Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 86
Filtrar
1.
Mucosal Immunol ; 3(3): 291-300, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20404812

RESUMEN

Severe respiratory syncytial virus (RSV)-induced bronchiolitis has been associated with a mixed "Th1" and "Th2" cytokine storm. We hypothesized that differentiation of "alternatively activated" macrophages (AA-M phi) would mediate the resolution of RSV-induced lung injury. RSV induced interleukin (IL)-4 and IL-13 by murine lung and peritoneal macrophages, IL-4R alpha/STAT6-dependent AA-M phi differentiation, and significantly enhanced inflammation in the lungs of IL-4R alpha(-/-) mice. Adoptive transfer of wildtype macrophages to IL-4R alpha(-/-) mice restored RSV-inducible AA-M phi phenotype and diminished lung pathology. RSV-infected Toll-like receptor (TLR)4(-/-) and interferon (IFN)-beta(-/-) macrophages and mice also failed to express AA-M phi markers, but exhibited sustained proinflammatory cytokine production (e.g., IL-12) in vitro and in vivo and epithelial damage in vivo. TLR4 signaling is required for peroxisome proliferator-activated receptor gamma expression, a DNA-binding protein that induces AA-M phi genes, whereas IFN-beta regulates IL-4, IL-13, IL-4R alpha, and IL-10 expression in response to RSV. RSV-infected cotton rats treated with a cyclooxygenase-2 inhibitor increased expression of lung AA-M phi. These data suggest new treatment strategies for RSV that promote AA-M phi differentiation.


Asunto(s)
Interferón beta/inmunología , Lesión Pulmonar/inmunología , Activación de Macrófagos/inmunología , Macrófagos Alveolares/inmunología , Macrófagos Peritoneales/inmunología , Receptores de Superficie Celular/inmunología , Infecciones por Virus Sincitial Respiratorio/inmunología , Virus Sincitiales Respiratorios/inmunología , Receptor Toll-Like 4/inmunología , Animales , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Inhibidores de la Ciclooxigenasa 2/farmacología , Citocinas/genética , Citocinas/inmunología , Citocinas/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Regulación de la Expresión Génica/inmunología , Humanos , Interferón beta/genética , Interferón beta/metabolismo , Lesión Pulmonar/metabolismo , Lesión Pulmonar/virología , Activación de Macrófagos/efectos de los fármacos , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/virología , Macrófagos Peritoneales/metabolismo , Macrófagos Peritoneales/virología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Neumonía/inmunología , Neumonía/metabolismo , Neumonía/virología , Ratas , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Infecciones por Virus Sincitial Respiratorio/genética , Infecciones por Virus Sincitial Respiratorio/metabolismo , Infecciones por Virus Sincitial Respiratorio/virología , Virus Sincitiales Respiratorios/metabolismo , Sigmodontinae , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo
2.
Clin Exp Immunol ; 148(2): 218-29, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17335559

RESUMEN

Epidemiological studies show an association between early exposure to respiratory syncytial virus (RSV) and the development or exacerbation of asthma. This idea is supported by studies in mice that demonstrate worsened airway hyper-reactivity (AHR) when RSV-infected animals are exposed to allergen. The effect of allergen on RSV disease, however, has not been reported. Cotton rats (Sigmodon hispidus) that have been used as a model to study RSV pathogenesis were sensitized to extracts of Aspergillus fumigatus (Af), a common household mould. The allergic response to Af included eosinophilia, formation of granulomas and induction of Th2 type cytokines. RSV infection prior to allergen challenge resulted in exacerbation of the inflammatory response as well as increased airway responsiveness to methacholine. The exacerbated response was indeed dependent on virus replication. Virus replication in turn was influenced by the allergic response, with persistence in the noses for 2 days longer in animals challenged with allergen. This diminished clearance corresponded to decreased induction of mRNA for IFN-gamma, a Th1-type cytokine that is characteristic of viral infection. Treatment of RSV-infected Af-challenged animals with recombinant IFN-gamma reduced the allergic inflammatory response as well as the relative levels of Th1 and Th2 cytokine mRNA. However, this treatment did not reduce airway reactivity, showing that these pathologic and physiologic measures of exacerbated disease are independent. We speculate that the reciprocal effect of the allergic response on viral immunity may benefit the host by limiting exacerbation of physiologic responses that are IFN-gamma-dependent.


Asunto(s)
Alérgenos/inmunología , Hipersensibilidad Respiratoria/virología , Infecciones por Virus Sincitial Respiratorio/virología , Virus Sincitiales Respiratorios/fisiología , Infecciones del Sistema Respiratorio/virología , Animales , Antivirales/uso terapéutico , Aspergillus fumigatus/inmunología , Citocinas/biosíntesis , Citocinas/genética , Femenino , Granuloma/inmunología , Granuloma/patología , Granuloma/virología , Interferón gamma/uso terapéutico , Masculino , ARN Mensajero/genética , Proteínas Recombinantes , Hipersensibilidad Respiratoria/inmunología , Hipersensibilidad Respiratoria/patología , Infecciones por Virus Sincitial Respiratorio/tratamiento farmacológico , Infecciones por Virus Sincitial Respiratorio/inmunología , Infecciones por Virus Sincitial Respiratorio/patología , Infecciones del Sistema Respiratorio/tratamiento farmacológico , Infecciones del Sistema Respiratorio/inmunología , Infecciones del Sistema Respiratorio/patología , Sigmodontinae , Células Th2/inmunología , Replicación Viral/inmunología
3.
Bone Marrow Transplant ; 29(2): 117-20, 2002 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11850705

RESUMEN

Respiratory syncytial virus (RSV) is widely recognized as a leading cause of pneumonia, with substantial mortality, in bone marrow transplant recipients. We tested the efficacy of a systemic monoclonal antibody (MAB) preparation possessing a high titer of anti-RSV neutralizing antibody, palivizumab (Synagis) for prophylaxis and therapy of RSV infection in cytoxan (CY) immunosuppressed cotton rats, a model in which the efficacy of a polyclonal anti-RSV product (Respigam) has been demonstrated. Both prophylaxis and therapy with this MAB were highly effective in reducing pulmonary viral replication. However, multiple sequential therapeutic doses of MAB were necessary to control rebound viral replication in continually suppressed animals.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antivirales/uso terapéutico , Huésped Inmunocomprometido , Infecciones por Virus Sincitial Respiratorio/tratamiento farmacológico , Infecciones por Virus Sincitial Respiratorio/inmunología , Animales , Anticuerpos Monoclonales Humanizados , Evaluación Preclínica de Medicamentos , Modelos Animales , Palivizumab , Ratas , Ratas Endogámicas , Infecciones por Virus Sincitial Respiratorio/prevención & control , Virus Sincitiales Respiratorios/efectos de los fármacos , Virus Sincitiales Respiratorios/crecimiento & desarrollo , Trasplante de Células Madre/efectos adversos , Resultado del Tratamiento , Replicación Viral/efectos de los fármacos
4.
Arch Virol ; 147(1): 167-79, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-11855629

RESUMEN

The majority of the human population has been infected with herpes simplex virus type 1 (HSV-1). During a typical primary episode, HSV-1 spreads from the oral pharynx to the trigeminal ganglia, where a latent HSV-1 infection is established. Cold sores at the mucocutaneous junction of the lip are the typical manifestation of recurrent HSV-1. We investigated whether HSV-1 also infects the brain during the primary infection. We used HSV-1 infected BALB/c mice and inbred cotton rats as models. While both species were susceptible to HSV-1 infection, the time course of lesion formation and healing in the cotton rat more closely reflected that seen in humans. In both species, HSV-1 replicated in the brainstem and cerebellum, as well as the trigeminal ganglia during a primary infection of the lip. The brain infection was produced by a low inoculation dose, and did not cause observable neurologic signs or mortality. Using PCR and RT-PCR techniques, we demonstrated HSV-1 thymidine kinase in the absence of infectivity in the brains of both species 30-40 days after primary infection.


Asunto(s)
Encéfalo/virología , Herpesvirus Humano 1/fisiología , Enfermedades de los Labios/virología , Ganglio del Trigémino/virología , Latencia del Virus , Animales , Modelos Animales de Enfermedad , Herpes Simple/virología , Herpesvirus Humano 1/aislamiento & purificación , Herpesvirus Humano 1/patogenicidad , Humanos , Ratones , Ratones Endogámicos BALB C , Reacción en Cadena de la Polimerasa , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sigmodontinae
5.
J Virol ; 75(24): 12446-51, 2001 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11711635

RESUMEN

The envelope of human parainfluenza virus type 3 (HPF3) contains two viral glycoproteins, the hemagglutinin-neuraminidase (HN) protein and the fusion (F) protein. In a previous study, highly fusogenic variant HPF3 viruses were isolated, including two, C-0 and C-22, that exhibit increased avidity for sialic acid receptors due to single amino acid changes in the HN protein and one, C-28, that has decreased neuraminidase activity relative to that of the wild type (wt) and is delayed in the release of virus particles into the supernatant fluid. These variants form very large plaques and destroy a cell monolayer more rapidly than does wt HPF3 in cell culture. These variant viruses allowed us to formulate hypotheses about the roles of HN in pathogenesis. We investigated the behavior of wt HPF3 and the three variant viruses in the cotton rat model. In the cotton rat, there was no delayed clearance of any of the variant viruses compared to that of the wt. The variant plaque morphology was preserved in vivo, and there was no reversion to the wt phenotype in the infected animals. In spite of a slight advantage of wt virus in viral titer, there were no differences in the severities of peribronchiolitis between wt viruses and the variants. However, there were marked differences in severities in alveolitis and interstitial pneumonitis when each of the three variants was compared to the wt, with the variants causing enhanced disease. Thus, despite similar or lower viral titers and similar clearance rates, the variants caused more extensive disease in the lung. The results show that mutations in HN conferring altered fusion properties in cell culture also confer striking differences in the ability of HPF3 to cause extensive disease in the cotton rat lung and that this effect is dissociated from any effect on viral replication.


Asunto(s)
Virus de la Parainfluenza 3 Humana/fisiología , Receptores Virales/fisiología , Infecciones por Respirovirus/etiología , Animales , Guanidinas , Pulmón/patología , Pulmón/virología , Neuraminidasa/metabolismo , Nariz/patología , Nariz/virología , Piranos , Ácidos Siálicos/farmacología , Sigmodontinae , Replicación Viral , Zanamivir
6.
Vaccine ; 19(15-16): 2048-54, 2001 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-11228376

RESUMEN

The mechanisms by which administration of a formalin-inactivated respiratory syncytial virus vaccine resulted in enhanced disease among children after they later became naturally infected with the virus remains largely undefined. After immunization and live virus challenge, the cotton rat demonstrated the histopathologic marker of the enhanced disease, polymorphonuclear leukocyte infiltration of lung alveolar spaces. We now report that immunization with formalin-inactivated vaccine formulated with the adjuvant, 3-deacylated monophosphoryl lipid A, dramatically reduces or eliminates the polymorphonuclear leukocyte infiltration within the alveoli of cotton rats post-challenge. We suggest, that this or similar adjuvants may be beneficial components of candidate non-replicating respiratory syncytial virus vaccines, whose development has been hampered by safety concerns.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Lípido A/administración & dosificación , Virus Sincitiales Respiratorios/inmunología , Vacunas Virales/efectos adversos , Vacunas Virales/toxicidad , Animales , Niño , Femenino , Formaldehído , Humanos , Lípido A/análogos & derivados , Masculino , Alveolos Pulmonares/patología , Infecciones por Virus Sincitial Respiratorio/inmunología , Infecciones por Virus Sincitial Respiratorio/patología , Infecciones por Virus Sincitial Respiratorio/prevención & control , Seguridad , Sigmodontinae , Vacunas de Productos Inactivados/efectos adversos , Vacunas de Productos Inactivados/toxicidad
7.
Expert Opin Investig Drugs ; 10(2): 297-308, 2001 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11178342

RESUMEN

Respiratory syncytial virus (RSV), the most common cause of lower respiratory tract disease in infants and young children, is a ubiquitous respiratory pathogen, infecting or reinfecting much of the population every year and causing severe, sometimes fatal disease in high-risk populations of infants and adults, particularly in developing countries. Spurred by the medical and economic burdens of RSV disease and enticed by the economic potential of therapeutic drugs, particularly in the absence to date of a safe and effective RSV vaccine, scientists in many industrial, academic and government laboratories have developed a wide variety of candidate RSV antiviral agents. Most of these have been screened thus far only in cell culture, a few in animal models. Aside from ribavirin, however, none has proven effective in therapeutic clinical trials and even ribavirin usage has declined precipitously in recent years due to concerns over efficacy, safety, ease of use and cost. All of the antiviral compounds discussed in this review were evaluated primarily for their ability to reduce viral load, with little or no attention paid to the role of host inflammation in the pathogenesis of RSV disease. Recent research has highlighted the prominent role of inflammatory mediators and an increasing number of reports suggest that a therapeutic strategy that combines antiviral and anti-inflammatory components will be the most effective way of treating RSV disease.


Asunto(s)
Antivirales/farmacología , Virus Sincitiales Respiratorios/efectos de los fármacos , Oligonucleótidos Antisentido/farmacología , ARN Viral/biosíntesis , Virus Sincitiales Respiratorios/fisiología
8.
J Virol ; 74(22): 10287-92, 2000 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11044072

RESUMEN

Several formulations of a recombinant chimeric respiratory syncytial virus (RSV) vaccine consisting of the extramembrane domains of the F and G glycoproteins (FG) were tested in cotton rats to evaluate efficacy and safety. The FG vaccine was highly immunogenic, providing nearly complete resistance to pulmonary infection at doses as low as 25 ng in spite of inducing relatively low levels of serum neutralizing antibody at low vaccine doses. Upon RSV challenge animals primed with FG vaccine showed quite mild alveolitis and interstitial pneumonitis, which were eliminated by the addition of monophosphoryl lipid A to the formulation.


Asunto(s)
Infecciones por Virus Sincitial Respiratorio/prevención & control , Vacunas contra Virus Sincitial Respiratorio , Virus Sincitiales Respiratorios/inmunología , Proteínas Virales/inmunología , Animales , Anticuerpos Antivirales/sangre , Pulmón/patología , Pulmón/virología , Enfermedades Pulmonares Intersticiales/prevención & control , Pruebas de Neutralización , Neumonía Viral/prevención & control , Proteínas Recombinantes de Fusión/inmunología , Vacunas contra Virus Sincitial Respiratorio/administración & dosificación , Vacunas contra Virus Sincitial Respiratorio/inmunología , Virus Sincitiales Respiratorios/aislamiento & purificación , Sigmodontinae , Vacunación , Proteínas Virales/genética
9.
Viral Immunol ; 13(2): 231-6, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-10893002

RESUMEN

Vaccine-induced potentiation was studied in cotton rats immunized with formalin-inactivated human parainfluenza type 3, ultraviolet light-inactivated virus, or infection with live virus. Immunized animals and unimmunized controls were later challenged by intranasal inoculation of live virus and evaluated for pulmonary pathology 4 days later. Animals immunized with either of the inactivated vaccines developed marked peribronchiolitis, perivasculitis, and an alveolar cellular infiltration much more severe than seen in animals infected previously, or in unmanipulated but challenged animals. Disease enhancement after immunization with killed virus is thus a characteristic of a member of each of three genera of the family, Paramyxoviridae, and is not restricted to immunization with formalin-inactivated virus.


Asunto(s)
Pulmón/patología , Vacunas contra la Parainfluenza/inmunología , Virus de la Parainfluenza 3 Humana/inmunología , Infecciones por Respirovirus/patología , Animales , Modelos Animales de Enfermedad , Formaldehído , Humanos , Infecciones por Respirovirus/inmunología , Infecciones por Respirovirus/virología , Sigmodontinae , Vacunación , Vacunas de Productos Inactivados/inmunología
10.
Lab Invest ; 79(11): 1385-92, 1999 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-10576209

RESUMEN

Infection of the cotton rat lung with a human strain of respiratory syncytial virus results in substantial virus replication and is associated with mild-to-moderate peribronchiolitis, perivasculitis, and bronchitis. Reinfection after 49 days did not result in detectable virus replication, but surprisingly, was associated with an earlier appearance and accentuation of the three types of lesions seen in cotton rats undergoing primary infection. Animals primed with formalin-inactivated virus and challenged after 49 days had pulmonary viral titers 1/10 to 1/100 of that seen in naive animals, but developed markedly accentuated lesions of the same type as in animals undergoing primary or secondary infection. In addition, the animals with the vaccine-enhanced disease developed alveolitis and interstitial pneumonitis, which seem to be specific markers for the vaccine enhancement. These latter markers may be useful in determining the safety of nonreplicating vaccines.


Asunto(s)
Pulmón/efectos de los fármacos , Pulmón/patología , Infecciones por Virus Sincitial Respiratorio/patología , Vacunas Virales/farmacología , Animales , Bronquiolitis/patología , Bronquiolitis/virología , Bronquitis/patología , Bronquitis/virología , Femenino , Humanos , Pulmón/virología , Enfermedades Pulmonares Intersticiales/patología , Enfermedades Pulmonares Intersticiales/virología , Masculino , Alveolos Pulmonares/patología , Ratas , Recurrencia , Virus Sincitiales Respiratorios/aislamiento & purificación , Virus Sincitiales Respiratorios/fisiología , Sigmodontinae , Vacunas de Productos Inactivados/farmacología , Replicación Viral
11.
Proc Natl Acad Sci U S A ; 96(18): 10409-11, 1999 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-10468621

RESUMEN

Comparison of the inflammatory response of Sigmodon hispidus cotton rats to pulmonary infection with wild-type 5 adenovirus (Ad5) or with a viral mutant, in which the early region 1B gene encoding a 55-kDa protein, Ad5dl110 (dl110), was deleted, indicated that the inflammation in animals infected with dl110 was markedly reduced compared with the inflammation in animals infected with wild-type Ad5, although both viruses replicated to the same extent. Comparable experiments done with C57BL/6 mice yielded identical results, even though only the early phase of gene expression essential for viral replication occurs in mice. Cytokine analysis of infected mouse lungs indicated that tumor necrosis factor-alpha and IL-6 were produced in relatively large quantities in wild-type Ad5-infected mice and at significantly lower levels in dl110-infected mice during the early stages of infection.


Asunto(s)
Infecciones por Adenoviridae/patología , Proteínas E1B de Adenovirus/genética , Adenovirus Humanos/genética , Adenovirus Humanos/patogenicidad , Pulmón/patología , Infecciones por Adenoviridae/inmunología , Adenovirus Humanos/fisiología , Animales , Línea Celular , Citocinas/análisis , Eliminación de Gen , Humanos , Inflamación , Células KB , Pulmón/inmunología , Pulmón/virología , Ratones , Ratones Endogámicos C57BL , Sigmodontinae , Replicación Viral
12.
Bone Marrow Transplant ; 24(1): 41-5, 1999 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-10435733

RESUMEN

Respiratory syncytial virus (RSV) has emerged as a leading cause of pneumonia, with high mortality, in bone marrow transplant (BMT) recipients, as well as in other profoundly immunocompromised patients, such as myelosuppressed adults with leukemia. We tested the efficacy of immunoglobulin with high anti-RSV neutralizing antibody levels (RSVIG) for prophylaxis and therapy of RSV infection in cotton rats undergoing prolonged immunosuppression with cyclophosphamide. These animals experience persistent infection, a model which is similar to the disease seen in post-BMT humans. Both prophylaxis and therapy reduced pulmonary viral replication over 500-fold to nearly undetectable levels. In animals receiving continual immunosuppression, the use of multiple therapeutic doses of RSVIG was able to prevent rebound viral replication, though virus was not completely eliminated.


Asunto(s)
Anticuerpos Antivirales/uso terapéutico , Inmunoglobulinas Intravenosas/uso terapéutico , Inmunosupresores/uso terapéutico , Infecciones por Virus Sincitial Respiratorio/prevención & control , Virus Sincitiales Respiratorios/inmunología , Animales , Ciclofosfamida/farmacología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta Inmunológica , Inmunoglobulinas Intravenosas/efectos adversos , Inmunosupresores/efectos adversos , Pulmón/virología , Nariz/virología , Ratas , Ratas Endogámicas , Virus Sincitiales Respiratorios/crecimiento & desarrollo , Sigmodontinae , Replicación Viral
13.
J Infect Dis ; 180(1): 35-40, 1999 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-10353858

RESUMEN

Two humanized monoclonal antibodies, MEDI-493 and RSHZ19, were developed independently as potential improvements over RSV-IGIV for prevention of respiratory syncytial virus (RSV) infection. RSV-IGIV is a polyclonal human antibody preparation for intravenous infusion enriched for RSV neutralizing activity. A phase III clinical trial showed that MEDI-493 significantly reduced hospitalizations due to RSV infection. In a separate trial, RSHZ19 failed to show significant efficacy. In new studies, the in vitro and in vivo activities of MEDI-493 and RSHZ19 were compared to determine whether the different clinical results are related to differences in biologic activity. MEDI-493 was consistently 4- to 5-fold more potent than RSHZ19 in antigen binding, RSV neutralization, and fusion inhibition assays. Although both MEDI-493 and RSHZ19 were effective against A and B subtypes of RSV in the cotton rat model of RSV infection, 2- to 4-fold higher doses of RSHZ19 were required for similar protection. The enhanced activity of MEDI-493 compared with RSHZ19 may, in part, explain its better clinical effect.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Proteína HN , Infecciones por Virus Sincitial Respiratorio/tratamiento farmacológico , Virus Sincitiales Respiratorios/inmunología , Animales , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Monoclonales Humanizados , Afinidad de Anticuerpos , Fusión Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Inmunoglobulina G/inmunología , Inmunoterapia , Pruebas de Neutralización , Palivizumab , Sigmodontinae , Proteínas del Envoltorio Viral , Proteínas Virales de Fusión/inmunología , Proteínas Virales/inmunología
14.
Infect Control Hosp Epidemiol ; 20(1): 57-8, 1999 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-9927268

RESUMEN

In an in-vitro test, generic liquid hand dishwashing detergents were as much as 100-fold more effective than proprietary antibacterial soaps in inactivating respiratory syncytial virus (RSV). The use of such detergents for hand washing during annual RSV epidemics, or the incorporation of their antiviral components into antibacterial soaps might be considered to limit nosocomial spread.


Asunto(s)
Detergentes/farmacología , Desinfección de las Manos , Infecciones por Virus Sincitial Respiratorio/prevención & control , Virus Sincitial Respiratorio Humano/patogenicidad , Infección Hospitalaria/prevención & control , Humanos
15.
Proc Natl Acad Sci U S A ; 95(24): 14355-60, 1998 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-9826704

RESUMEN

Cotton rats (Sigmodon hispidus and S. fulviventer) are susceptible to many viruses that infect humans (e.g., poliovirus, respiratory syncytial virus, influenza virus, adenovirus, and parainfluenza virus) and have been influential in developing therapeutic clinical intervention strategies for many viral infections of man. This study set out to determine whether cotton rats are susceptible to infection with HIV type 1 (HIV-1). Results indicate that HIV-1 does infect the cotton rat and S. fulviventer is more susceptible than S. hispidus. The virus was passaged from animal to animal for a total of three serial passages; but HIV replicated poorly in vivo, was only detectable as proviral DNA, and never exceeded one provirus per 1.8 x 10(5) cotton rat peripheral blood mononuclear cells. Infection induced a distinct and characteristic anti-HIV antibody response that, in some animals, included neutralizing antibodies, recognized all of the major HIV-1 antigens and the antibodies lasted out to 52 wk post-infection. Neonate S. fulviventer were not more susceptible to infection than adults. In vitro culture studies produced indirect evidence of viral replication by detection of viral gag gene RNA in reverse transcriptase-PCR assays on viral culture supernatants. Collectively, these results indicate that HIV-1 can replicate in a nontransgenic rodent and that this system may have potential as an animal model for HIV-1 infection if viral replication rates can be improved in vivo.


Asunto(s)
Infecciones por VIH/fisiopatología , VIH-1/fisiología , VIH-1/patogenicidad , Animales , Células Cultivadas , ADN Viral/sangre , Susceptibilidad a Enfermedades , Genes gag , Infecciones por VIH/transmisión , VIH-1/aislamiento & purificación , Humanos , Leucocitos Mononucleares/virología , Provirus/genética , Provirus/aislamiento & purificación , ARN Viral/sangre , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sigmodontinae , Especificidad de la Especie , Replicación Viral
16.
J Infect Dis ; 178(4): 925-32, 1998 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-9806017

RESUMEN

Antigenic differences between the two major groups of respiratory syncytial (RS) virus may contribute to reinfections with these viruses. Additional variability occurs within the two major groups; the importance of intra-group variability in reinfections with RS virus has not been defined. Two pairs of group A viruses that had caused sequential infections in children showed G protein amino acid differences of up to 15%. Vaccinia viruses were constructed that expressed the G proteins from 2 of the paired group A isolates. Immunization of cotton rats with the recombinant vaccinia viruses provided equal protection against intranasal challenge by either of the RS viruses. Despite the amino acid differences between the two group A RS virus G proteins, these animal studies did not reveal differences in protection after immunization with the two G proteins. Precise definition of the role of RS virus antigenic variability in the establishment of reinfections in humans will require further investigations in humans.


Asunto(s)
Antígenos Virales/genética , Proteína HN , Infecciones por Virus Sincitial Respiratorio/virología , Virus Sincitial Respiratorio Humano/genética , Proteínas Virales/genética , Secuencia de Aminoácidos , Animales , Variación Antigénica , Antígenos Virales/inmunología , Preescolar , Femenino , Variación Genética , Humanos , Lactante , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/inmunología , Recurrencia , Infecciones por Virus Sincitial Respiratorio/inmunología , Virus Sincitial Respiratorio Humano/clasificación , Virus Sincitial Respiratorio Humano/inmunología , Homología de Secuencia de Aminoácido , Sigmodontinae , Vacunación , Virus Vaccinia/genética , Proteínas del Envoltorio Viral , Proteínas Virales/biosíntesis , Proteínas Virales/inmunología
17.
Am J Respir Crit Care Med ; 157(5 Pt 1): 1506-11, 1998 May.
Artículo en Inglés | MEDLINE | ID: mdl-9603130

RESUMEN

A dysfunction of pathways that normally cause contraction or relaxation of airways has been proposed to explain heightened levels of responsiveness produced by various insults to the airway. For example, we previously reported (4) that infection of cotton rats with the human respiratory syncytial virus (HRSV) leads to a significant decrease in an airway's nonadrenergic noncholinergic inhibitory (NANCi) response shortly after the infection. In the present study we addressed the more chronic effects of HRSV infection on airway function in young ferrets during a period of rapid somatic growth. Animals 1 wk old received HRSV or uninfected cell culture medium intranasally. In vitro studies of airway function were performed on tracheal smooth muscle (TSM) segments at 4, 8, and 24 wk of age. To evaluate neurally mediated contractile responses, frequency-response curves to electrical field stimulation (EFS) were performed with results expressed in terms of the frequency causing 50% of the maximal contractile response (ES50). In addition, contractile responses of TSM to methacholine (MCh) were also assessed with results expressed as the concentration needed to produce 50% of the maximal contractile response (EC50). To gauge NANCi responses, TSM was contracted with neurokinin A in the presence of atropine, propranolol, and indomethacin. Relaxant responses to EFS were assessed at frequencies from 5 to 30 Hz, with results expressed as mean percent relaxation. We found increased contractile responses to EFS in infected animals compared with that in the control group in both 4- and 8-wk old animals (p = 0.001 and p = 0.008, respectively). This difference had resolved by 24 wk of age. There was no difference in TSM responses to MCh between the groups at any age. Although there were no NANCi responses in 4-wk-old ferrets from either group, NANCi responses were significantly decreased in 8-wk-old ferrets previously infected with HRSV in the first week of life (p = 0.0001). A significant difference persisted (p = 0.008), albeit to a lesser degree, at 24 wk of age. These findings demonstrate that HRSV produces prolonged alterations of TSM function in ferret airways in vitro.


Asunto(s)
Infecciones por Virus Sincitial Respiratorio/fisiopatología , Virus Sincitial Respiratorio Humano , Tráquea/inervación , Animales , Hurones , Técnicas In Vitro , Cloruro de Metacolina/farmacología , Contracción Muscular/efectos de los fármacos , Músculo Liso/efectos de los fármacos , Músculo Liso/fisiopatología , Tráquea/fisiopatología
18.
J Infect Dis ; 176(5): 1215-24, 1997 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-9359721

RESUMEN

Neutralizing polyclonal antibody to respiratory syncytial virus (RSV) has been shown to be an effective prophylactic agent when administered intravenously in high-risk infants. This study describes the generation of a humanized monoclonal antibody, MEDI-493, that recognizes a conserved neutralizing epitope on the F glycoprotein of RSV. The affinity of MEDI-493 was found to be equal to or slightly better than an isotype-matched chimeric derivative of the parent antibody. In plaque reduction, microneutralization, and fusion-inhibition assays, MEDI-493 was significantly more potent than the polyclonal preparation. Broad neutralization of a panel of 57 clinical isolates of the RSV A and B subtypes was demonstrated. Pretreatment of cotton rats with MEDI-493 resulted in 99% reduction of lung RSV titers at a dose of 2.5 mg/kg, corresponding to a serum concentration of 25-30 microg/mL. Further, MEDI-493 did not induce increased RSV infection or pathology in either a primary or a secondary challenge.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Anticuerpos Antivirales/inmunología , Proteína HN , Infecciones por Virus Sincitial Respiratorio/prevención & control , Virus Sincitial Respiratorio Humano/inmunología , Proteínas Virales/inmunología , Secuencia de Aminoácidos , Animales , Humanos , Datos de Secuencia Molecular , Sigmodontinae , Proteínas del Envoltorio Viral
19.
J Infect Dis ; 175(4): 932-4, 1997 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-9086152

RESUMEN

Two antigenic groups of respiratory syncytial virus (RSV) have been identified: A (RSV/A) and B (RSV/B). Topical administration of human IgG screened for high titers of antibody to RSV/A (RSVIg) is protective against RSV/A infection in the cotton rat model. The study attempted to determine if topical RSVIg would also be protective against RSV/B. Cotton rats were pretreated intranasally with RSVIg or with monospecific antiserum obtained from animals previously infected with RSV/A or RSV/B (day 0), challenged intranasally with RSV/A or RSV/B (day 1), and sacrificed for virus titration (day 5). Cotton rat antiserum to RSV/B protected against RSV/A and RSV/B, while antiserum to RSV/A protected only against RSV/A. RSVIg, although prepared on the basis of activity against RSV/A, was also protective against RSV/B.


Asunto(s)
Infecciones por Virus Sincitial Respiratorio/prevención & control , Virus Sincitiales Respiratorios/inmunología , Animales , Humanos , Sueros Inmunes/inmunología , Inmunoglobulina G/inmunología , Sigmodontinae
20.
Clin Infect Dis ; 25(6): 1363-8, 1997 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-9431379

RESUMEN

Over the past two decades, animal models of respiratory syncytial virus (RSV) infection have been developed using primates, cotton rats, mice, calves, guinea pigs, ferrets, and hamsters. Use of these models has shed light on the mechanisms of vaccine-enhanced disease seen in clinical trials of a formalin-inactivated RSV vaccine and has provided a means for testing efficacy and safety of candidate prophylactic and therapeutic strategies. The development of multiple animal models has coincided with the realization that RSV disease in humans is a multifaceted disease whose clinical manifestations and sequelae depend upon age, genetic makeup, immunologic status, and concurrent disease within subpopulations. There is no single human subpopulation in whom all forms of RSV disease manifest, nor is there a single animal model that duplicates all forms of RSV disease. The choice of an experimental model will be governed by the specific manifestation of disease to be studied.


Asunto(s)
Modelos Animales de Enfermedad , Infecciones por Virus Sincitial Respiratorio/etiología , Animales , Bovinos , Cricetinae , Hurones , Cobayas , Humanos , Ratones , Pan troglodytes , Ratas , Infecciones por Virus Sincitial Respiratorio/terapia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...