Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Cell Rep Med ; 2(4): 100230, 2021 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-33754147

RESUMEN

The deployment of a vaccine that limits transmission and disease likely will be required to end the coronavirus disease 2019 (COVID-19) pandemic. We recently described the protective activity of an intranasally administered chimpanzee adenovirus-vectored vaccine encoding a pre-fusion stabilized spike (S) protein (ChAd-SARS-CoV-2-S [chimpanzee adenovirus-severe acute respiratory syndrome-coronavirus-2-S]) in the upper and lower respiratory tracts of mice expressing the human angiotensin-converting enzyme 2 (ACE2) receptor. Here, we show the immunogenicity and protective efficacy of this vaccine in non-human primates. Rhesus macaques were immunized with ChAd-Control or ChAd-SARS-CoV-2-S and challenged 1 month later by combined intranasal and intrabronchial routes with SARS-CoV-2. A single intranasal dose of ChAd-SARS-CoV-2-S induces neutralizing antibodies and T cell responses and limits or prevents infection in the upper and lower respiratory tracts after SARS-CoV-2 challenge. As ChAd-SARS-CoV-2-S confers protection in non-human primates, it is a promising candidate for limiting SARS-CoV-2 infection and transmission in humans.

2.
bioRxiv ; 2021 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-33532770

RESUMEN

The deployment of a vaccine that limits transmission and disease likely will be required to end the Coronavirus Disease 2019 (COVID-19) pandemic. We recently described the protective activity of an intranasally-administered chimpanzee adenovirus-vectored vaccine encoding a pre-fusion stabilized spike (S) protein (ChAd-SARS-CoV-2-S) in the upper and lower respiratory tract of mice expressing the human angiotensin-converting enzyme 2 (ACE2) receptor. Here, we show the immunogenicity and protective efficacy of this vaccine in non-human primates. Rhesus macaques were immunized with ChAd-Control or ChAd-SARS-CoV-2-S and challenged one month later by combined intranasal and intrabronchial routes with SARS-CoV-2. A single intranasal dose of ChAd-SARS-CoV-2-S induced neutralizing antibodies and T cell responses and limited or prevented infection in the upper and lower respiratory tract after SARS-CoV-2 challenge. As this single intranasal dose vaccine confers protection against SARS-CoV-2 in non-human primates, it is a promising candidate for limiting SARS-CoV-2 infection and transmission in humans.

3.
Nature ; 585(7824): 273-276, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32516797

RESUMEN

Effective therapies to treat coronavirus disease 2019 (COVID-19) are urgently needed. While many investigational, approved, and repurposed drugs have been suggested as potential treatments, preclinical data from animal models can guide the search for effective treatments by ruling out those that lack efficacy in vivo. Remdesivir (GS-5734) is a nucleotide analogue prodrug with broad antiviral activity1,2 that is currently being investigated in COVID-19 clinical trials and recently received Emergency Use Authorization from the US Food and Drug Administration3,4. In animal models, remdesivir was effective against infection with Middle East respiratory syndrome coronavirus (MERS-CoV) and severe acute respiratory syndrome coronavirus (SARS-CoV)2,5,6. In vitro, remdesivir inhibited replication of SARS-CoV-27,8. Here we investigate the efficacy of remdesivir in a rhesus macaque model of SARS-CoV-2 infection9. Unlike vehicle-treated animals, macaques treated with remdesivir did not show signs of respiratory disease; they also showed reduced pulmonary infiltrates on radiographs and reduced virus titres in bronchoalveolar lavages twelve hours after the first dose. Virus shedding from the upper respiratory tract was not reduced by remdesivir treatment. At necropsy, remdesivir-treated animals had lower lung viral loads and reduced lung damage. Thus, treatment with remdesivir initiated early during infection had a clinical benefit in rhesus macaques infected with SARS-CoV-2. Although the rhesus macaque model does not represent the severe disease observed in some patients with COVID-19, our data support the early initiation of remdesivir treatment in patients with COVID-19 to prevent progression to pneumonia.


