategy to mitigate severe disease caused by CCHFV. Most efforts have focused on targeting the viral glycoproteins. However, glycoproteins are poorly conserved among viral strains. The CCHFV nucleocapsid protein (NP) is highly conserved between CCHFV strains. Here, we investigate the protective efficacy of a CCHFV monoclonal antibody targeting the NP. We find that an anti-NP monoclonal antibody (mAb-9D5) protected female mice against lethal CCHFV infection or resulted in a significant delay in mean time-to-death in mice that succumbed to disease compared to isotype control animals. Antibody protection is independent of Fc-receptor functionality and complement activity. The antibody bound NP from several CCHFV strains and exhibited robust cross-protection against the heterologous CCHFV strain Afg09-2990. Our work demonstrates that the NP is a viable target for antibody-based therapeutics, providing another direction for developing immunotherapeutics against CCHFV.


Assuntos
Vírus da Febre Hemorrágica da Crimeia-Congo , Febre Hemorrágica da Crimeia , Feminino , Animais , Camundongos , Vírus da Febre Hemorrágica da Crimeia-Congo/metabolismo , Proteínas do Nucleocapsídeo/metabolismo , Anticorpos Monoclonais , Febre Hemorrágica da Crimeia/prevenção & controle , Glicoproteínas/metabolismo , Anticorpos Antivirais
10.
Microbiol Spectr ; 12(4): e0358623, 2024 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-38391232

RESUMO

Although smallpox has been eradicated, other orthopoxviruses continue to be a public health concern as exemplified by the ongoing Mpox (formerly monkeypox) global outbreak. While medical countermeasures (MCMs) previously approved by the Food and Drug Administration for the treatment of smallpox have been adopted for Mpox, previously described vulnerabilities coupled with the questionable benefit of at least one of the therapeutics during the 2022 Mpox outbreak reinforce the need for identifying and developing other MCMs against orthopoxviruses. Here, we screened a panel of Merck proprietary small molecules and identified a novel nucleoside inhibitor with potent broad-spectrum antiviral activity against multiple orthopoxviruses. Efficacy testing of a 7-day dosing regimen of the orally administered nucleoside in a murine model of severe orthopoxvirus infection yielded a dose-dependent increase in survival. Treated animals had greatly reduced lesions in the lung and nasal cavity, particularly in the 10 µg/mL dosing group. Viral levels were also markedly lower in the UMM-766-treated animals. This work demonstrates that this nucleoside analog has anti-orthopoxvirus efficacy and can protect against severe disease in a murine orthopox model.IMPORTANCEThe recent monkeypox virus pandemic demonstrates that members of the orthopoxvirus, which also includes variola virus, which causes smallpox, remain a public health issue. While currently FDA-approved treatment options exist, risks that resistant strains of orthopoxviruses may arise are a great concern. Thus, continued exploration of anti-poxvirus treatments is warranted. Here, we developed a template for a high-throughput screening assay to identify anti-poxvirus small-molecule drugs. By screening available drug libraries, we identified a compound that inhibited orthopoxvirus replication in cell culture. We then showed that this drug can protect animals against severe disease. Our findings here support the use of existing drug libraries to identify orthopoxvirus-targeting drugs that may serve as human-safe products to thwart future outbreaks.


Assuntos
Mpox , Orthopoxvirus , Varíola , Vírus da Varíola , Animais , Camundongos , Humanos , Nucleosídeos/uso terapêutico , Varíola/tratamento farmacológico , Varíola/prevenção & controle , Modelos Animais de Doenças
11.
J Virol ; 86(2): 1238-43, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22072772

RESUMO

Virion uncoating is an essential early event in reovirus infection. In natural enteric infections, rapid proteolytic uncoating of virions is mediated by pancreatic serine proteases. The proteases that promote reovirus disassembly and cell entry in the respiratory tract remain unknown. In this report, we show that endogenous respiratory and inflammatory proteases can promote reovirus infection in vitro and that preexisting inflammation augments in vivo infection in the murine respiratory tract.


Assuntos
Interações Hospedeiro-Patógeno , Orthoreovirus de Mamíferos/fisiologia , Infecções por Reoviridae/enzimologia , Sistema Respiratório/enzimologia , Serina Proteases/metabolismo , Animais , Humanos , Camundongos , Camundongos Endogâmicos CBA , Orthoreovirus de Mamíferos/genética , Infecções por Reoviridae/imunologia , Infecções por Reoviridae/virologia , Sistema Respiratório/imunologia , Sistema Respiratório/virologia , Serina Proteases/genética
12.
Virus Res ; 334: 199173, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37459918

RESUMO

Crimean-Congo hemorrhagic fever (CCHF) is a World Health Organization prioritized disease because its broad distribution and severity of disease make it a global health threat. Despite advancements in preclinical vaccine development for CCHF virus (CCHFV), including multiple platforms targeting multiple antigens, a clear definition of the adaptive immune correlates of protection is lacking. Levels of neutralizing antibodies in vaccinated animal models do not necessarily correlate with protection, suggesting that cellular immunity, such as CD8+ T cells, might have an important role in protection in this model. Using a well-established IFN-I antibody blockade mouse model (IS) and a DNA-based vaccine encoding the CCHFV M-segment glycoprotein precursor, we investigated the role of humoral and T cell immunity in vaccine-mediated protection in mice genetically devoid of these immune compartments. We found that in the absence of the B-cell compartment (µMT knockout mice), protection provided by the vaccine was not reduced. In contrast, in the absence of CD8+ T cells (CD8+ knockout mice) the vaccine-mediated protection was significantly diminished. Importantly, humoral responses to the vaccine in CD8+ T-cell knockout mice were equivalent to wild-type mice. These findings indicated that CD8+ T-cell responses are necessary and sufficient to promote protection in mice vaccinated with the M-segment DNA vaccine. Identifying a crucial role of the cellular immunity to protect against CCHFV should help guide the development of CCHFV-targeting vaccines.


Assuntos
Vírus da Febre Hemorrágica da Crimeia-Congo , Febre Hemorrágica da Crimeia , Vacinas de DNA , Animais , Camundongos , Vírus da Febre Hemorrágica da Crimeia-Congo/genética , Vacinas de DNA/genética , Linfócitos T CD8-Positivos , Camundongos Knockout
13.
PLoS Negl Trop Dis ; 17(9): e0011620, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37682988

RESUMO

Numerous arenaviruses have been identified throughout the Americas and a subset of these viruses cause viral hemorrhagic fever in humans. This study compared the pathology and viral RNA distribution in Hartley guinea pigs challenged with two human-disease causing New World arenaviruses, Junin virus (JUNV) or Guanarito virus (GTOV). Histopathologic analysis and RNA in situ hybridization revealed similar pathology and viral RNA distribution for both groups of animals challenged with either JUNV or GTOV on days 3, 7, 10 and 12 post exposure (PE). Gross lesions were first observed on day 7 and primarily involved the lungs and liver. The most severe histologic lesions occurred in the lymph nodes, spleen, and thymus and included lymphoid depletion and necrosis which increased in severity over time. Extensive necrosis was also observed in the bone marrow on day 12. Minimal to mild inflammation with and without necrosis was observed in the choroid plexus of the brain, choroid of the eye, intestinal tract, lung and adrenal gland. Significant liver lesions were rare, consisting predominantly of hepatocyte vacuolation. Viral RNA labeling was identified in nearly all organs examined, was often extensive in certain organs and generally increased over time starting on day 7. Our data demonstrate the guinea pig may serve as a useful model to study New World arenavirus infection in humans and for the evaluation and development of medical countermeasures.


Assuntos
Arenavirus do Novo Mundo , Vírus Junin , Humanos , Cobaias , Animais , RNA Viral/genética , Fígado , Encéfalo
14.
Nat Commun ; 13(1): 7298, 2022 11 26.
Artigo em Inglês | MEDLINE | ID: mdl-36435827

RESUMO

Crimean-Congo Hemorrhagic Fever Virus (CCHFV) causes a life-threatening disease with up to a 40% mortality rate. With no approved medical countermeasures, CCHFV is considered a public health priority agent. The non-neutralizing mouse monoclonal antibody (mAb) 13G8 targets CCHFV glycoprotein GP38 and protects mice from lethal CCHFV challenge when administered prophylactically or therapeutically. Here, we reveal the structures of GP38 bound with a human chimeric 13G8 mAb and a newly isolated CC5-17 mAb from a human survivor. These mAbs bind overlapping epitopes with a shifted angle. The broad-spectrum potential of c13G8 and CC5-17 and the practicality of using them against Aigai virus, a closely related nairovirus were examined. Binding studies demonstrate that the presence of non-conserved amino acids in Aigai virus corresponding region prevent CCHFV mAbs from binding Aigai virus GP38. This information, coupled with in vivo efficacy, paves the way for future mAb therapeutics effective against a wide swath of CCHFV strains.


Assuntos
Vírus da Febre Hemorrágica da Crimeia-Congo , Febre Hemorrágica da Crimeia , Camundongos , Humanos , Animais , Vírus da Febre Hemorrágica da Crimeia-Congo/química , Febre Hemorrágica da Crimeia/prevenção & controle , Anticorpos Neutralizantes , Anticorpos Antivirais , Epitopos , Anticorpos Monoclonais
15.
mBio ; 13(1): e0290621, 2022 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-35073750

RESUMO

The rapid emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has created a global health emergency. While most human disease is mild to moderate, some infections lead to a severe disease characterized by acute respiratory distress, hypoxia, anosmia, ageusia, and, in some instances, neurological involvement. Small-animal models reproducing severe disease, including neurological sequela, are needed to characterize the pathophysiological mechanism(s) of disease and to identify medical countermeasures. Transgenic mice expressing the human angiotensin-converting enzyme 2 (hACE2) viral receptor under the control of the K18 promoter develop severe and lethal respiratory disease subsequent to SARS-CoV-2 intranasal challenge when high viral doses are used. Here, we report on SARS-CoV-2 infection of hamsters engineered to express the hACE2 receptor under the control of the K18 promoter. K18-hACE2 hamsters infected with a relatively low dose of 100 or 1,000 PFU of SARS-CoV-2 developed a severe and lethal disease, with most animals succumbing by day 5 postinfection. Hamsters developed severe lesions and inflammation within the upper and lower respiratory system, including infection of the nasal cavities causing marked destruction of the olfactory epithelium as well as severe bronchopneumonia that extended deep into the alveoli. Additionally, SARS-CoV-2 infection spread to the central nervous system (CNS), including the brain stem and spinal cord. Wild-type (WT) hamsters naturally support SARS-CoV-2 infection, with the primary lesions present in the respiratory tract and nasal cavity. Overall, infection in the K18-hACE2 hamsters is more extensive than that in WT hamsters, with more CNS involvement and a lethal outcome. These findings demonstrate the K18-hACE2 hamster model will be valuable for studying SARS-CoV-2. IMPORTANCE The rapid emergence of SARS-CoV-2 has created a global health emergency. While most human SARS-CoV-2 disease is mild, some people develop severe, life-threatening disease. Small-animal models mimicking the severe aspects of human disease are needed to more clearly understand the pathophysiological processes driving this progression. Here, we studied SARS-CoV-2 infection in hamsters engineered to express the human angiotensin-converting enzyme 2 viral receptor under the control of the K18 promoter. SARS-CoV-2 produces a severe and lethal infection in transgenic hamsters that mirrors the most severe aspects of COVID-19 in humans, including respiratory and neurological injury. In contrast to other animal systems, hamsters manifest disease with levels of input virus more consistent with natural human infection. This system will be useful for the study of SARS-CoV-2 disease and the development of drugs targeting this virus.


Assuntos
COVID-19 , SARS-CoV-2 , Camundongos , Animais , Cricetinae , Humanos , COVID-19/patologia , Enzima de Conversão de Angiotensina 2 , Peptidil Dipeptidase A , Pulmão/patologia , Camundongos Transgênicos , Modelos Animais de Doenças
16.
Virol J ; 8: 441, 2011 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-21933385

RESUMO

BACKGROUND: Previously we demonstrated that DNA vaccination of nonhuman primates (NHP) with a small subset of vaccinia virus (VACV) immunogens (L1, A27, A33, B5) protects against lethal monkeypox virus challenge. The L1 and A27 components of this vaccine target the mature virion (MV) whereas A33 and B5 target the enveloped virion (EV). RESULTS: Here, we demonstrated that the antibodies produced in vaccinated NHPs were sufficient to confer protection in a murine model of lethal Orthopoxvirus infection. We further explored the concept of using DNA vaccine technology to produce immunogen-specific polyclonal antibodies that could then be combined into cocktails as potential immunoprophylactic/therapeutics. Specifically, we used DNA vaccines delivered by muscle electroporation to produce polyclonal antibodies against the L1, A27, A33, and B5 in New Zealand white rabbits. The polyclonal antibodies neutralized both MV and EV in cell culture. The ability of antibody cocktails consisting of anti-MV, anti-EV, or a combination of anti-MV/EV to protect BALB/c mice was evaluated as was the efficacy of the anti-MV/EV mixture in a mouse model of progressive vaccinia. In addition to evaluating weight loss and lethality, bioimaging technology was used to characterize the spread of the VACV infections in mice. We found that the anti-EV cocktail, but not the anti-MV cocktail, limited virus spread and lethality. CONCLUSIONS: A combination of anti-MV/EV antibodies was significantly more protective than anti-EV antibodies alone. These data suggest that DNA vaccine technology could be used to produce a polyclonal antibody cocktail as a possible product to replace vaccinia immune globulin.


Assuntos
Anticorpos Antivirais/administração & dosagem , Antígenos Virais/imunologia , Vacina Antivariólica/genética , Vacinas de DNA/genética , Vaccinia virus/imunologia , Vacínia/prevenção & controle , Proteínas do Envelope Viral/genética , Animais , Anticorpos Heterófilos , Antígenos Virais/química , Antígenos Virais/genética , Peso Corporal , Modelos Animais de Doenças , Eletroporação , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Imagem Molecular , Músculos/imunologia , Testes de Neutralização , Coelhos , Vacina Antivariólica/imunologia , Vacinas de DNA/imunologia , Vacínia/imunologia , Vaccinia virus/efeitos dos fármacos , Vaccinia virus/genética , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/imunologia , Vírion/efeitos dos fármacos , Vírion/imunologia
17.
NPJ Vaccines ; 6(1): 31, 2021 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-33654101

RESUMO

Crimean-Congo hemorrhagic fever virus (CCHFV) is a tick-borne virus that causes severe hemorrhagic fever disease in humans. Currently, no licensed CCHF vaccines exist, and the protective epitopes remain unclear. Previously, we tested a DNA vaccine expressing the M-segment glycoprotein precursor gene of the laboratory CCHFV strain IbAr 10200 (CCHFV-M10200). CCHFV-M10200 provided >60% protection against homologous CCHFV-IbAr 10200 challenge in mice. Here, we report that increasing the dose of CCHFV-M10200 provides complete protection from homologous CCHFV challenge in mice, and significant (80%) protection from challenge with the clinically relevant heterologous strain CCHFV-Afg09-2990. We also report complete protection from CCHFV-Afg09-2990 challenge following vaccination with a CCHFV-Afg09-2990 M-segment DNA vaccine (CCHFV-MAfg09). Finally, we show that the non-structural M-segment protein, GP38, influences CCHF vaccine immunogenicity and provides significant protection from homologous CCHFV challenge. Our results demonstrate that M-segment DNA vaccines elicit protective CCHF immunity and further illustrate the immunorelevance of GP38.

18.
J Thorac Imaging ; 36(2): 108-115, 2021 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-32251234

RESUMO

PURPOSE: In this study, we analyzed the prognostic value of coronary computed tomography angiography-derived morphologic and quantitative plaque markers and plaque scores for major adverse cardiovascular events (MACEs). MATERIALS AND METHODS: We analyzed the data of patients with suspected coronary artery disease (CAD). Various plaque markers were obtained using a semiautomated software prototype or derived from the results of the software analysis. Several risk scores were calculated, and follow-up data concerning MACE were collected from all patients. RESULTS: A total of 131 patients (65±12 y, 73% male) were included in our study. MACE occurred in 11 patients within the follow-up period of 34±25 months.CAD-Reporting and Data System score (odds ratio [OR]=11.62), SYNTAX score (SS) (OR=1.11), Leiden-risk score (OR=1.37), segment involvement score (OR=1.76), total plaque volume (OR=1.20), and percentage aggregated plaque volume (OR=1.32) were significant predictors for MACE (all P≤0.05). Moreover, the difference of the corrected coronary opacification (ΔCCO) correlated significantly with the occurrence of MACE (P<0.0001). The CAD-Reporting and Data System score, SS, and Leiden-risk score showed substantial sensitivity for predicting MACE (90.9%). The SS and Leiden-risk score displayed high specificities of 80.8% and 77.5%, respectively. These plaque markers and risk scores all provided high negative predictive value (>90%). CONCLUSION: The coronary computed tomography angiography-derived plaque markers of segment involvement score, total plaque volume, percentage aggregated plaque volume, and ΔCCO, and the risk scores exhibited predictive value for the occurrence of MACE and can likely aid in identifying patients at risk for future cardiac events.


Assuntos
Doença da Artéria Coronariana , Placa Aterosclerótica , Angiografia por Tomografia Computadorizada , Angiografia Coronária , Doença da Artéria Coronariana/diagnóstico por imagem , Feminino , Humanos , Masculino , Placa Aterosclerótica/diagnóstico por imagem , Valor Preditivo dos Testes , Prognóstico , Medição de Risco , Fatores de Risco , Software
19.
Eur J Radiol ; 138: 109633, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33735700

RESUMO

OBJECTIVE: To evaluate 30 day rate of major adverse cardiac events (MACE) utilizing cCTA and FFRCT for evaluation of patients presenting to the Emergency Department (ED) with acute chest pain. MATERIALS AND METHODS: Patients between the ages of 18-95 years who underwent clinically indicated cCTA and FFRCT in the evaluation of acute chest pain in the emergency department were retrospectively evaluated for 30 day MACE, repeat presentation/admission for chest pain, revascularization, and additional testing. RESULTS: A total of 59 patients underwent CCTA and subsequent FFRCT for the evaluation of acute chest pain in the ED over the enrollment period. 32 out of 59 patients (54 %) had negative FFRCT (>0.80) out of whom 18 patients (55 %) were discharged from the ED. Out of the 32 patients without functionally significant CAD by FFRCT, 32 patients (100 %) underwent no revascularization and 32 patients (100 %) had no MACE at the 30-day follow-up period. CONCLUSION: In this limited retrospective study, patients presenting to the ED with acute chest pain and with CCTA with subsequent FFRCT of >0.8 had no MACE at 30 days; however, for many of these patients results were not available at time of clinical decision making by the ED physician.


Assuntos
Doença da Artéria Coronariana , Reserva Fracionada de Fluxo Miocárdico , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Dor no Peito/diagnóstico por imagem , Angiografia por Tomografia Computadorizada , Angiografia Coronária , Humanos , Pessoa de Meia-Idade , Valor Preditivo dos Testes , Estudos Retrospectivos , Adulto Jovem
20.
Eur J Radiol ; 122: 108744, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31790934

RESUMO

PURPOSE: The study aimed to compare morphological and anatomic plaque markers derived from coronary computed tomography angiography (cCTA) for the detection of lesion specific ischemia with invasive instantaneous wave free ratio (iFR®) as the reference standard. METHODS: In our prospective study, we enrolled patients with suspected coronary artery disease (CAD), who had undergone cCTA, using a low-dose third-generation dual-source CT and invasive coronary angiography (ICA) with iFR® measurement. Various plaque markers were assessed on cCTA. Discriminatory power of these markers for the detection of ischemia-inducing coronary artery disease was evaluated against invasive iFR®. RESULTS: Our study cohort included 39 patients (66.6 ± 12.0 years, 72 % male). Among 54 vessel-specific lesions, 15 lesions (28 %) were characterized as hemodynamically significant by iFR® ≤0.89. The area under the curve (AUC) of lesion length/ minimal luminal diameter4 (LL/MLD4) (0.84) was greater than the AUC of minimal luminal area (MLA) (0.82), MLD (0.81), the degree of luminal diameter stenosis (0.81), corrected coronary opacification (CCO) (0.79), remodeling index (RI) (0.75), and percentage aggregate plaque volume (%APV) (0.72). LL, vessel volume (VV), total plaque volume (TPV), calcified and non-calcified plaque volume (CPV and NCPV) did not reach statistical significance and were unable to discriminate between vessels with and without ischemia-inducing coronary stenosis. CONCLUSION: LL/MLD4, MLA, MLD, the degree of luminal diameter stenosis, CCO, RI, and %APV derived from cCTA can support the detection of hemodynamically significant coronary stenosis as compared with iFR®, with LL/MLD4 showing the greatest discriminatory power.


Assuntos
Estenose Coronária/diagnóstico por imagem , Hemodinâmica/fisiologia , Placa Aterosclerótica/diagnóstico por imagem , Idoso , Área Sob a Curva , Biomarcadores/metabolismo , Angiografia por Tomografia Computadorizada/métodos , Angiografia Coronária/métodos , Doença da Artéria Coronariana/diagnóstico por imagem , Doença da Artéria Coronariana/fisiopatologia , Estenose Coronária/fisiopatologia , Feminino , Reserva Fracionada de Fluxo Miocárdico/fisiologia , Humanos , Masculino , Pessoa de Meia-Idade , Placa Aterosclerótica/fisiopatologia , Estudos Prospectivos , Estudos Retrospectivos , Tomografia Computadorizada por Raios X/métodos
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa