Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Br J Nutr ; 122(12): 1359-1367, 2019 12 28.
Artigo em Inglês | MEDLINE | ID: mdl-31554524

RESUMO

Subcutaneous adipose tissue (scAT) and peripheral blood mononuclear cells (PBMC) play a significant role in obesity-associated systemic low-grade inflammation. High-fat diet (HFD) is known to induce inflammatory changes in both scAT and PBMC. However, the time course of the effect of HFD on these systems is still unknown. The aim of the present study was to determine the time course of the effect of HFD on PBMC and scAT. New Zealand white rabbits were fed HFD for 5 or 10 weeks (i.e. HFD-5 and HFD-10) or regular chow (i.e. control (CNT)-5 and CNT-10). Thereafter, metabolic and inflammatory parameters of PBMC and scAT were quantified. HFD induced hyperfattyacidaemia in HFD-5 and HFD-10 groups, with the development of insulin resistance in HFD-10, while no changes were observed in scAT lipid metabolism and inflammatory status. HFD activated the inflammatory pathways in PBMC of HFD-5 group and induced modified autophagy in that of HFD-10. The rate of fat oxidation in PBMC was directly associated with the expression of inflammatory markers and tended to inversely associate with autophagosome formation markers in PBMC. HFD affected systemic substrate metabolism, and the metabolic, inflammatory and autophagy pathways in PBMC in the absence of metabolic and inflammatory changes in scAT. Dietary approaches or interventions to avert HFD-induced changes in PBMC could be essential to prevent metabolic and inflammatory complications of obesity and promote healthier living.


Assuntos
Dieta Hiperlipídica , Leucócitos Mononucleares/metabolismo , Gordura Subcutânea/metabolismo , Aumento de Peso , Animais , Autofagia , Carnitina/análogos & derivados , Carnitina/metabolismo , Homeostase , Inflamação , Insulina/sangue , Resistência à Insulina , Metabolismo dos Lipídeos , Fígado/metabolismo , Masculino , Obesidade , Coelhos
2.
Proc Natl Acad Sci U S A ; 111(19): 7114-9, 2014 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-24778254

RESUMO

The pathophysiology of hantavirus pulmonary syndrome (HPS) remains unclear because of a lack of surrogate disease models with which to perform pathogenesis studies. Nonhuman primates (NHP) are considered the gold standard model for studying the underlying immune activation/suppression associated with immunopathogenic viruses such as hantaviruses; however, to date an NHP model for HPS has not been described. Here we show that rhesus macaques infected with Sin Nombre virus (SNV), the primary etiological agent of HPS in North America, propagated in deer mice develop HPS, which is characterized by thrombocytopenia, leukocytosis, and rapid onset of respiratory distress caused by severe interstitial pneumonia. Despite establishing a systemic infection, SNV differentially activated host responses exclusively in the pulmonary endothelium, potentially the mechanism leading to acute severe respiratory distress. This study presents a unique chronological characterization of SNV infection and provides mechanistic data into the pathophysiology of HPS in a closely related surrogate animal model. We anticipate this model will advance our understanding of HPS pathogenesis and will greatly facilitate research toward the development of effective therapeutics and vaccines against hantaviral diseases.


Assuntos
Modelos Animais de Doenças , Síndrome Pulmonar por Hantavirus/fisiopatologia , Macaca mulatta/virologia , Doenças dos Macacos/virologia , Peromyscus/virologia , Vírus Sin Nombre/genética , Animais , Chlorocebus aethiops , Síndrome Pulmonar por Hantavirus/diagnóstico por imagem , Síndrome Pulmonar por Hantavirus/transmissão , Pulmão/diagnóstico por imagem , Pulmão/virologia , Dados de Sequência Molecular , Doenças dos Macacos/fisiopatologia , Doenças dos Macacos/transmissão , América do Norte , RNA Viral/genética , Radiografia , Células Vero , Viremia/fisiopatologia
3.
Proc Natl Acad Sci U S A ; 110(5): 1893-8, 2013 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-23319647

RESUMO

Ebola viruses cause hemorrhagic disease in humans and nonhuman primates with high fatality rates. These viruses pose a significant health concern worldwide due to the lack of approved therapeutics and vaccines as well as their potential misuse as bioterrorism agents. Although not licensed for human use, recombinant vesicular stomatitis virus (rVSV) expressing the filovirus glycoprotein (GP) has been shown to protect macaques from Ebola virus and Marburg virus infections, both prophylactically and postexposure in a homologous challenge setting. However, the immune mechanisms of protection conferred by this vaccine platform remain poorly understood. In this study, we set out to investigate the role of humoral versus cellular immunity in rVSV vaccine-mediated protection against lethal Zaire ebolavirus (ZEBOV) challenge. Groups of cynomolgus macaques were depleted of CD4+ T, CD8+ T, or CD20+ B cells before and during vaccination with rVSV/ZEBOV-GP. Unfortunately, CD20-depleted animals generated a robust IgG response. Therefore, an additional group of vaccinated animals were depleted of CD4+ T cells during challenge. All animals were subsequently challenged with a lethal dose of ZEBOV. Animals depleted of CD8+ T cells survived, suggesting a minimal role for CD8+ T cells in vaccine-mediated protection. Depletion of CD4+ T cells during vaccination caused a complete loss of glycoprotein-specific antibodies and abrogated vaccine protection. In contrast, depletion of CD4+ T cells during challenge resulted in survival of the animals, indicating a minimal role for CD4+ T-cell immunity in rVSV-mediated protection. Our results suggest that antibodies play a critical role in rVSV-mediated protection against ZEBOV.