Asunto(s)
Adenosina Monofosfato/análogos & derivados , Alanina/análogos & derivados , Betacoronavirus/efectos de los fármacos , Infecciones por Coronavirus/tratamiento farmacológico , Infecciones por Coronavirus/virología , Modelos Animales de Enfermedad , Macaca mulatta/virología , Neumonía Viral/prevención & control , Adenosina Monofosfato/farmacocinética , Adenosina Monofosfato/farmacología , Adenosina Monofosfato/uso terapéutico , Alanina/farmacocinética , Alanina/farmacología , Alanina/uso terapéutico , Animales , Betacoronavirus/genética , Betacoronavirus/patogenicidad , Líquido del Lavado Bronquioalveolar/virología , COVID-19 , Infecciones por Coronavirus/patología , Infecciones por Coronavirus/fisiopatología , Análisis Mutacional de ADN , Progresión de la Enfermedad , Farmacorresistencia Viral , Femenino , Pulmón/efectos de los fármacos , Pulmón/patología , Pulmón/fisiopatología , Pulmón/virología , Masculino , Pandemias , Neumonía Viral/tratamiento farmacológico , Neumonía Viral/patología , Neumonía Viral/fisiopatología , Neumonía Viral/virología , SARS-CoV-2 , Prevención Secundaria , Factores de Tiempo , Carga Viral/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Esparcimiento de Virus/efectos de los fármacos
4.
mBio ; 10(6)2019 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-31848292

RESUMEN

Klebsiella pneumoniae is a human gut communal organism and notorious opportunistic pathogen. The relative high burden of asymptomatic colonization by K. pneumoniae is often compounded by multidrug resistance-a potential problem for individuals with significant comorbidities or other risk factors for infection. A carbapenem-resistant K. pneumoniae strain classified as multilocus sequence type 258 (ST258) is widespread in the United States and is usually multidrug resistant. Thus, treatment of ST258 infections is often difficult. Inasmuch as new preventive and/or therapeutic measures are needed for treatment of such infections, we developed an ST258 pneumonia model in cynomolgus macaques and tested the ability of an ST258 capsule polysaccharide type 2 (CPS2) vaccine to moderate disease severity. Compared with sham-vaccinated animals, those vaccinated with ST258 CPS2 had significantly less disease as assessed by radiography 24 h after intrabronchial installation of 108 CFU of ST258. All macaques vaccinated with CPS2 ultimately developed ST258-specific antibodies that significantly enhanced serum bactericidal activity and killing of ST258 by macaque neutrophils ex vivo Consistent with a protective immune response to CPS2, transcripts encoding inflammatory mediators were increased in infected lung tissues obtained from CPS-vaccinated animals compared with control, sham-vaccinated macaques. Taken together, our data provide support for the idea that vaccination with ST258 CPS can be used to prevent or moderate infections caused by ST258. As with studies performed decades earlier, we propose that this prime-boost vaccination approach can be extended to include multiple capsule types.IMPORTANCE Multidrug-resistant bacteria continue to be a major problem worldwide, especially among individuals with significant comorbidities and other risk factors for infection. K. pneumoniae is among the leading causes of health care-associated infections, and the organism is often resistant to multiple classes of antibiotics. A carbapenem-resistant K. pneumoniae strain known as multilocus sequence type 258 (ST258) is the predominant carbapenem-resistant Enterobacteriaceae in the health care setting in the United States. Infections caused by ST258 are often difficult to treat and new prophylactic measures and therapeutic approaches are needed. To that end, we developed a lower respiratory tract infection model in cynomolgus macaques in which to test the ability of ST258 CPS to protect against severe ST258 infection.


Asunto(s)
Vacunas Bacterianas/inmunología , Farmacorresistencia Bacteriana Múltiple , Klebsiella pneumoniae/efectos de los fármacos , Klebsiella pneumoniae/inmunología , Infecciones del Sistema Respiratorio/microbiología , Infecciones del Sistema Respiratorio/prevención & control , Animales , Biopsia , Inmunización , Infecciones por Klebsiella/diagnóstico , Infecciones por Klebsiella/microbiología , Infecciones por Klebsiella/prevención & control , Primates , Radiografía , Infecciones del Sistema Respiratorio/diagnóstico , Transcriptoma , Vacunación
5.
J Infect Dis ; 218(suppl_5): S458-S465, 2018 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-30215737

RESUMEN

Filoviruses are among the most pathogenic infectious agents known to human, with high destructive potential, as evidenced by the recent Ebola virus epidemic in West Africa. As members of the filovirus family, marburgviruses have caused similar devastating outbreaks, albeit with lower case numbers. In this study we compare the pathogenesis of Ravn virus (RAVV) and Marburg virus (MARV) strains Angola, Musoke, and Ozolin in rhesus and cynomolgus macaques, the 2 nonhuman primate species most commonly used in filovirus research. Our results reveal the most pathogenic MARV strain to be Angola, followed by Musoke, whereas Ozolin is the least pathogenic. We also demonstrate that RAVV is highly pathogenic in cynomolgus macaques but less pathogenic in rhesus macaques. Our results demonstrate a preferential infection of endothelial cells by MARVs; in addition, analysis of tissue samples suggests that lymphocyte and hepatocyte apoptosis might play a role in MARV pathogenicity. This information expands our knowledge about pathogenicity and virulence of marburgviruses.


Asunto(s)
Enfermedad del Virus de Marburg/etiología , Marburgvirus/patogenicidad , Animales , Apoptosis , Hepatocitos/patología , Macaca fascicularis , Macaca mulatta , Macrófagos/patología , Masculino , Enfermedad del Virus de Marburg/inmunología , Enfermedad del Virus de Marburg/patología , Fenotipo
6.
PLoS Negl Trop Dis ; 12(7): e0006628, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-30011277

RESUMEN

Crimean-Congo hemorrhagic fever (CCHF) is an acute, often fatal viral disease characterized by rapid onset of febrile symptoms followed by hemorrhagic manifestations. The etiologic agent, CCHF orthonairovirus (CCHFV), can infect several mammals in nature but only seems to cause clinical disease in humans. Over the past two decades there has been an increase in total number of CCHF case reports, including imported CCHF patients, and an expansion of CCHF endemic areas. Despite its increased public health burden there are currently no licensed vaccines or treatments to prevent CCHF. We here report the development and assessment of the protective efficacy of an adenovirus (Ad)-based vaccine expressing the nucleocapsid protein (N) of CCHFV (Ad-N) in a lethal immunocompromised mouse model of CCHF. The results show that Ad-N can protect mice from CCHF mortality and that this platform should be considered for future CCHFV vaccine strategies.


Asunto(s)
Virus de la Fiebre Hemorrágica de Crimea-Congo/inmunología , Fiebre Hemorrágica de Crimea/prevención & control , Proteínas de la Nucleocápside/inmunología , Vacunas Virales/inmunología , Animales , Anticuerpos Antivirales/inmunología , Modelos Animales de Enfermedad , Femenino , Virus de la Fiebre Hemorrágica de Crimea-Congo/genética , Fiebre Hemorrágica de Crimea/inmunología , Fiebre Hemorrágica de Crimea/virología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas de la Nucleocápside/administración & dosificación , Proteínas de la Nucleocápside/genética , Vacunas Virales/administración & dosificación , Vacunas Virales/genética
7.
Am J Respir Cell Mol Biol ; 57(5): 536-546, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28613916

RESUMEN

During severe influenza A virus (IAV) infections, a large amount of damage to the pulmonary epithelium is the result of the antiviral immune response. Specifically, whilst CD8+ T cells are important for killing IAV-infected cells, during a severe IAV infection, they can damage uninfected epithelial cells. At present, the mechanisms by which this occurs are unclear. Here, we used a novel in vitro coculture model of human NCl-H441 cells and CD8+ T cells to provide a new insight into how CD8+ T cells may affect uninfected epithelial cells during severe IAV infections. Using this model, we show that human IAV-specific CD8+ T cells produce soluble factors that reduce the barrier integrity of noninfected epithelial cells (referred to as "bystander damage"). We show that this bystander damage is the result of a combination of TNF-α and IFN-γ. This bystander damage occurred in the absence of widespread epithelial cell death and was instead associated with decreased expression of epithelial cell ion channels and pumps. Together, these data suggest that ameliorating the function of epithelial cell ion channels and pumps may help reduce immunopathology during severe IAV infections.


Asunto(s)
Linfocitos T CD8-positivos/virología , Células Epiteliales/virología , Subtipo H1N1 del Virus de la Influenza A , Gripe Humana/virología , Pulmón/virología , Linfocitos T CD8-positivos/inmunología , Humanos , Pulmón/patología , Factor de Necrosis Tumoral alfa/metabolismo
8.
Cell Rep ; 18(3): 816-829, 2017 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-28099857

RESUMEN

The unprecedented 2013-2016 outbreak of Ebola virus (EBOV) resulted in over 11,300 human deaths. Host resistance to RNA viruses requires RIG-I-like receptor (RLR) signaling through the adaptor protein, mitochondrial antiviral signaling protein (MAVS), but the role of RLR-MAVS in orchestrating anti-EBOV responses in vivo is not known. Here we apply a systems approach to MAVS-/- mice infected with either wild-type or mouse-adapted EBOV. MAVS controlled EBOV replication through the expression of IFNα, regulation of inflammatory responses in the spleen, and prevention of cell death in the liver, with macrophages implicated as a major cell type influencing host resistance. A dominant role for RLR signaling in macrophages was confirmed following conditional MAVS deletion in LysM+ myeloid cells. These findings reveal tissue-specific MAVS-dependent transcriptional pathways associated with resistance to EBOV, and they demonstrate that EBOV adaptation to cause disease in mice involves changes in two distinct events, RLR-MAVS antagonism and suppression of RLR-independent IFN-I responses.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Ebolavirus/fisiología , Fiebre Hemorrágica Ebola/patología , Proteínas Adaptadoras Transductoras de Señales/antagonistas & inhibidores , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Proteína 58 DEAD Box/antagonistas & inhibidores , Proteína 58 DEAD Box/metabolismo , Modelos Animales de Enfermedad , Fiebre Hemorrágica Ebola/metabolismo , Fiebre Hemorrágica Ebola/mortalidad , Humanos , Interferón Tipo I/metabolismo , Estimación de Kaplan-Meier , Hígado/metabolismo , Hígado/patología , Hígado/virología , Macrófagos/citología , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Mieloides/citología , Células Mieloides/metabolismo , Células Mieloides/virología , Transducción de Señal , Bazo/metabolismo , Bazo/patología , Bazo/virología , Replicación Viral
9.
J Infect Dis ; 214(suppl 3): S308-S318, 2016 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-27601621

RESUMEN

The study of Ebola virus (EBOV) pathogenesis in vivo has been limited to nonhuman primate models or use of an adapted virus to cause disease in rodent models. Herein we describe wild-type EBOV (Makona variant) infection of mice engrafted with human hematopoietic CD34+ stem cells (Hu-NSG™-SGM3 mice; hereafter referred to as SGM3 HuMice). SGM3 HuMice support increased development of myeloid immune cells, which are primary EBOV targets. In SGM3 HuMice, EBOV replicated to high levels, and disease was observed following either intraperitoneal or intramuscular inoculation. Despite the high levels of viral antigen and inflammatory cell infiltration in the liver, the characteristic histopathology of Ebola virus disease was not observed, and this absence of severe immunopathology may have contributed to the recovery and survival of some of the animals. Future investigations into the underlying mechanisms of the atypical disease presentation in SGM3 HuMice will provide additional insights into the immunopathogenesis of severe EBOV disease.


Asunto(s)
Antígenos Virales/inmunología , Ebolavirus/fisiología , Fiebre Hemorrágica Ebola/virología , Animales , Modelos Animales de Enfermedad , Ebolavirus/inmunología , Células Madre Hematopoyéticas/inmunología , Células Madre Hematopoyéticas/patología , Células Madre Hematopoyéticas/virología , Fiebre Hemorrágica Ebola/inmunología , Fiebre Hemorrágica Ebola/patología , Humanos , Hígado/inmunología , Hígado/patología , Hígado/virología , Linfocitos/patología , Linfocitos/virología , Ratones , Ratones Transgénicos , Células Mieloides/inmunología , Células Mieloides/patología , Células Mieloides/virología , Bazo/inmunología , Bazo/patología , Bazo/virología , Transgenes , Replicación Viral
10.
Am J Pathol ; 186(3): 630-8, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26724387

RESUMEN

Middle East respiratory syndrome coronavirus (MERS-CoV) was first identified in a human with severe pneumonia in 2012. Since then, infections have been detected in >1500 individuals, with disease severity ranging from asymptomatic to severe, fatal pneumonia. To elucidate the pathogenesis of this virus and investigate mechanisms underlying disease severity variation in the absence of autopsy data, a rhesus macaque and common marmoset model of MERS-CoV disease were analyzed. Rhesus macaques developed mild disease, and common marmosets exhibited moderate to severe, potentially lethal, disease. Both nonhuman primate species exhibited respiratory clinical signs after inoculation, which were more severe and of longer duration in the marmosets, and developed bronchointerstitial pneumonia. In marmosets, the pneumonia was more extensive, with development of severe airway lesions. Quantitative analysis showed significantly higher levels of pulmonary neutrophil infiltration and higher amounts of pulmonary viral antigen in marmosets. Pulmonary expression of the MERS-CoV receptor, dipeptidyl peptidase 4, was similar in marmosets and macaques. These results suggest that increased virus replication and the local immune response to MERS-CoV infection likely play a role in pulmonary pathology severity. Together, the rhesus macaque and common marmoset models of MERS-CoV span the wide range of disease severity reported in MERS-CoV-infected humans, which will aid in investigating MERS-CoV disease pathogenesis.


Asunto(s)
Antígenos Virales/sangre , Infecciones por Coronavirus/inmunología , Coronavirus del Síndrome Respiratorio de Oriente Medio/inmunología , Neumonía Viral/inmunología , Replicación Viral/inmunología , Animales , Antígenos Virales/análisis , Callithrix , Infecciones por Coronavirus/virología , Dipeptidil Peptidasa 4/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Pulmón/inmunología , Pulmón/patología , Macaca mulatta , Macrófagos Alveolares/clasificación , Masculino , Coronavirus del Síndrome Respiratorio de Oriente Medio/patogenicidad , Coronavirus del Síndrome Respiratorio de Oriente Medio/fisiología , Neutrófilos/inmunología , Conejos , Carga Viral , Virulencia
11.
Eur Respir J ; 47(3): 954-66, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26743480

RESUMEN

A major cause of respiratory failure during influenza A virus (IAV) infection is damage to the epithelial-endothelial barrier of the pulmonary alveolus. Damage to this barrier results in flooding of the alveolar lumen with proteinaceous oedema fluid, erythrocytes and inflammatory cells. To date, the exact roles of pulmonary epithelial and endothelial cells in this process remain unclear.Here, we used an in vitro co-culture model to understand how IAV damages the pulmonary epithelial-endothelial barrier. Human epithelial cells were seeded on the upper half of a transwell membrane while human endothelial cells were seeded on the lower half. These cells were then grown in co-culture and IAV was added to the upper chamber.We showed that the addition of IAV (H1N1 and H5N1 subtypes) resulted in significant barrier damage. Interestingly, we found that, while endothelial cells mounted a pro-inflammatory/pro-coagulant response to a viral infection in the adjacent epithelial cells, damage to the alveolar epithelial-endothelial barrier occurred independently of endothelial cells. Rather, barrier damage was associated with disruption of tight junctions amongst epithelial cells, and specifically with loss of tight junction protein claudin-4.Taken together, these data suggest that maintaining epithelial cell integrity is key in reducing pulmonary oedema during IAV infection.


Asunto(s)
Células Epiteliales/virología , Subtipo H1N1 del Virus de la Influenza A/patogenicidad , Subtipo H5N1 del Virus de la Influenza A/patogenicidad , Alveolos Pulmonares/virología , Uniones Estrechas/ultraestructura , Línea Celular , Técnicas de Cocultivo , Citocinas/metabolismo , Células Epiteliales/patología , Humanos
12.
PLoS One ; 8(12): e82096, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24324751

RESUMEN

Virulent Francisella tularensis ssp tularensis is an intracellular, Gram negative bacterium that causes acute lethal disease following inhalation of fewer than 15 organisms. Pathogenicity of Francisella infections is tied to its unique ability to evade and suppress inflammatory responses in host cells. It has been proposed that induction of alternative activation of infected macrophages is a mechanism by which attenuated Francisella species modulate host responses. In this report we reveal that neither attenuated F. tularensis Live Vaccine Strain (LVS) nor virulent F. tularensis strain SchuS4 induce alternative activation of macrophages in vitro or in vivo. LVS, but not SchuS4, provoked production of arginase1 independent of alternative activation in vitro and in vivo. However, absence of arginase1 did not significantly impact intracellular replication of LVS or SchuS4. Together our data establish that neither induction of alternative activation nor expression of arginase1 are critical features of disease mediated by attenuated or virulent Francisella species.


Asunto(s)
Arginasa/biosíntesis , Francisella tularensis/fisiología , Activación de Macrófagos , Macrófagos/enzimología , Macrófagos/microbiología , Animales , Citocinas/metabolismo , Inducción Enzimática , Francisella tularensis/crecimiento & desarrollo , Francisella tularensis/patogenicidad , Pulmón/inmunología , Pulmón/microbiología , Pulmón/patología , Macrófagos/inmunología , Macrófagos/patología , Ratones , Ratones Endogámicos C57BL , Tularemia/inmunología , Tularemia/microbiología , Tularemia/prevención & control , Vacunas Atenuadas/inmunología
13.
J Infect Dis ; 207(8): 1316-27, 2013 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-23303805

RESUMEN

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.


Asunto(s)
Fiebre de Lassa/patología , Virus Lassa/patogenicidad , Animales , Quimiocina CCL3/sangre , Quimiocina CCL3/inmunología , Chlorocebus aethiops , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Factor Estimulante de Colonias de Granulocitos/sangre , Factor Estimulante de Colonias de Granulocitos/inmunología , Cobayas , Pruebas Hematológicas , Inmunohistoquímica , Interleucina-1beta/sangre , Interleucina-1beta/inmunología , Interleucina-6/sangre , Interleucina-6/inmunología , Fiebre de Lassa/inmunología , Fiebre de Lassa/virología , Virus Lassa/genética , Virus Lassa/inmunología , Virus Lassa/aislamiento & purificación , Pulmón/patología , Pulmón/virología , Macaca fascicularis , Masculino , Malí , ARN Viral/sangre , Factores de Tiempo , Factor de Necrosis Tumoral alfa/sangre , Factor de Necrosis Tumoral alfa/inmunología , Células Vero , Carga Viral , Viremia/virología , Virulencia
14.
Proc Natl Acad Sci U S A ; 110(5): 1893-8, 2013 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-23319647

RESUMEN

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.


Asunto(s)
Anticuerpos Antivirales/inmunología , Vacunas contra el Virus del Ébola/inmunología , Ebolavirus/inmunología , Fiebre Hemorrágica Ebola/inmunología , Glicoproteínas de Membrana/inmunología , Proteínas del Envoltorio Viral/inmunología , Animales , Anticuerpos Antivirales/sangre , Citocinas/sangre , Citocinas/inmunología , Vacunas contra el Virus del Ébola/administración & dosificación , Ebolavirus/genética , Ensayo de Inmunoadsorción Enzimática , Fiebre Hemorrágica Ebola/sangre , Fiebre Hemorrágica Ebola/prevención & control , Humanos , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Hígado/inmunología , Hígado/parasitología , Hígado/patología , Linfocitos/inmunología , Macaca fascicularis , Masculino , Marburgvirus/genética , Marburgvirus/inmunología , Glicoproteínas de Membrana/genética , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Bazo/inmunología , Bazo/parasitología , Bazo/patología , Factores de Tiempo , Virus de la Estomatitis Vesicular Indiana/genética , Virus de la Estomatitis Vesicular Indiana/inmunología , Proteínas del Envoltorio Viral/genética , Carga Viral/genética
16.
J Parasitol ; 91(3): 695-7, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16108571

RESUMEN

Toxoplasma gondii infection in marine mammals is intriguing and indicative of contamination of the ocean environment with oocysts. T. gondii was identified in a Hawaiian monk seal (Monachus schauinslandi) that had visceral and cerebral lesions. Tachyzoites were found in the lymph nodes, spleen, diaphragm, heart, adrenal glands, and brain. A few tissue cysts were found in sections of the cerebrum. The diagnosis was confirmed serologically, by immunohistochemical staining with T. gondii-specific polyclonal rabbit serum, and by the detection of T. gondii DNA. The genotype was determined to be type III by restriction fragment length polymorphisms of the SAG2 gene. This is the first report of T. gondii infection in a Hawaiian monk seal.


Asunto(s)
Phocidae/parasitología , Toxoplasma/aislamiento & purificación , Toxoplasmosis Animal/diagnóstico , Corteza Suprarrenal/parasitología , Corteza Suprarrenal/patología , Animales , Encéfalo/parasitología , Encéfalo/patología , ADN Protozoario/análisis , Diafragma/parasitología , Diafragma/patología , Genotipo , Hawaii , Corazón/parasitología , Inmunohistoquímica/veterinaria , Ganglios Linfáticos/parasitología , Ganglios Linfáticos/patología , Masculino , Bazo/parasitología , Bazo/patología , Toxoplasma/clasificación , Toxoplasma/genética , Toxoplasmosis Animal/parasitología , Toxoplasmosis Animal/patología
17.
Am J Pathol ; 163(6): 2347-70, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-14633608

RESUMEN

Ebola virus (EBOV) infection causes a severe and fatal hemorrhagic disease that in many ways appears to be similar in humans and nonhuman primates; however, little is known about the development of EBOV hemorrhagic fever. In the present study, 21 cynomolgus monkeys were experimentally infected with EBOV and examined sequentially over a 6-day period to investigate the pathological events of EBOV infection that lead to death. Importantly, dendritic cells in lymphoid tissues were identified as early and sustained targets of EBOV, implicating their important role in the immunosuppression characteristic of EBOV infections. Bystander lymphocyte apoptosis, previously described in end-stage tissues, occurred early in the disease-course in intravascular and extravascular locations. Of note, apoptosis and loss of NK cells was a prominent finding, suggesting the importance of innate immunity in determining the fate of the host. Analysis of peripheral blood mononuclear cell gene expression showed temporal increases in tumor necrosis factor-related apoptosis-inducing ligand and Fas transcripts, revealing a possible mechanism for the observed bystander apoptosis, while up-regulation of NAIP and cIAP2 mRNA suggest that EBOV has evolved additional mechanisms to resist host defenses by inducing protective transcripts in cells that it infects. The sequence of pathogenetic events identified in this study should provide new targets for rational prophylactic and chemotherapeutic interventions.


Asunto(s)
Células Dendríticas/virología , Fiebre Hemorrágica Ebola/virología , Animales , Quimiocinas/sangre , Enfermedad Crítica , Citocinas/sangre , Endotoxinas/sangre , Citometría de Flujo , Fiebre Hemorrágica Ebola/metabolismo , Fiebre Hemorrágica Ebola/patología , Fiebre Hemorrágica Ebola/fisiopatología , Recuento de Leucocitos , Estudios Longitudinales , Macaca fascicularis , Masculino , Nitratos/sangre , Factores de Tiempo , Carga Viral , Viremia/virología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA