Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
Sci Rep ; 13(1): 757, 2023 01 14.
Artículo en Inglés | MEDLINE | ID: mdl-36641520

RESUMEN

Heterogeneity of COVID-19 manifestations in human population is vast, for reasons unknown. Cotton rats are a clinically relevant small animal model of human respiratory viral infections. Here, we demonstrate for the first time that SARS-CoV-2 infection in cotton rats affects multiple organs and systems, targeting species- and age-specific biological processes. Infection of S. fulviventer, which developed a neutralizing antibody response and were more susceptible to SARS-CoV-2 replication in the upper respiratory tract, was accompanied by hyperplasia of lacrimal drainage-associated lymphoid tissue (LDALT), a first known report of mucosa-associated lymphoid tissue activation at the portal of SARS-CoV-2 entry. Although less permissive to viral replication, S. hispidus showed hyperplasia of bone marrow in the facial bones and increased pulmonary thrombosis in aged males. Augmentation of these features by SARS-CoV-2 infection suggests a virus-induced breach in regulatory mechanisms which could be devastating for people of all ages with underlying conditions and in particular for elderly with a multitude of ongoing disorders.


Asunto(s)
COVID-19 , Masculino , Animales , Humanos , Anciano , Sigmodontinae , Hiperplasia , SARS-CoV-2 , Factores de Edad
2.
Sci Transl Med ; 13(616): eabj7843, 2021 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-34669442

RESUMEN

In 1967, two toddlers immunized with a formalin-inactivated vaccine against respiratory syncytial virus (FIRSV) in the United States died from enhanced RSV disease (ERD), a severe form of illness resulting from aberrant priming of the antiviral immune response during vaccination. Up to 80% of immunized children subsequently exposed to wild-type virus were hospitalized. These events hampered RSV vaccine development for decades. Here, we provide a characterization of the clinical, immunopathological, and transcriptional signature of fatal human ERD, outlining evidence for safety evaluation of RSV vaccines and a framework for understanding disease enhancement for pathogens in general.


Asunto(s)
Enfermedades Transmisibles , Infecciones por Virus Sincitial Respiratorio , Vacunas contra Virus Sincitial Respiratorio , Preescolar , Humanos , Infecciones por Virus Sincitial Respiratorio/epidemiología , Virus Sincitiales Respiratorios
3.
J Virol ; 91(8)2017 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-28148790

RESUMEN

Respiratory syncytial virus (RSV) infection of children previously immunized with a nonlive, formalin-inactivated (FI)-RSV vaccine has been associated with serious enhanced respiratory disease (ERD). Consequently, detailed studies of potential ERD are a critical step in the development of nonlive RSV vaccines targeting RSV-naive children and infants. The fusion glycoprotein (F) of RSV in either its postfusion (post-F) or prefusion (pre-F) conformation is a target for neutralizing antibodies and therefore an attractive antigen candidate for a pediatric RSV subunit vaccine. Here, we report the evaluation of RSV post-F and pre-F in combination with glucopyranosyl lipid A (GLA) integrated into stable emulsion (SE) (GLA-SE) and alum adjuvants in the cotton rat model. Immunization with optimal doses of RSV F antigens in the presence of GLA-SE induced high titers of virus-neutralizing antibodies and conferred complete lung protection from virus challenge, with no ERD signs in the form of alveolitis. To mimic a waning immune response, and to assess priming for ERD under suboptimal conditions, an antigen dose de-escalation study was performed in the presence of either GLA-SE or alum. At low RSV F doses, alveolitis-associated histopathology was unexpectedly observed with either adjuvant at levels comparable to FI-RSV-immunized controls. This occurred despite neutralizing-antibody titers above the minimum levels required for protection and with no/low virus replication in the lungs. These results emphasize the need to investigate a pediatric RSV vaccine candidate carefully for priming of ERD over a wide dose range, even in the presence of strong neutralizing activity, Th1 bias-inducing adjuvant, and protection from virus replication in the lower respiratory tract.IMPORTANCE RSV disease is of great importance worldwide, with the highest burden of serious disease occurring upon primary infection in infants and children. FI-RSV-induced enhanced disease, observed in the 1960s, presented a major and ongoing obstacle for the development of nonlive RSV vaccine candidates. The findings presented here underscore the need to evaluate a nonlive RSV vaccine candidate during preclinical development over a wide dose range in the cotton rat RSV enhanced-disease model, as suboptimal dosing of several RSV F subunit vaccine candidates led to the priming for ERD. These observations are relevant to the validity of the cotton rat model itself and to safe development of nonlive RSV vaccines for seronegative infants and children.


Asunto(s)
Acrecentamiento Dependiente de Anticuerpo , Infecciones por Virus Sincitial Respiratorio/inmunología , Vacunas contra Virus Sincitial Respiratorio/inmunología , Células TH1/inmunología , Células Th2/inmunología , Proteínas Virales de Fusión/inmunología , Adyuvantes Inmunológicos/administración & dosificación , Compuestos de Alumbre/administración & dosificación , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Modelos Animales de Enfermedad , Lípido A/administración & dosificación , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Infecciones por Virus Sincitial Respiratorio/prevención & control , Vacunas contra Virus Sincitial Respiratorio/genética , Sigmodontinae , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Proteínas Virales de Fusión/genética
4.
Hum Vaccin ; 6(6): 482-92, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20671419

RESUMEN

Respiratory Syncytial Virus (RSV) is the leading cause of pneumonia and bronchiolitis in infants and children <1 year old, resulting in significant morbidity and mortality worldwide. There is currently no RSV vaccine. In the 1960s, a formalin-inactivated RSV (FI-RSV) vaccine trial led to exacerbated disease upon natural infection of vaccinees, including two deaths. The causes involved in the disastrous results of these vaccine trials are still unclear but they remain the engine for searching new avenues to develop a safe vaccine that can provide long-term protection against this important pathogen. This article reviews some of the early history of RSV vaccine development,as well as more recent information on the interaction between RSV and the host innate and adaptive immune responses. A safe and efficacious vaccine for RSV will require "re-education" of the host immune response against RSV to prevent vaccine-enhanced or severe RSV disease.


Asunto(s)
Infecciones por Virus Sincitial Respiratorio/prevención & control , Vacunas contra Virus Sincitial Respiratorio/inmunología , Virus Sincitiales Respiratorios/inmunología , Inmunidad Adaptativa , Animales , Ensayos Clínicos como Asunto , Interacciones Huésped-Patógeno , Humanos , Inmunidad Innata , Infecciones por Virus Sincitial Respiratorio/patología , Infecciones por Virus Sincitial Respiratorio/virología , Vacunas contra Virus Sincitial Respiratorio/efectos adversos , Virus Sincitiales Respiratorios/patogenicidad , Receptor Toll-Like 4/inmunología , Receptor Toll-Like 4/metabolismo , Proteínas Virales de Fusión/metabolismo
5.
Antiviral Res ; 88(1): 31-7, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20619294

RESUMEN

Human metapneumovirus (HMPV) is a paramyxovirus causing acute respiratory tract infections in humans. The effects of a monoclonal antibody (MAb 338, MedImmune, Inc.) directed against the HMPV fusion protein were assessed in vivo. Different groups of BALB/c mice received an intraperitoneal injection of 25 or 50mg/kg of MAb 338 either 24h before or 48h after viral infection. Lung samples were collected on days 5 and 42 after infection for determination of viral titers and histopathological changes. Pulmonary functions were also evaluated by plethysmography. On day 5 post-infection, lung viral titers were significantly decreased in mice treated with 25 or 50mg/kg before or after viral infection compared to HMPV-infected control mice. Similarly, HMPV copy numbers on day 42 were decreased for all prophylactic and therapeutic interventions. Histopathological changes were also less severe in all treated groups of mice on days 5 and 42 post-infection, correlating with decreased airways obstruction. Finally, on day 42, all treated groups had a significant decrease in airways hyperresponsiveness following treatment with MAb 338. Both prophylactic and, to a lesser extent, therapeutic administration of MAb 338 improved acute and late consequences of HMPV infection in a relevant mouse model.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Metapneumovirus/inmunología , Infecciones por Paramyxoviridae/tratamiento farmacológico , Infecciones por Paramyxoviridae/prevención & control , Infecciones del Sistema Respiratorio/tratamiento farmacológico , Infecciones del Sistema Respiratorio/prevención & control , Proteínas Virales de Fusión/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Antivirales/inmunología , Anticuerpos Antivirales/uso terapéutico , Antivirales/inmunología , Antivirales/uso terapéutico , Modelos Animales de Enfermedad , Pulmón/inmunología , Pulmón/patología , Pulmón/virología , Enfermedades Pulmonares Obstructivas/tratamiento farmacológico , Enfermedades Pulmonares Obstructivas/patología , Ratones , Ratones Endogámicos BALB C , Infecciones por Paramyxoviridae/patología , Infecciones por Paramyxoviridae/virología , Neumonía/tratamiento farmacológico , Neumonía/inmunología , Reacción en Cadena de la Polimerasa , Hipersensibilidad Respiratoria , Sistema Respiratorio , Infecciones del Sistema Respiratorio/patología , Infecciones del Sistema Respiratorio/virología
6.
Curr Protoc Cell Biol ; Chapter 26: Unit26.6, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20235102

RESUMEN

Viral infection is normally detected either by viral culture or by PCR methods. Rarely is a combination of the two techniques used in the same study. Yet, when applied simultaneously, viral culture and PCR may reveal important features of viral biology, such as an abortive replication, as in the case of respiratory syncytial virus (RSV) infection. In this unit, we describe methods for detecting abortive RSV replication in a cotton rat model by using the plaque-forming unit assay and the real-time reverse-transcription PCR (qRT-PCR) assay. All steps of the process of monitoring viral replication in vivo are described, starting from the design of animal infection protocols. We continue on to the methods for extracting and processing lung samples for viral culture and RNA extraction, and finish with the actual methods of viral titration by the qRT-PCR and the plaque-forming unit assays.


Asunto(s)
Pulmón/virología , Virus Sincitiales Respiratorios/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Cultivo de Virus/métodos , Replicación Viral/fisiología , Animales , Línea Celular , Células Epiteliales/virología , Pulmón/patología , Ratas , Infecciones por Virus Sincitial Respiratorio/genética , Infecciones por Virus Sincitial Respiratorio/virología
7.
Virol J ; 7: 20, 2010 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-20102602

RESUMEN

BACKGROUND: Infectivity of retroviruses such as HIV-1 and MuLV can be abrogated by compounds targeting zinc finger motif in viral nucleocapsid protein (NC), involved in controlling the processivity of reverse transcription and virus infectivity. Although a member of a different viral family (Pneumoviridae), respiratory syncytial virus (RSV) contains a zinc finger protein M2-1 also involved in control of viral polymerase processivity. Given the functional similarity between the two proteins, it was possible that zinc finger-reactive compounds inactivating retroviruses would have a similar effect against RSV by targeting RSV M2-1 protein. Moreover, inactivation of RSV through modification of an internal protein could yield a safer whole virus vaccine than that produced by RSV inactivation with formalin which modifies surface proteins. RESULTS: Three compounds were evaluated for their ability to reduce RSV infectivity: 2,2'-dithiodipyridine (AT-2), tetraethylthiuram disulfide and tetramethylthiuram disulfide. All three were capable of inactivating RSV, with AT-2 being the most potent. The mechanism of action of AT-2 was analyzed and it was found that AT-2 treatment indeed results in the modification of RSV M2-1. Altered intramolecular disulfide bond formation in M2-1 protein of AT-2-treated RSV virions might have been responsible for abrogation of RSV infectivity. AT-2-inactivated RSV was found to be moderately immunogenic in the cotton rats S.hispidus and did not cause a vaccine-enhancement seen in animals vaccinated with formalin-inactivated RSV. Increasing immunogenicity of AT-2-inactivated RSV by adjuvant (Ribi), however, led to vaccine-enhanced disease. CONCLUSIONS: This work presents evidence that compounds that inactivate retroviruses by targeting the zinc finger motif in their nucleocapsid proteins are also effective against RSV. AT-2-inactivated RSV vaccine is not strongly immunogenic in the absence of adjuvants. In the adjuvanted form, however, vaccine induces immunopathologic response. The mere preservation of surface antigens of RSV, therefore may not be sufficient to produce a highly-efficacious inactivated virus vaccine that does not lead to an atypical disease.


Asunto(s)
Antivirales/farmacología , Virus Sincitiales Respiratorios/efectos de los fármacos , Proteínas Virales/antagonistas & inhibidores , Replicación Viral/efectos de los fármacos , Dedos de Zinc , 2,2'-Dipiridil/análogos & derivados , 2,2'-Dipiridil/metabolismo , 2,2'-Dipiridil/farmacología , Adyuvantes Inmunológicos/administración & dosificación , Animales , Antivirales/metabolismo , Esqueleto de la Pared Celular/administración & dosificación , Factores Cordón/administración & dosificación , Disulfuros/metabolismo , Disulfuros/farmacología , Disulfiram/metabolismo , Disulfiram/farmacología , Lípido A/administración & dosificación , Lípido A/análogos & derivados , Unión Proteica , Ratas , Vacunas contra Virus Sincitial Respiratorio/inmunología , Virus Sincitiales Respiratorios/inmunología , Virus Sincitiales Respiratorios/fisiología , Sigmodontinae/virología , Tiram/metabolismo , Tiram/farmacología , Vacunas Atenuadas/inmunología , Proteínas Virales/metabolismo
8.
J Interferon Cytokine Res ; 30(4): 229-42, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20038196

RESUMEN

Interferon (IFN) therapy in humans often causes flu-like symptoms by an unknown mechanism. Poly ICLC is a synthetic dsRNA and a Toll-like receptor 3 (TLR3) agonist with a strong IFN-inducing ability. In this work, we analyzed the effect of poly ICLC on pulmonary responses to influenza and respiratory syncytial virus (RSV) infections in the cotton rat (Sigmodon hispidus) model. Viral replication, pulmonary inflammation, and expression of IFN, TLR, and chemokines were monitored and compared. Antiviral effect of poly ICLC against influenza virus and RSV was best achieved at high poly ICLC concentrations that, in the absence of virus infection, induced a strong IFN response. The antiviral doses of poly ICLC, however, also increased lung inflammation, an unexpected finding because of the reported poly ICLC safety in BALB/c mice. Similarly, in contrast to murine model, pathology of RSV infection was increased in cotton rats treated with poly ICLC. Augmented lung inflammation was accompanied by an earlier induction of IFN and TLR responses and a stronger chemokine expression. Overall, these findings indicate significant association between antiviral IFN action and pulmonary inflammation and highlight important animal model-specific variations in the potential of IFN to cause pathology.


Asunto(s)
Interferones/inmunología , Pulmón/virología , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Virus Sincitial Respiratorio/inmunología , Sigmodontinae/inmunología , Sigmodontinae/virología , Receptor Toll-Like 3/metabolismo , Animales , Lavado Broncoalveolar , Carboximetilcelulosa de Sodio/análogos & derivados , Carboximetilcelulosa de Sodio/farmacología , Quimiocinas/genética , Quimiocinas/metabolismo , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Virus de la Influenza A/efectos de los fármacos , Virus de la Influenza A/inmunología , Pulmón/efectos de los fármacos , Pulmón/inmunología , Ratones , Infecciones por Orthomyxoviridae/virología , Neumonía/inmunología , Neumonía/virología , Poli I-C/farmacología , Polilisina/análogos & derivados , Polilisina/farmacología , Infecciones por Virus Sincitial Respiratorio/virología , Virus Sincitiales Respiratorios/efectos de los fármacos , Virus Sincitiales Respiratorios/inmunología , Carga Viral/efectos de los fármacos
9.
BMC Pulm Med ; 9: 28, 2009 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-19515232

RESUMEN

BACKGROUND: Increased respiratory rate (tachypnea) is frequently observed as a clinical sign of influenza pneumonia in pediatric patients admitted to the hospital. We previously demonstrated that influenza infection of adult cotton rats (Sigmodon hispidus) also results in tachypnea and wanted to establish whether this clinical sign was observed in infected infant cotton rats. We hypothesized that age-dependent differences in lung mechanics result in differences in ventilatory characteristics following influenza infection. METHODS: Lung tidal volume, dynamic elastance, resistance, and pleural pressure were measured in a resistance and compliance system on mechanically-ventilated anesthestized young (14-28 day old) and adult (6-12 week old) cotton rats. Animals at the same age were infected with influenza virus, and breathing rates and other respiratory measurements were recorded using a whole body flow plethysmograph. RESULTS: Adult cotton rats had significantly greater tidal volume (TV), and lower resistance and elastance than young animals. To evaluate the impact of this increased lung capacity and stiffening on respiratory disease, young and adult animals were infected intra-nasally with influenza A/Wuhan/359/95. Both age groups had increased respiratory rate and enhanced pause (Penh) during infection, suggesting lower airway obstruction. However, in spite of significant tachypnea, the infant (unlike the adult) cotton rats maintained the same tidal volume, resulting in an increased minute volume. In addition, the parameters that contribute to Penh were different: while relaxation time between breaths and time of expiration was decreased in both age groups, a disproportionate increase in peak inspiratory and expiratory flow contributed to the increase in Penh in infant animals. CONCLUSION: While respiratory rate is increased in both adult and infant influenza-infected cotton rats, the volume of air exchanged per minute (minute volume) is increased in the infant animals only. This is likely to be a consequence of greater lung elastance in the very young animals. This model replicates many respiratory features of humans and consequently may be a useful tool to investigate new strategies to treat respiratory disease in influenza-infected infants.


Asunto(s)
Envejecimiento/fisiología , Resistencia de las Vías Respiratorias/fisiología , Hiperventilación/fisiopatología , Hiperventilación/virología , Virus de la Influenza A/patogenicidad , Presión Esfenoidal Pulmonar/fisiología , Sistema Respiratorio/fisiopatología , Animales , Animales Recién Nacidos , Modelos Animales de Enfermedad , Femenino , Masculino , Infecciones por Orthomyxoviridae/complicaciones , Ratas , Mecánica Respiratoria/fisiología , Sistema Respiratorio/patología , Sistema Respiratorio/virología , Sigmodontinae , Volumen de Ventilación Pulmonar/fisiología
10.
Virol J ; 6: 57, 2009 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-19442298

RESUMEN

BACKGROUND: Current small animal models for studying HIV-1 infection are very limited, and this continues to be a major obstacle for studying HIV-1 infection and pathogenesis, as well as for the urgent development and evaluation of effective anti-HIV-1 therapies and vaccines. Previously, it was shown that HIV-1 can infect cotton rats as indicated by development of antibodies against all major proteins of the virus, the detection of viral cDNA in spleen and brain of challenged animals, the transmission of infectious virus, albeit with low efficiency, from animal to animal by blood, and an additional increase in the mortality in the infected groups. RESULTS: Using in vitro experiments, we now show that cotton rat cell lines engineered to express human receptor complexes for HIV-1 (hCD4 along with hCXCR4 or hCCR5) support virus entry, viral cDNA integration, and the production of infectious virus. CONCLUSION: These results further suggest that the development of transgenic cotton rats expressing human HIV-1 receptors may prove to be useful small animal model for HIV infection.


Asunto(s)
Antígenos CD4/biosíntesis , VIH-1/fisiología , Receptores del VIH/biosíntesis , Proteínas Recombinantes/biosíntesis , Animales , Antígenos CD4/genética , Línea Celular , VIH-1/crecimiento & desarrollo , Humanos , Datos de Secuencia Molecular , Receptores del VIH/genética , Proteínas Recombinantes/genética , Análisis de Secuencia de ADN , Sigmodontinae , Internalización del Virus , Replicación Viral
11.
Biologicals ; 37(3): 152-9, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19394861

RESUMEN

Development of successful vaccines against human infectious diseases depends on using appropriate animal models for testing vaccine efficacy and safety. For some viral infections the task is further complicated by the frequently changing genetic make-up of the virus, as in the case of influenza, or by the existence of the little-understood phenomenon of vaccine-enhanced disease, as in the case of respiratory syncytial virus (RSV). The cotton rat Sigmodon hispidus has been used for years as an excellent small animal model of the RSV vaccine-enhanced disease. Recently, using cotton rats, we have demonstrated that vaccination against another paramyxovirus, human metapneumovirus (hMPV), can also lead to vaccine-enhanced disease. In addition to the study of paramyxoviruses, S. hispidus presents important advantages for the study of orthomyxoviruses such as influenza. The cotton rat is susceptible to infection with unadapted human influenza strains, and heterosubtypic immunity to influenza can be evoked in S. hispidus. The mechanisms of influenza, RSV, and hMPV pathogenesis and immunity can now be investigated in the cotton rat with the development of species-specific reagents for this animal model.


Asunto(s)
Modelos Animales de Enfermedad , Infecciones del Sistema Respiratorio/virología , Animales , Metapneumovirus/patogenicidad , Orthomyxoviridae/patogenicidad , Ratas , Virus Sincitiales Respiratorios/patogenicidad , Sigmodontinae
12.
J Virol ; 83(3): 1341-9, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19019962

RESUMEN

Human metapneumovirus (hMPV) is a recently described paramyxovirus that causes respiratory tract infections. Prior clinical studies have highlighted the importance of respiratory viruses, such as influenza virus, in facilitating secondary bacterial infections and increasing host immunopathology. The objective of the present work was to evaluate the effects of initial viral infection with hMPV or influenza A virus followed by Streptococcus pneumoniae superinfection 5 days later in a murine model. Both groups of superinfected mice demonstrated significant weight loss (mean of 15%) and higher levels of airway obstruction (mean enhanced pause value of 2.7) compared to those of mice infected with hMPV, influenza virus, or pneumococcus alone. Bacterial counts increased from 5 x 10(2) CFU/lung in mice infected with pneumococcus only to 10(7) and 10(9) CFU/lung in mice with prior infections with hMPV and influenza A virus, respectively. A more pronounced interstitial and alveolar inflammation correlated with higher levels of inflammatory cytokines and chemokines such as interleukin-1alpha (IL-1alpha), IL-1beta, IL-6, IL-12, monocyte chemotactic protein 1, macrophage inflammatory protein 1alpha, KC, and granulocyte colony-stimulating factor, as well as greater expression of Toll-like receptor 2 (TLR2), TLR6, TLR7, and TLR13 in the lungs of superinfected animals compared to results for single infections, with similar immunological effects seen in both coinfection models. Prior infection with either hMPV or influenza A virus predisposes mice to severe pneumococcus infection.


Asunto(s)
Metapneumovirus/patogenicidad , Neumonía Neumocócica/microbiología , Animales , Línea Celular , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Femenino , Pulmón/patología , Ratones , Ratones Endogámicos BALB C , Neumonía Neumocócica/fisiopatología , Neumonía Neumocócica/virología
13.
Virol J ; 5: 44, 2008 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-18355405

RESUMEN

BACKGROUND: Influenza virus infection or vaccination evokes an antibody response to viral hemagglutinin (HA) and neuraminidase (NA) surface glycoproteins, which results in immunity against influenza A viruses of the same HA and NA subtype. A heterosubtypic immune response that offers some protection against different influenza A subtypes has been suggested from epidemiologic studies in human influenza outbreaks, and has been induced in experimental animal models. Original studies of such cross-protection showed that cytotoxic T lymphocytes (CTL) protect H3N2-immune mice from a lethal H1N1 infection. More recent studies in mice demonstrate that antibodies also contribute to heterosubtypic immunity (HSI). We previously demonstrated that HSI in cotton rats (Sigmodon hispidus) is characterized by protection of H3N2-immune animals from influenza H1N1-induced increase in respiratory rate (tachypnea). Alternatively, H1N1-immune animals are protected from H3N2-induced tachypnea. The experiments described in this report were designed to elucidate the immune mechanism that prevents this very early sign of disease. RESULTS: Our results show that cotton rats provided with H1N1-immune serum prior to challenge with an H3N2 virus were protected from influenza-associated tachypnea, with the degree of protection correlating with the antibody titer transferred. Immunization with an inactivated preparation of virus delivered intramuscularly also provided some protection suggesting that CTL and/or mucosal antibody responses are not required for protection. Antibodies specific for conserved epitopes present on the virus exterior are likely to facilitate this protection since prophylactic treatment of cotton rats with anti-M2e (the extracellular domain of M2) but not anti-nucleoprotein (NP) reduced virus-induced tachypnea. CONCLUSION: In the cotton rat model of heterosubtypic immunity, humoral immunity plays a role in protecting animals from influenza-induced tachypea. Partial protection against respiratory disease caused by different influenza A subtypes can be attained with either live virus administered intranasally or inactivated virus delivered intramuscularly suggesting that either vaccine regimen may provide some protection against potential pandemic outbreaks in humans.


Asunto(s)
Anticuerpos Antivirales/inmunología , Reacciones Cruzadas , Subtipo H1N1 del Virus de la Influenza A/inmunología , Subtipo H3N2 del Virus de la Influenza A/inmunología , Infecciones por Orthomyxoviridae/fisiopatología , Trastornos Respiratorios/prevención & control , Sigmodontinae , Animales , Anticuerpos Antivirales/sangre , Especificidad de Anticuerpos , Modelos Animales de Enfermedad , Femenino , Humanos , Sueros Inmunes/inmunología , Subtipo H1N1 del Virus de la Influenza A/patogenicidad , Subtipo H3N2 del Virus de la Influenza A/patogenicidad , Vacunas contra la Influenza/administración & dosificación , Vacunas contra la Influenza/inmunología , Masculino , Pruebas de Neutralización , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/prevención & control , Infecciones por Orthomyxoviridae/virología , Trastornos Respiratorios/inmunología , Trastornos Respiratorios/fisiopatología , Trastornos Respiratorios/virología
14.
J Gen Virol ; 89(Pt 1): 261-270, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18089750

RESUMEN

Respiratory syncytial virus (RSV) is the primary cause of bronchiolitis in young children. In general, RSV is considered to be a poor inducer of type I (alpha/beta) interferons (IFNs). Measurement of active type I IFN production during infection in vivo is demanding, as multiple IFN subtypes with overlapping activities are produced. In contrast, Mx gene expression, which is tightly regulated by type I IFN expression, is easily determined. This study therefore measured Mx expression as a reliable surrogate marker of type I IFN activity during RSV infection in vivo in a cotton rat model. It was shown that expression of Mx genes was dramatically augmented in the lungs of infected animals in a dose- and virus strain-dependent manner. The expression of Mx genes in the lungs was paralleled by their induction in the nose and spleen, although in spleen no simultaneous virus gene expression was detected. Reinfection of RSV-immune animals leads to abortive virus replication in the lungs. Thus, type I IFN and Mx gene expression was triggered in reinfected animals, even though virus could not be isolated from their lungs. Furthermore, it was demonstrated that immunity to RSV wanes with time. Virus replication and Mx gene expression became more prominent with increasing intervals between primary infection and reinfection. These results highlight the role of type I IFN in modulation of the immune response to RSV.


Asunto(s)
Proteínas de Unión al GTP/genética , Regulación de la Expresión Génica , Interferón Tipo I/biosíntesis , Infecciones por Virus Sincitial Respiratorio/inmunología , Virus Sincitiales Respiratorios/fisiología , Animales , Pulmón/fisiopatología , Pulmón/virología , Proteínas de Resistencia a Mixovirus , Especificidad de Órganos , ARN Mensajero/genética , Recurrencia , Infecciones por Virus Sincitial Respiratorio/fisiopatología , Virus Sincitiales Respiratorios/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sigmodontinae , Replicación Viral
15.
Antimicrob Agents Chemother ; 52(1): 279-87, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17967906

RESUMEN

Human metapneumovirus (hMPV) can cause acute upper and lower respiratory tract infections that are particularly severe in young children, elderly subjects, and immunocompromised patients. To date, no treatments or vaccines are available for hMPV infections. Our objective was to assess the inhibitory potential of several peptides derived from the heptad repeat A and B (HRA and HRB) domains of the hMPV fusion protein. Nine candidate peptides were expressed in Escherichia coli or obtained synthetically and tested in vitro and in an animal model. Excellent in vitro inhibition of an hMPV strain of the A1 subgroup was obtained with five peptides, with 50% inhibitory concentrations ranging from 1.4 nM to 3.3 microM. One peptide, HRA2, displayed very potent activity against all four hMPV subgroups. It was also moderately active against human respiratory syncytial virus (strain A2) but displayed no activity against human parainfluenza virus type 3. BALB/c mice that received the HRA2 peptide and a lethal hMPV intranasal challenge simultaneously were completely protected from clinical symptoms and mortality. On day 5 postinfection, HRA2-treated mice had undetectable lung viral loads which were significantly less than those of untreated mice (3 x 10(4) 50% tissue culture infective doses/lung). Pulmonary inflammation, levels of proinflammatory cytokines/chemokines (RANTES, gamma interferon, and monocyte chemoattractant protein 1) and airway obstruction were also significantly decreased in HRA2-treated mice. The results of this study demonstrate that potent antivirals can be derived from the hMPV fusion protein HR domains. Moreover, hMPV, compared to other paramyxoviruses and to the human immunodeficiency virus, seems to be more susceptible to HRA- than HRB-derived peptides.


Asunto(s)
Antivirales/farmacología , Fusión de Membrana/efectos de los fármacos , Metapneumovirus/efectos de los fármacos , Infecciones por Paramyxoviridae/tratamiento farmacológico , Péptidos/farmacología , Proteínas Virales de Fusión/química , Secuencia de Aminoácidos , Animales , Antivirales/química , Antivirales/uso terapéutico , Línea Celular , Línea Celular Tumoral , Femenino , Humanos , Concentración 50 Inhibidora , Metapneumovirus/patogenicidad , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Infecciones por Paramyxoviridae/virología , Péptidos/química , Péptidos/uso terapéutico
16.
Virology ; 369(1): 131-42, 2007 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-17727912

RESUMEN

Oncolytic adenoviruses as a treatment for cancer have demonstrated limited clinical activity. Contributing to this may be the relevance of preclinical animal models used to study these agents. Syngeneic mouse tumor models are generally non-permissive for adenoviral replication, whereas human tumor xenograft models exhibit attenuated immune responses to the vector. The cotton rat (Sigmodon hispidus) is susceptible to human adenovirus infection, permissive for viral replication and exhibits similar inflammatory pathology to humans with adenovirus replicating in the lungs, respiratory passages and cornea. We evaluated three transplantable tumorigenic cotton rat cell lines, CCRT, LCRT and VCRT as models for the study of oncolytic adenoviruses. All three cells lines were readily infected with adenovirus type-5-based vectors and exhibited high levels of transgene expression. The cell lines supported viral replication demonstrated by the induction of cytopathogenic effect (CPE) in tissue culture, increase in virus particle numbers and assembly of virions seen on transmission electron microscopy. In vivo, LCRT and VCRT tumors demonstrated delayed growth after injection with replicating adenovirus. No in vivo antitumor activity was seen in CCRT tumors despite in vitro oncolysis. Adenovirus was also rapidly cleared from the CCRT tumors compared to LCRT and VCRT tumors. The effect observed with the different cotton rat tumor cell lines mimics the variable results of human clinical trials highlighting the potential relevance of this model for assessing the activity and toxicity of oncolytic adenoviruses.


Asunto(s)
Adenoviridae/crecimiento & desarrollo , Modelos Animales de Enfermedad , Neoplasias/terapia , Neoplasias/virología , Viroterapia Oncolítica/métodos , Animales , Línea Celular Tumoral , Efecto Citopatogénico Viral , Microscopía Electrónica de Transmisión , Ratas , Sigmodontinae , Trasplante Isogénico , Ensayo de Placa Viral , Virión/ultraestructura
17.
Microb Pathog ; 43(5-6): 208-16, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17689046

RESUMEN

Bacterial super-infection of influenza patients is the primary cause of excess mortality during influenza pandemics, with Staphylococcus aureus (S. aureus) having the highest fatality rate. The cotton rat (Sigmodon hispidus) is an excellent model for both influenza and S. aureus pathogenesis, and therefore a potential tool to model co-infection. We compared physiologic and pathologic changes in cotton rats infected with both S. aureus and influenza A/Wuhan/359/95 (H3N2), with animals infected with each pathogen alone. Co-infected cotton rats demonstrated significantly higher mortality, lower temperatures on 2 and 3 days post-inoculation (p.i.), higher levels of bacteremia and pulmonary bacterial load 4 days p.i., and worse pathology 7 days p.i. Early indicators of exacerbated disease coincided with higher pulmonary mRNA levels for IL-1beta, IL-6, IL-10 and IFNy, supporting the idea that these may contribute to disease severity. Our results demonstrate that the cotton rat is a good model of influenza and S. aureus co-infection, with increased mortality and hypothermia as well as prolonged bacterial duration indicative of synergistic disease that may be the result of increased induction of both pro- and anti-inflammatory cytokines.


Asunto(s)
Subtipo H3N2 del Virus de la Influenza A , Infecciones por Orthomyxoviridae/complicaciones , Infecciones Estafilocócicas/complicaciones , Staphylococcus aureus/aislamiento & purificación , Animales , Anticuerpos Antivirales/sangre , Modelos Animales de Enfermedad , Subtipo H3N2 del Virus de la Influenza A/aislamiento & purificación , Pulmón/patología , Pulmón/virología , Sigmodontinae
18.
J Virol ; 81(17): 9443-50, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17596309

RESUMEN

Respiratory syncytial virus (RSV) is a major cause of bronchiolitis and viral pneumonia in young children and a serious health risk in immunocompromised individuals and the elderly. Immunity to RSV is not completely understood. In this work, we established a method for monitoring RSV infection by real-time PCR and applied this method for analysis of RSV replication in vivo in the cotton rat model in naïve animals and in animals rendered immune to RSV by prior RSV infection. We found that even though no virus could be isolated from the lungs of RSV-challenged immune animals, RSV infection in fact took place and an accumulation of viral RNA transcripts was observed. This type of replication, therefore, can be termed "abortive," as RSV is capable of entering the cells in the lungs of immune animals, yet the production of progeny viruses is impaired. Similar patterns of RSV gene expression gradient were observed between naïve and reinfected animals, indicating that the skewing of mRNA gradient of viral gene expression, a mechanism documented during latent infection by other viruses, is not likely to be responsible for abortive replication of RSV during reinfection. We found that passive administration of antibodies to RSV prevents productive infection normally accompanied by viral release in the lung, but it does not prevent abortive replication of the virus. To the best of our knowledge, this is the first evidence of abortive replication of RSV in vivo.


Asunto(s)
Pulmón/virología , Infecciones por Virus Sincitial Respiratorio/inmunología , Infecciones por Virus Sincitial Respiratorio/virología , Virus Sincitiales Respiratorios/crecimiento & desarrollo , Virus Sincitiales Respiratorios/inmunología , Animales , Línea Celular , ADN Viral/análisis , Modelos Animales de Enfermedad , Humanos , Reacción en Cadena de la Polimerasa/métodos , ARN Mensajero/análisis , ARN Viral/análisis , Virus Sincitiales Respiratorios/genética , Virus Sincitiales Respiratorios/aislamiento & purificación , Sigmodontinae , Carga Viral , Ensayo de Placa Viral
19.
Vaccine ; 25(27): 5034-40, 2007 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-17543425

RESUMEN

Cotton rats (Sigmodon hispidus) are susceptible to the recently discovered human metapneumovirus (hMPV), an agent closely related to human respiratory syncytial virus. Since certain respiratory syncytial virus vaccines can induce enhanced disease upon viral challenge, we have done similar experiments with hMPV in cotton rats. Young adult cotton rats were vaccinated with a formalin-inactivated preparation of hMPV strain C-85473, or with a mock preparation of the vaccine on day 0 and again on day 28. All animals were challenged intranasally on day 49 with 10(7) TCID50 of the same hMPV strain. Animals were sacrificed on days 4, 7, and 10 post-challenge and lungs were removed for viral quantitation, histopathology, and cytokine mRNA expression analysis (interferon-gamma (IFN-gamma) and interleukin-4 (IL-4)). Although the vaccinated animals showed almost complete protection from viral replication in the lungs (<10(2.0) TCID50 per gram), there was a dramatic increase in the lung pathology, particularly the interstitial pneumonitis and alveolitis with elevated serum neutralizing antibody titer prior to challenge. Cytokine profiles were distinctive from those observed during primary infection and re-infection. The data raise safety concerns for hMPV vaccine preparations.


Asunto(s)
Enfermedades Pulmonares/etiología , Enfermedades Pulmonares/patología , Metapneumovirus/inmunología , Infecciones por Paramyxoviridae/inmunología , Infecciones por Paramyxoviridae/prevención & control , Vacunas Virales/efectos adversos , Vacunas Virales/inmunología , Animales , Citocinas/metabolismo , Inmunización , Interferón gamma/biosíntesis , Interleucina-4/biosíntesis , Pulmón/metabolismo , Pulmón/patología , Enfermedades Pulmonares/metabolismo , Pruebas de Neutralización , Infecciones por Paramyxoviridae/virología , Ratas , Sigmodontinae , Vacunas de Productos Inactivados/efectos adversos , Replicación Viral
20.
J Infect Dis ; 195(4): 511-8, 2007 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-17230410

RESUMEN

BACKGROUND: Respiratory syncytial virus (RSV) is the major cause of severe lower respiratory tract infection in infants and young children. Recently, RSV has also been recognized as a serious health risk in elderly individuals, but the pathogenesis of RSV infection in elderly individuals remains unknown. METHODS: Dynamics of pulmonary cytokine response (including interferon- gamma , interleukin [IL]-4, IL-10, IL-6, monocyte chemoattractant protein-1, and growth-regulated oncogene [GRO] mRNA) during acute RSV infection were investigated in young (<2 months old) and aged (>9 months old) cotton rats (Sigmodon hispidus). Therapeutic treatments that diminish viral replication (antiviral antibody) and pulmonary inflammation (anti-inflammatory corticosteroid) in RSV-infected cotton rats were used to evaluate the contribution of virus replication and inflammation to the development of RSV disease with respect to age. RESULTS: The time of the peak expression of the majority of cytokines was shifted with respect to age. Antiviral and anti-inflammatory treatments had a similar effect on cytokine expression in aged and young cotton rats. GRO mRNA transcripts were more abundant in the lungs of aged cotton rats. CONCLUSIONS: The present study reports an age-related delay in the pulmonary cytokine response to RSV and an imbalance in chemokine production with respect to age and underscores different components of RSV pathogenesis with respect to their molecular signature.


Asunto(s)
Envejecimiento/inmunología , Antiinflamatorios/uso terapéutico , Antivirales/uso terapéutico , Citocinas/biosíntesis , Pulmón/inmunología , Infecciones por Virus Sincitial Respiratorio/inmunología , Animales , Citocinas/genética , Modelos Animales de Enfermedad , Expresión Génica , ARN Mensajero/análisis , ARN Mensajero/genética , Ratas , Infecciones por Virus Sincitial Respiratorio/tratamiento farmacológico , Infecciones por Virus Sincitial Respiratorio/patología , Virus Sincitiales Respiratorios/inmunología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sigmodontinae
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...