Assuntos
Anticorpos Antivirais/imunologia , Vacinas contra Ebola/imunologia , Ebolavirus/imunologia , Doença pelo Vírus Ebola/imunologia , Glicoproteínas de Membrana/imunologia , Proteínas do Envelope Viral/imunologia , Animais , Anticorpos Antivirais/sangue , Citocinas/sangue , Citocinas/imunologia , Vacinas contra Ebola/administração & dosagem , Ebolavirus/genética , Ensaio de Imunoadsorção Enzimática , Doença pelo Vírus Ebola/sangue , Doença pelo Vírus Ebola/prevenção & controle , Humanos , Imunoglobulina G/sangue , Imunoglobulina G/imunologia , Fígado/imunologia , Fígado/parasitologia , Fígado/patologia , Linfócitos/imunologia , Macaca fascicularis , Masculino , Marburgvirus/genética , Marburgvirus/imunologia , Glicoproteínas de Membrana/genética , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Baço/imunologia , Baço/parasitologia , Baço/patologia , Fatores de Tempo , Vírus da Estomatite Vesicular Indiana/genética , Vírus da Estomatite Vesicular Indiana/imunologia , Proteínas do Envelope Viral/genética , Carga Viral/genética
4.
Proc Natl Acad Sci U S A ; 110(41): 16598-603, 2013 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-24062443

RESUMO

In 2012, a novel betacoronavirus, designated Middle East respiratory syndrome coronavirus or MERS-CoV and associated with severe respiratory disease in humans, emerged in the Arabian Peninsula. To date, 108 human cases have been reported, including cases of human-to-human transmission. The availability of an animal disease model is essential for understanding pathogenesis and developing effective countermeasures. Upon a combination of intratracheal, ocular, oral, and intranasal inoculation with 7 × 10(6) 50% tissue culture infectious dose of the MERS-CoV isolate HCoV-EMC/2012, rhesus macaques developed a transient lower respiratory tract infection. Clinical signs, virus shedding, virus replication in respiratory tissues, gene expression, and cytokine and chemokine profiles peaked early in infection and decreased over time. MERS-CoV caused a multifocal, mild to marked interstitial pneumonia, with virus replication occurring mainly in alveolar pneumocytes. This tropism of MERS-CoV for the lower respiratory tract may explain the severity of the disease observed in humans and the, up to now, limited human-to-human transmission.


Assuntos
Doenças Transmissíveis Emergentes/virologia , Modelos Animais de Doenças , Pulmão/patologia , Macaca mulatta , Síndrome Respiratória Aguda Grave/patologia , Síndrome Respiratória Aguda Grave/virologia , Animais , Pulmão/virologia , Microscopia Eletrônica de Transmissão , Especificidade da Espécie , Vírion/ultraestrutura , Replicação Viral/fisiologia , Eliminação de Partículas Virais/fisiologia
5.
J Infect Dis ; 207(8): 1316-27, 2013 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-23303805

RESUMO

The virulence of Soromba-R, a Lassa virus strain recently isolated from southern Mali, was assessed in 2 animal models of Lassa fever: inbred strain 13 guinea pigs and cynomolgus macaques. In both models, the Malian isolate demonstrated tissue tropism and viral titers similar to those of historical Lassa virus isolates from Sierra Leone (Josiah) and Liberia (Z-132); however, the Soromba-R isolate was found to be less pathogenic, as determined by decreased mortality and prolonged time to euthanasia in macaques. Interestingly, in addition to the classic indicators of Lassa fever, Soromba-R infection presented with moderate to severe pulmonary manifestations in the macaque model. Analysis of host responses demonstrated increased immune activation in Soromba-R-infected macaques, particularly in neutrophil-activating or -potentiating proinflammatory cytokines or growth factors, including tumor necrosis factor α, macrophage inflammatory protein 1α, interleukin 1ß, and granulocyte colony-stimulating factor, as well as interleukin 5, which may be responsible for the decreased lethality and uncharacteristic clinical presentation. These results suggest that the strain of Lassa virus circulating in Mali might be less pathogenic than strains circulating in the historical region of endemicity and may result in an atypical presentation for Lassa fever, which could complicate clinical diagnosis.


Assuntos
Febre Lassa/patologia , Vírus Lassa/patogenicidade , Animais , Quimiocina CCL3/sangue , Quimiocina CCL3/imunologia , Chlorocebus aethiops , Modelos Animais de Doenças , Progressão da Doença , Feminino , Fator Estimulador de Colônias de Granulócitos/sangue , Fator Estimulador de Colônias de Granulócitos/imunologia , Cobaias , Testes Hematológicos , Imuno-Histoquímica , Interleucina-1beta/sangue , Interleucina-1beta/imunologia , Interleucina-6/sangue , Interleucina-6/imunologia , Febre Lassa/imunologia , Febre Lassa/virologia , Vírus Lassa/genética , Vírus Lassa/imunologia , Vírus Lassa/isolamento & purificação , Pulmão/patologia , Pulmão/virologia , Macaca fascicularis , Masculino , Mali , RNA Viral/sangue , Fatores de Tempo , Fator de Necrose Tumoral alfa/sangue , Fator de Necrose Tumoral alfa/imunologia , Células Vero , Carga Viral , Viremia/virologia , Virulência
6.
Nutrients ; 16(3)2024 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-38337648

RESUMO

Exclusive enteral nutrition (EEN) is an established dietary treatment for Crohn's disease (CD) by alleviating inflammation and inducing remission. However, the mechanisms of action of EEN are incompletely understood. As CD is associated with gut microbiome dysbiosis, we investigated the effect of EEN on the microbiome in a rat model of CD-like colitis. The rat model of CD-like colitis was established by an intracolonic instillation of TNBS at 65 mg/kg in 250 µL of 40% ethanol. Sham control rats were instilled with saline. Rats were fed ad libitum with either regular pellet food or EEN treatment with a clear liquid diet (Ensure). Rats were euthanized at 7 days. Fecal pellets were collected from the distal colon for 16S rRNA sequencing analysis of gut microbiota. In addition, colon tissues were taken for histological and molecular analyses in all the groups of rats. EEN administration to TNBS-induced CD rats significantly improved the body weight change, inflammation scores, and disease activity index. The mRNA expression of IL-17A and interferon-γ was significantly increased in the colonic tissue in TNBS rats when fed with regular food. However, EEN treatment significantly attenuated the increase in IL-17A and interferon-γ in TNBS rats. Our 16S rRNA sequencing analysis found that gut microbiota diversity and compositions were significantly altered in TNBS rats, compared to controls. However, EEN treatment improved alpha diversity and increased certain beneficial bacteria such as Lactobacillus and Dubosiella and decreased bacteria such as Bacteroides and Enterorhabdus in CD-like rats, compared to CD-like rats with the regular pellet diet. In conclusion, EEN treatment increases the diversity of gut microbiota and the composition of certain beneficial bacteria. These effects may contribute to the reduced inflammation by EEN in the rat model of CD-like colitis.


Assuntos
Colite , Doença de Crohn , Microbioma Gastrointestinal , Ratos , Animais , Doença de Crohn/microbiologia , Nutrição Enteral , RNA Ribossômico 16S/genética , Interleucina-17 , Interferon gama , Colite/induzido quimicamente , Colite/terapia , Bactérias , Inflamação/terapia , Indução de Remissão
7.
PLoS Pathog ; 7(12): e1002426, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22194683

RESUMO

Hantavirus pulmonary syndrome (HPS), also referred to as hantavirus cardiopulmonary syndrome (HCPS), is a rare but frequently fatal disease caused by New World hantaviruses. In humans HPS is associated with severe pulmonary edema and cardiogenic shock; however, the pathogenesis of this disease remains unclear largely due to a lack of suitable animal models for the study of disease progression. In this study we monitored clinical, virological, pathophysiological parameters and host immunological responses to decipher pathological factors and events in the lethal Syrian hamster model of HPS following intranasal inoculation of Andes virus. Transcriptional profiling of the host gene responses demonstrated a suppression of innate immune responses in most organs analyzed during the early stage of infection, except for in the lung which had low level activation of several pro-inflammatory genes. During this phase Andes virus established a systemic infection in hamsters, with viral antigen readily detectable in the endothelium of the majority of tissues analyzed by 7-8 days post-inoculation. Despite wide-spread infection, histological analysis confirmed pathological abnormalities were almost exclusively found in the lungs. Immediately preceding clinical signs of disease, intense activation of pro-inflammatory and Th1/Th2 responses were observed in the lungs as well as the heart, but not in peripheral organs, suggesting that localized immune-modulations by infection is paramount to pathogenesis. Throughout the course of infection a strong suppression of regulatory T-cell responses was noted and is hypothesized to be the basis of the aberrant immune activations. The unique and comprehensive monitoring of host immune responses to hantavirus infection increases our understanding of the immuno-pathogenesis of HPS and will facilitate the development of treatment strategies targeting deleterious host immunological responses.


Assuntos
Síndrome Pulmonar por Hantavirus/imunologia , Síndrome Pulmonar por Hantavirus/virologia , Administração Intranasal , Animais , Cricetinae , Modelos Animais de Doenças , Feminino , Orthohantavírus/isolamento & purificação , Orthohantavírus/patogenicidade , Síndrome Pulmonar por Hantavirus/patologia , Interações Hospedeiro-Patógeno , Pulmão/patologia , Pulmão/virologia , Mesocricetus , Linfócitos T Reguladores/imunologia
8.
J Virol ; 85(15): 7658-71, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21593160

RESUMO

Nipah virus (NiV) and Hendra virus (HeV) are emerging zoonotic viruses and the causative agents of severe respiratory disease and encephalitis in humans. Little is known about the mechanisms that govern the development of respiratory and neurological disease. Using a hamster model of lethal NiV and HeV infection, we describe the role of the route and dose of infection on the clinical outcome and determine virus tropism and host responses following infection. Infection of hamster with a high dose of NiV or HeV resulted in acute respiratory distress. NiV initially replicated in the upper respiratory tract epithelium, whereas HeV initiated infection primarily in the interstitium. In contrast, infection with a low dose of NiV or HeV resulted in the development of neurological signs and more systemic spread of the virus through involvement of the endothelium. The development of neurological signs coincided with disruption of the blood-brain barrier (BBB) and expression of tumor necrosis alpha (TNF-α) and interleukin 1 ß (IL-1ß). In addition, interferon-inducible protein 10 (IP-10) was identified as playing an important role in NiV and HeV pathogenesis. These studies reveal novel information on the development and progression of NiV and HeV clinical disease, provide a mechanism for the differences in transmission observed between NiV and HeV outbreaks, and identify specific cytokines and chemokines that serve as important targets for treatment.


Assuntos
Infecções por Henipavirus/patologia , Henipavirus/fisiologia , Animais , Barreira Hematoencefálica , Cricetinae , Modelos Animais de Doenças , Feminino , Infecções por Henipavirus/virologia , Imuno-Histoquímica , Mesocricetus , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Resultado do Tratamento , Tropismo Viral , Replicação Viral
9.
J Virol ; 85(3): 1214-23, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21084481

RESUMO

The first influenza pandemic of the new millennium was caused by a newly emerged swine-origin influenza virus (SOIV) (H1N1). This new virus is characterized by a previously unknown constellation of gene segments derived from North American and Eurasian swine lineages and the absence of common markers predictive of human adaptation. Overall, human infections appeared to be mild, but an alarming number of young individuals presented with symptoms atypical for seasonal influenza. The new SOIV also showed a sustained human-to-human transmissibility and higher reproduction ratio than common seasonal viruses, altogether indicating a higher pathogenic potential for this newly emerged virus. To study the virulence of the SOIV, we used a recently established cynomolgus macaque model and compared parameters of clinical disease, virology, host responses, and pathology/histopathology with a current seasonal H1N1 virus. We here show that infection of macaques with two genetically similar but clinically distinct SOIV isolates from the early stage of the pandemic (A/Mexico/4108/2009 and A/Mexico/InDRE4487/2009) resulted in upper and lower respiratory tract infections and clinical disease ranging from mild to severe pneumonia that was clearly advanced over the mild infection caused by A/Kawasaki/UTK-4/2009, a current seasonal strain. Unexpectedly, we observed heterogeneity among the two SOIV isolates in virus replication, host transcriptional and cytokine responses, and disease progression, demonstrating a higher pathogenic potential for A/Mexico/InDRE4487/2009. Differences in virulence may explain more severe disease, as was seen with certain individuals infected with the emerged pandemic influenza virus. Thus, the nonhuman primate model closely mimics influenza in humans.


Assuntos
Vírus da Influenza A Subtipo H1N1/isolamento & purificação , Vírus da Influenza A Subtipo H1N1/patogenicidade , Infecções por Orthomyxoviridae/patologia , Infecções por Orthomyxoviridae/virologia , Doenças dos Primatas/patologia , Doenças dos Primatas/virologia , Animais , Pré-Escolar , Citocinas/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Perfilação da Expressão Gênica , Variação Genética , Humanos , Influenza Humana/virologia , Macaca , Masculino , Pneumonia Viral/patologia , Pneumonia Viral/virologia , Infecções Respiratórias/patologia , Infecções Respiratórias/virologia , Índice de Gravidade de Doença , Virulência
10.
J Infect Dis ; 204 Suppl 3: S991-9, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21987781

RESUMO

To gain further insight into the interdependent pathogenic processes in Ebola hemorrhagic fever (EHF), we have examined the dynamics of host responses in individual rhesus macaques infected with Zaire ebolavirus over the entire disease course. Examination of coagulation parameters revealed that decreased coagulation inhibitor activity triggered severe coagulopathy as indicated by prolonged coagulation times and decreased fibrinogen levels. This has been proposed as one of the significant mechanisms underlying disseminated intravascular coagulation in EHF patients. Furthermore, monitoring of expression levels for cytokines/chemokines suggested a mixed anti-inflammatory response syndrome (MARS), which indicates that a catastrophic uncontrolled immunological status contributes to the development of fatal hemorrhagic fever. These results highlight the pathological analogies between EHF and severe sepsis and not only contribute to our understanding of the pathogenic process, but will also help to establish novel postexposure treatment modalities.


Assuntos
Ebolavirus/patogenicidade , Doença pelo Vírus Ebola/imunologia , Animais , Coagulação Sanguínea , Quimiocinas/metabolismo , Chlorocebus aethiops , Citocinas/metabolismo , Hemorragia , Doença pelo Vírus Ebola/sangue , Doença pelo Vírus Ebola/patologia , Interações Hospedeiro-Patógeno , Inflamação , Macaca mulatta , Masculino , Fatores de Tempo , Células Vero , Viremia , Tempo de Coagulação do Sangue Total
11.
J Virol ; 84(19): 9831-9, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20660198

RESUMO

The henipaviruses, Hendra virus (HeV) and Nipah virus (NiV), are emerging zoonotic paramyxoviruses that can cause severe and often lethal neurologic and/or respiratory disease in a wide variety of mammalian hosts, including humans. There are presently no licensed vaccines or treatment options approved for human or veterinarian use. Guinea pigs, hamsters, cats, and ferrets, have been evaluated as animal models of human HeV infection, but studies in nonhuman primates (NHP) have not been reported, and the development and approval of any vaccine or antiviral for human use will likely require efficacy studies in an NHP model. Here, we examined the pathogenesis of HeV in the African green monkey (AGM) following intratracheal inoculation. Exposure of AGMs to HeV produced a uniformly lethal infection, and the observed clinical signs and pathology were highly consistent with HeV-mediated disease seen in humans. Ribavirin has been used to treat patients infected with either HeV or NiV; however, its utility in improving outcome remains, at best, uncertain. We examined the antiviral effect of ribavirin in a cohort of nine AGMs before or after exposure to HeV. Ribavirin treatment delayed disease onset by 1 to 2 days, with no significant benefit for disease progression and outcome. Together our findings introduce a new disease model of acute HeV infection suitable for testing antiviral strategies and also demonstrate that, while ribavirin may have some antiviral activity against the henipaviruses, its use as an effective standalone therapy for HeV infection is questionable.


Assuntos
Antivirais/farmacologia , Vírus Hendra , Infecções por Henipavirus/tratamento farmacológico , Infecções por Henipavirus/etiologia , Ribavirina/farmacologia , Animais , Sequência de Bases , Encéfalo/patologia , Chlorocebus aethiops , Primers do DNA/genética , DNA Viral/genética , Modelos Animais de Doenças , Feminino , Vírus Hendra/genética , Vírus Hendra/patogenicidade , Vírus Hendra/fisiologia , Infecções por Henipavirus/patologia , Infecções por Henipavirus/virologia , Humanos , Pulmão/diagnóstico por imagem , Pulmão/patologia , Masculino , Radiografia , Replicação Viral
12.
Sci Transl Med ; 13(602)2021 07 14.
Artigo em Inglês | MEDLINE | ID: mdl-34261800

RESUMO

Although substantial progress has been made with Ebola virus (EBOV) vaccine measures, the immune correlates of vaccine-mediated protection remain uncertain. Here, five mucosal vaccine vectors based on human and avian paramyxoviruses provided nonhuman primates with varying degrees of protection, despite expressing the same EBOV glycoprotein (GP) immunogen. Each vaccine produced antibody responses that differed in Fc-mediated functions and isotype composition, as well as in magnitude and coverage toward GP and its conformational and linear epitopes. Differences in the degree of protection and comprehensive characterization of the response afforded the opportunity to identify which features and functions were elevated in survivors and could therefore serve as vaccine correlates of protection. Pairwise network correlation analysis of 139 immune- and vaccine-related parameters was performed to demonstrate relationships with survival. Total GP-specific antibodies, as measured by biolayer interferometry, but not neutralizing IgG or IgA titers, correlated with survival. Fc-mediated functions and the amount of receptor binding domain antibodies were associated with improved survival outcomes, alluding to the protective mechanisms of these vaccines. Therefore, functional qualities of the antibody response, particularly Fc-mediated effects and GP specificity, rather than simply magnitude of the response, appear central to vaccine-induced protection against EBOV. The heterogeneity of the response profile between the vaccines indicates that each vaccine likely exhibits its own protective signature and the requirements for an efficacious EBOV vaccine are complex.


Assuntos
Vacinas contra Ebola , Ebolavirus , Doença pelo Vírus Ebola , Animais , Anticorpos Neutralizantes , Anticorpos Antivirais , Especificidade de Anticorpos , Doença pelo Vírus Ebola/prevenção & controle , Humanos , Primatas
13.
Comp Med ; 59(3): 257-65, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19619416

RESUMO

Bacillus anthracis, the causative agent of anthrax, is a category A priority pathogen that causes extensive damage in humans. For this reason, B. anthracis has been the focus of numerous studies using various animal models. In this study, we explored physiologic parameters in Dutch belted rabbits with inhalation anthrax to characterize the disease progression in this model. To this end, we infected Dutch belted rabbits with 100 LD(50) B. anthracis Ames spores by nasal instillation and continuously recorded various physiologic parameters by using telemetry. In addition, samples were collected at selected times for serum chemistry, hematology, and blood gas analysis. The animals exhibited hemodynamic and respiratory changes that coincided with those reported in human cases of inhalational anthrax infection, including hypotension, altered heart rate, and respiratory distress. Likewise, hematology, serum chemistry, and blood gas analysis revealed trends comparable to human anthrax-related pathophysiology. The Dutch belted rabbit model of inhalational anthrax exhibited most of the physiologic, hematologic, and biochemical sequelae noted in human cases. Therefore, this rabbit model fulfills several of the criteria of a useful animal model for studying disease pathogenesis and evaluating therapeutics during inhalational anthrax.


Assuntos
Antraz/fisiopatologia , Bacillus anthracis/fisiologia , Síndrome do Desconforto Respiratório/fisiopatologia , Insuficiência Respiratória/fisiopatologia , Animais , Antraz/microbiologia , Antraz/transmissão , Bacillus anthracis/patogenicidade , Gasometria , Modelos Animais de Doenças , Feminino , Testes Hematológicos , Hemodinâmica , Exposição por Inalação , Coelhos , Síndrome do Desconforto Respiratório/microbiologia , Insuficiência Respiratória/microbiologia
14.
Comp Med ; 67(3): 232-241, 2017 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-28662752

RESUMO

Arthropod-borne viruses (arboviruses) have continued to emerge in recent years, posing a significant health threat to millions of people worldwide. The majority of arboviruses that are pathogenic to humans are transmitted by mosquitoes and ticks, but other types of arthropod vectors can also be involved in the transmission of these viruses. To alleviate the health burdens associated with arbovirus infections, it is necessary to focus today's research on disease control and therapeutic strategies. Animal models for arboviruses are valuable experimental tools that can shed light on the pathophysiology of infection and will enable the evaluation of future treatments and vaccine candidates. Ideally an animal model will closely mimic the disease manifestations observed in humans. In this review, we outline the currently available animal models for several viruses vectored by mosquitoes, ticks, and midges, for which there are no standardly available vaccines or therapeutics.


Assuntos
Arbovírus , Modelos Animais de Doenças , Animais , Projetos de Pesquisa
15.
Virulence ; 4(8): 707-15, 2013 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-24104465

RESUMO

Staphylococcus aureus community-acquired pneumonia is often associated with influenza or an influenza-like syndrome. Morbidity and mortality due to methicillin-resistant S. aureus (MRSA) or influenza and pneumonia, which includes bacterial co-infection, are among the top causes of death by infectious diseases in the United States. We developed a non-lethal influenza A virus (IAV) (H3N2)/S. aureus co-infection model in cynomolgus macaques (Macaca fascicularis) to test the hypothesis that seasonal IAV infection predisposes non-human primates to severe S. aureus pneumonia. Infection and disease progression were monitored by clinical assessment of animal health; analysis of blood chemistry, nasal swabs, and X-rays; and gross pathology and histopathology of lungs from infected animals. Seasonal IAV infection in healthy cynomolgus macaques caused mild pneumonia, but unexpectedly, did not predispose these animals to subsequent severe infection with the community-associated MRSA clone USA300. We conclude that in our co-infection model, seasonal IAV infection alone is not sufficient to promote severe S. aureus pneumonia in otherwise healthy non-human primates. The implication of these findings is that comorbidity factors in addition to IAV infection are required to predispose individuals to secondary S. aureus pneumonia.


Assuntos
Coinfecção/microbiologia , Coinfecção/virologia , Vírus da Influenza A Subtipo H3N2/crescimento & desenvolvimento , Interações Microbianas , Infecções por Orthomyxoviridae/complicações , Pneumonia Estafilocócica/complicações , Staphylococcus aureus/crescimento & desenvolvimento , Animais , Coinfecção/patologia , Modelos Animais de Doenças , Feminino , Humanos , Pulmão/patologia , Macaca fascicularis , Masculino , Infecções por Orthomyxoviridae/patologia , Infecções por Orthomyxoviridae/virologia , Pneumonia Estafilocócica/microbiologia , Pneumonia Estafilocócica/patologia
16.
PLoS One ; 7(7): e39990, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22808082

RESUMO

The triple reassortant H2N3 virus isolated from diseased pigs in the United States in 2006 is pathogenic for certain mammals without prior adaptation and transmits among swine and ferrets. Adaptation, in the H2 hemagglutinin derived from an avian virus, includes the ability to bind to the mammalian receptor, a significant prerequisite for infection of mammals, in particular humans, which poses a big concern for public health. Here we investigated the pathogenic potential of swine H2N3 in Cynomolgus macaques, a surrogate model for human influenza infection. In contrast to human H2N2 virus, which served as a control and largely caused mild pneumonia similar to seasonal influenza A viruses, the swine H2N3 virus was more pathogenic causing severe pneumonia in nonhuman primates. Both viruses replicated in the entire respiratory tract, but only swine H2N3 could be isolated from lung tissue on day 6 post infection. All animals cleared the infection whereas swine H2N3 infected macaques still presented with pathologic changes indicative of chronic pneumonia at day 14 post infection. Swine H2N3 virus was also detected to significantly higher titers in nasal and oral swabs indicating the potential for animal-to-animal transmission. Plasma levels of IL-6, IL-8, MCP-1 and IFNγ were significantly increased in swine H2N3 compared to human H2N2 infected animals supporting the previously published notion of increased IL-6 levels being a potential marker for severe influenza infections. In conclusion, the swine H2N3 virus represents a threat to humans with the potential for causing a larger outbreak in a non-immune or partially immune population. Furthermore, surveillance efforts in farmed pig populations need to become an integral part of any epidemic and pandemic influenza preparedness.


Assuntos
Vírus da Influenza A/patogenicidade , Macaca fascicularis/virologia , Infecções por Orthomyxoviridae/veterinária , Pneumonia Viral/veterinária , Vírus Reordenados/patogenicidade , Doenças dos Suínos/transmissão , Suínos/virologia , Animais , Quimiocina CCL2/biossíntese , Quimiocina CCL2/imunologia , Feminino , Humanos , Vírus da Influenza A Subtipo H2N2/imunologia , Vírus da Influenza A Subtipo H2N2/patogenicidade , Vírus da Influenza A/imunologia , Interferon gama/biossíntese , Interferon gama/imunologia , Interleucina-6/biossíntese , Interleucina-6/imunologia , Interleucina-8/biossíntese , Interleucina-8/imunologia , Pulmão/imunologia , Pulmão/patologia , Pulmão/virologia , Macaca fascicularis/imunologia , Masculino , Infecções por Orthomyxoviridae/complicações , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/transmissão , Pneumonia Viral/etiologia , Pneumonia Viral/imunologia , Pneumonia Viral/patologia , Vírus Reordenados/imunologia , Índice de Gravidade de Doença , Suínos/imunologia , Doenças dos Suínos/imunologia , Doenças dos Suínos/virologia
17.
PLoS One ; 7(4): e36192, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22558378

RESUMO

Ebola virus (EBOV) is the causative agent of severe hemorrhagic fever in primates, with human case fatality rates up to 90%. Today, there is neither a licensed vaccine nor a treatment available for Ebola hemorrhagic fever (EHF). Single monoclonal antibodies (MAbs) specific for Zaire ebolavirus (ZEBOV) have been successfully used in passive immunization experiments in rodent models, but have failed to protect nonhuman primates from lethal disease. In this study, we used two clones of human-mouse chimeric MAbs (ch133 and ch226) with strong neutralizing activity against ZEBOV and evaluated their protective potential in a rhesus macaque model of EHF. Reduced viral loads and partial protection were observed in animals given MAbs ch133 and ch226 combined intravenously at 24 hours before and 24 and 72 hours after challenge. MAbs circulated in the blood of a surviving animal until virus-induced IgG responses were detected. In contrast, serum MAb concentrations decreased to undetectable levels at terminal stages of disease in animals that succumbed to infection, indicating substantial consumption of these antibodies due to virus replication. Accordingly, the rapid decrease of serum MAbs was clearly associated with increased viremia in non-survivors. Our results indicate that EBOV neutralizing antibodies, particularly in combination with other therapeutic strategies, might be beneficial in reducing viral loads and prolonging disease progression during EHF.


Assuntos
Anticorpos Monoclonais/imunologia , Anticorpos Neutralizantes/imunologia , Doença pelo Vírus Ebola/prevenção & controle , Animais , Anticorpos Monoclonais/química , Anticorpos Neutralizantes/química , Especificidade de Anticorpos , Células CHO , Chlorocebus aethiops , Cricetinae , Cricetulus , Modelos Animais de Doenças , Ebolavirus/imunologia , Ebolavirus/patogenicidade , Ebolavirus/fisiologia , Epitopos/imunologia , Humanos , Imunização Passiva , Macaca mulatta , Masculino , Camundongos , Modelos Moleculares , Conformação Proteica , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/imunologia , Células Vero , Carga Viral/imunologia
18.
PLoS One ; 6(4): e18558, 2011 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-21533129

RESUMO

The severe acute respiratory syndrome (SARS) epidemic was characterized by increased pathogenicity in the elderly due to an early exacerbated innate host response. SARS-CoV is a zoonotic pathogen that entered the human population through an intermediate host like the palm civet. To prevent future introductions of zoonotic SARS-CoV strains and subsequent transmission into the human population, heterologous disease models are needed to test the efficacy of vaccines and therapeutics against both late human and zoonotic isolates. Here we show that both human and zoonotic SARS-CoV strains can infect cynomolgus macaques and resulted in radiological as well as histopathological changes similar to those seen in mild human cases. Viral replication was higher in animals infected with a late human phase isolate compared to a zoonotic isolate. While there were significant differences in the number of host genes differentially regulated during the host responses between the three SARS-CoV strains, the top pathways and functions were similar and only apparent early during infection with the majority of genes associated with interferon signaling pathways. This study characterizes critical disease models in the evaluation and licensure of therapeutic strategies against SARS-CoV for human use.


Assuntos
Síndrome Respiratória Aguda Grave/patologia , Zoonoses , Animais , Citocinas/biossíntese , Humanos , Imuno-Histoquímica , Macaca fascicularis , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/fisiologia , Síndrome Respiratória Aguda Grave/transmissão , Replicação Viral
19.
Comp Med ; 60(5): 389-95, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21262125

RESUMO

Recent advances in the technology associated with digital radiography have created new opportunities for biomedical research applications. Here we evaluated the use of thoracic radiography as a noninvasive refinement methodology for the cynomologus macaque (Macaca fascicularis) model of H1N1 infection. Thoracic radiographic evaluations of macaques infected with any of 3 strains of emerging H1N1 swine-associated influenza virus isolated during the recent pandemic were compared with those of macaques infected with the currently circulating Kawasaki strain of H1N1 influenza. Ventrodorsal, right, and left lateral thoracic radiographs were obtained at days 0, 1, 6, 8, 11, and 14 after infection. A board-certified veterinary radiologist who was blinded to the study design evaluated the images. Numeric scores of extent and severity of lung involvement assigned to each radiograph were compared and demonstrated a significant and substantial difference among groups. The radiographic evaluation allowed for noninvasive assessment of lung involvement, disease onset, progression, and resolution of radiographic changes associated with H1N1 influenza infection.


Assuntos
Vírus da Influenza A Subtipo H1N1 , Pulmão/diagnóstico por imagem , Infecções por Orthomyxoviridae/diagnóstico por imagem , Animais , Progressão da Doença , Feminino , Pulmão/virologia , Macaca fascicularis , Masculino , Infecções por Orthomyxoviridae/virologia , Radiografia
20.
Vaccine ; 27(11): 1729-34, 2009 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-19186197

RESUMO

Eastern equine encephalitis virus (EEEV) is an arthropod-borne virus associated with life-threatening encephalitis in humans, equines, birds and many other domestic animals. To investigate the suitability of the Aotus nancymaae New World owl monkey as a viable animal model for EEE candidate vaccine testing we used clinical presentation, serology, viral isolation and PCR to evaluate pathogenesis and immunity in infected animals. Monkeys were inoculated subcutaneously (SQ) or intranasally (IN) with 10(4)pfu of virulent EEEV and were initially followed for 45 days. While none of the animals displayed clinical signs of disease, all of the SC inoculated animals (n=6) manifested a viremia averaging 3.2 days (+/-0.8 days). Likewise, serologic responses (IgM, IgG and PRNT) were observed in all SC infected animals. Interestingly, none of the IN inoculated animals (n=6) became viremic or mounted an antibody response and no pathological abnormalities were observed in two animals that were necropsied on day 6 post-infection (p.i.) from each group. To determine if the antibodies produced by the SC inoculated animals were protective against homologous challenge, three animals from the SC group were serologically evaluated on day 253 p.i. and were administered an inoculum identical to initial challenge on day 270 p.i. A positive control group of four naïve animals was also infected as before. All of the naïve positive control animals manifested a similar viremia as observed initially, averaging 2.75 days (+/-0.5 days) while none of the previously challenged animals became viremic. On days 45 and 253 p.i. geometric mean PRNT titers in the SC group were 453 and 101, respectively. This study demonstrates that the Aotus nancymaae can be reproducibly infected with EEE virus and can serve as a suitable model for infection and immunogenicity for the evaluation of candidate vaccines against EEEV.


Assuntos
Aotidae/imunologia , Aotidae/virologia , Vírus da Encefalite Equina do Leste/imunologia , Vírus da Encefalite Equina do Leste/patogenicidade , Encefalomielite Equina do Leste/imunologia , Encefalomielite Equina do Leste/virologia , Administração Intranasal , Animais , Anticorpos Antivirais/análise , Anticorpos Antivirais/biossíntese , Modelos Animais de Doenças , Vírus da Encefalite Equina do Leste/isolamento & purificação , Ensaio de Imunoadsorção Enzimática , Cavalos , Imunoglobulina G/análise , Imunoglobulina G/biossíntese , Imunoglobulina M/análise , Imunoglobulina M/biossíntese , Injeções Subcutâneas , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ensaio de Placa Viral , Viremia/virologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA