Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Front Immunol ; 15: 1406929, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39114655

RESUMEN

Numerous enveloped viruses, such as coronaviruses, influenza, and respiratory syncytial virus (RSV), utilize class I fusion proteins for cell entry. During this process, the proteins transition from a prefusion to a postfusion state, undergoing substantial and irreversible conformational changes. The prefusion conformation has repeatedly shown significant potential in vaccine development. However, the instability of this state poses challenges for its practical application in vaccines. While non-native disulfides have been effective in maintaining the prefusion structure, identifying stabilizing disulfide bonds remains an intricate task. Here, we present a general computational approach to systematically identify prefusion-stabilizing disulfides. Our method assesses the geometric constraints of disulfide bonds and introduces a ranking system to estimate their potential in stabilizing the prefusion conformation. We hypothesized that disulfides restricting the initial stages of the conformational switch could offer higher stability to the prefusion state than those preventing unfolding at a later stage. The implementation of our algorithm on the RSV F protein led to the discovery of prefusion-stabilizing disulfides that supported our hypothesis. Furthermore, the evaluation of our top design as a vaccine candidate in a cotton rat model demonstrated robust protection against RSV infection, highlighting the potential of our approach for vaccine development.


Asunto(s)
Disulfuros , Proteínas Virales de Fusión , Disulfuros/química , Animales , Proteínas Virales de Fusión/inmunología , Proteínas Virales de Fusión/química , Humanos , Infecciones por Virus Sincitial Respiratorio/inmunología , Infecciones por Virus Sincitial Respiratorio/prevención & control , Infecciones por Virus Sincitial Respiratorio/virología , Estabilidad Proteica , Diseño Asistido por Computadora , Conformación Proteica , Virus Sincitiales Respiratorios/inmunología , Vacunas contra Virus Sincitial Respiratorio/inmunología , Ratas , Modelos Moleculares
2.
Viruses ; 15(2)2023 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-36851691

RESUMEN

Human metapneumovirus (hMPV) is an important cause of respiratory disease in immunocompromised individuals, yet hMPV infection has not been modeled before in immunocompromised animals. In this work, cotton rats S. hispidus immunosuppressed by cyclophosphamide were infected with hMPV, and viral replication and pulmonary inflammation in these animals were compared to those in normal hMPV-infected S. hispidus. The efficacy of prophylactic and therapeutic administration of the anti-hMPV antibody MPV467 was also evaluated. Immunosuppressed animals had higher pulmonary and nasal titers of hMPV on day 5 post-infection compared to normal animals, and large amounts of hMPV were still present in the respiratory tract of immunosuppressed animals on days 7 and 9 post-infection, indicating prolonged viral replication. Immunosuppression was accompanied by reduced pulmonary histopathology in hMPV-infected cotton rats compared to normal animals; however, a delayed increase in pathology and pulmonary chemokine expression was seen in immunosuppressed cotton rats. Prophylactic and therapeutic MPV467 treatments protected both upper and lower respiratory tracts against hMPV infection. The lung pathology and pulmonary expression of IP-10 and MIP-1α mRNA were reduced by therapeutic MPV467 administration. These results indicate that immunosuppressed cotton rats represent a useful model for studying hMPV pathogenesis and for evaluating therapeutics that could alleviate hMPV-induced disease in immunocompromised subjects.


Asunto(s)
Huésped Inmunocomprometido , Metapneumovirus , Infecciones por Paramyxoviridae , Sigmodontinae , Animales , Humanos , Quimiocina CCL3 , Huésped Inmunocomprometido/inmunología , Terapia de Inmunosupresión , Infecciones por Paramyxoviridae/inmunología , Infecciones por Paramyxoviridae/virología , Sigmodontinae/inmunología , Sigmodontinae/virología , Modelos Animales de Enfermedad
3.
Vaccine ; 39(32): 4555-4563, 2021 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-34154864

RESUMEN

Despite the substantial health and economic burden caused by RSV-associated illness, no vaccine is available. The sole licensed treatment (palivizumab), composed of a monoclonal neutralizing antibody, blocks the fusion of the virus to the host cell but does not prevent infection. The development of a safe and efficacious RSV vaccine is therefore a priority, but also a considerable challenge, and new innovative strategies are warranted. Most of the adult population encounter regular RSV infections and can elicit a robust neutralizing antibody response, but unfortunately it wanes over time and reinfections during subsequent seasons are common. One approach to protect the mother and young infant from RSV infection is to administer a vaccine capable of boosting preexisting RSV immunity during pregnancy, which would provide protection to the fetus through passive transfer of maternal antibodies, thus preventing primary RSV infection in newborns during their first months of life. Here, we describe the preclinical evaluation of an epicutaneous RSV vaccine booster that combines epicutaneous patches as a delivery platform and a Synthetic Virus-Like Particles (SVLP)-based vaccine displaying multiple RSV F-protein site II (FsII, palivizumab epitope) mimetic as antigen (V-306). We demonstrated in mice that epicutaneous immunization with V-306 efficiently boosts preexisting immunity induced by the homologous V-306 administered subcutaneously. This boosting was characterized by a significant increase in F- and FsII-specific antibodies capable of competing with palivizumab for its target antigen and neutralize RSV. More importantly, epicutaneous booster immunization with V-306 significantly decreased lung viral replication in experimental mice after intranasal RSV challenge, without inducing enhanced RSV disease. In conclusion, an epicutaneous booster vaccine based on V-306 is safe and efficacious in enhancing RSV preexisting immunity in mice. This needle-free vaccine candidate would be potentially suited as a booster vaccine for vulnerable populations such as young infants via pregnant women, and the elderly.


Asunto(s)
Infecciones por Virus Sincitial Respiratorio , Vacunas contra Virus Sincitial Respiratorio , Virus Sincitial Respiratorio Humano , Anciano , Animales , Anticuerpos Neutralizantes , Anticuerpos Antivirales , Femenino , Humanos , Inmunización , Recién Nacido , Ratones , Embarazo , Infecciones por Virus Sincitial Respiratorio/prevención & control , Proteínas Virales de Fusión
4.
Vaccine ; 35(21): 2840-2847, 2017 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-28413132

RESUMEN

Respiratory syncytial virus (RSV) is a massive medical burden in infants, children and the elderly worldwide, and an effective, safe RSV vaccine remains an unmet need. Here we assess a novel vaccination strategy based on the intradermal delivery of a SynCon® DNA-based vaccine encoding engineered RSV-F antigen using a surface electroporation device (SEP) to target epidermal cells, in clinically relevant experimental models. We demonstrate the ability of this strategy to elicit robust immune responses. Importantly we demonstrate complete resistance to pulmonary infection at a single low dose of vaccine in the cotton rat RSV/A challenge model. In contrast to the formalin-inactivated RSV (FI-RSV) vaccine, there was no enhanced lung inflammation upon virus challenge after DNA vaccination. In summary the data presented outline the pre-clinical development of a highly efficacious, tolerable and safe non-replicating vaccine delivery strategy.


Asunto(s)
Electroporación/instrumentación , Infecciones por Virus Sincitial Respiratorio/prevención & control , Vacunas contra Virus Sincitial Respiratorio/administración & dosificación , Vacunas contra Virus Sincitial Respiratorio/inmunología , Vacunas de ADN/administración & dosificación , Vacunas de ADN/inmunología , Animales , Modelos Animales de Enfermedad , Femenino , Pulmón/patología , Sigmodontinae , Resultado del Tratamiento
5.
PLoS Negl Trop Dis ; 6(12): e1970, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23285308

RESUMEN

BACKGROUND: Chronic helminth infections induce a Th2 immune shift and establish an immunoregulatory milieu. As both of these responses can suppress Th1 immunity, which is necessary for control of Mycobacterium tuberculosis (MTB) infection, we hypothesized that chronic helminth infections may exacerbate the course of MTB. METHODOLOGY/PRINCIPAL FINDINGS: Co-infection studies were conducted in cotton rats as they are the natural host for the filarial nematode Litomosoides sigmodontis and are an excellent model for human MTB. Immunogical responses, histological studies, and quantitative mycobacterial cultures were assessed two months after MTB challenge in cotton rats with and without chronic L. sigmodontis infection. Spleen cell proliferation and interferon gamma production in response to purified protein derivative were similar between co-infected and MTB-only infected animals. In contrast to our hypothesis, MTB loads and occurrence and size of lung granulomas were not increased in co-infected animals. CONCLUSIONS/SIGNIFICANCE: These findings suggest that chronic filaria infections do not exacerbate MTB infection in the cotton rat model. While these results suggest that filaria eradication programs may not facilitate MTB control, they indicate that it may be possible to develop worm-derived therapies for autoimmune diseases that do not substantially increase the risk for infections.


Asunto(s)
Filariasis/complicaciones , Tuberculosis/complicaciones , Tuberculosis/patología , Animales , Carga Bacteriana , Proliferación Celular , Enfermedad Crónica , Coinfección/inmunología , Coinfección/microbiología , Coinfección/parasitología , Coinfección/patología , Modelos Animales de Enfermedad , Femenino , Filariasis/inmunología , Filarioidea/inmunología , Filarioidea/patogenicidad , Histocitoquímica , Interferón gamma/metabolismo , Leucocitos Mononucleares/inmunología , Pulmón/microbiología , Pulmón/patología , Mycobacterium tuberculosis/inmunología , Mycobacterium tuberculosis/patogenicidad , Sigmodontinae , Bazo/inmunología , Tuberculosis/inmunología
6.
J Leukoc Biol ; 89(3): 351-7, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21084628

RESUMEN

The 2009 outbreak of pandemic H1N1 influenza, increased drug resistance, and the significant delay in obtaining adequate numbers of vaccine doses have heightened awareness of the need to develop new antiviral drugs that can be used prophylactically or therapeutically. Previously, we showed that the experimental anti-tumor drug DMXAA potently induced IFN-ß but relatively low TNF-α expression in vitro. This study confirms these findings in vivo and demonstrates further that DMXAA induces potent antiviral activity in vitro and in vivo. In vitro, DMXAA protected RAW 264.7 macrophage-like cells from VSV-induced cytotoxicity and moreover, inhibited replication of influenza, including the Tamiflu®-resistant H1N1 influenza A/Br strain, in MDCK cells. In vivo, DMXAA protected WT C57BL/6J but not IFN-ß(-/-) mice from lethality induced by the mouse-adapted H1N1 PR8 influenza strain when administered before or after infection. Protection was accompanied by mitigation of weight loss, increased IFN-ß mRNA and protein levels in the lung, and significant inhibition of viral replication in vivo early after DMXAA treatment. Collectively, this study provides data to support the use of DMXAA as a novel antiviral agent.


Asunto(s)
Antineoplásicos/farmacología , Antivirales/farmacología , Subtipo H1N1 del Virus de la Influenza A/efectos de los fármacos , Interferón beta/metabolismo , Xantonas/farmacología , Adaptación Fisiológica/efectos de los fármacos , Animales , Antineoplásicos/administración & dosificación , Muerte Celular/efectos de los fármacos , Farmacorresistencia Viral/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Subtipo H1N1 del Virus de la Influenza A/fisiología , Interferón beta/genética , Lipopolisacáridos/farmacología , Ratones , Ratones Endogámicos C57BL , Oseltamivir/farmacología , Sustancias Protectoras/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Vesiculovirus/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Xantonas/administración & dosificación
7.
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
8.
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
9.
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
10.
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
11.
J Virol ; 79(23): 14632-9, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16282463

RESUMEN

Prevention of genital herpes and other sexually transmitted infections (STI) is a critical health priority because of the overwhelming impact on women and infants and the epidemiological association with human immunodeficiency virus (HIV)/AIDS. Small animal models are essential for evaluating strategies for prevention or treatment of STI. Neither the murine nor the guinea pig model of genital herpes fully recapitulates human disease. We demonstrate that herpes simplex virus type 2 (HSV-2) readily infects inbred cotton rats (Sigmodon hispidus). Consistent infection does not require pretreatment with medroxyprogesterone, and primary disease resembles that observed in humans. The animals develop genital lesions and fully recover. During primary infection, viral DNA is also detected in liver, lungs, brain, and kidneys. Clinical self-limited recurrences occur spontaneously but may also be induced by dexamethasone. Pretreatment of cotton rats with PRO 2000 gel, a candidate vaginal microbicide being evaluated in clinical trials to prevent HSV and HIV, protects cotton rats from HSV. Together, these studies suggest that the cotton rat may provide an excellent model to study genital herpes and to evaluate preventive strategies.


Asunto(s)
Modelos Animales de Enfermedad , Herpes Genital/veterinaria , Infecciones por Herpesviridae/veterinaria , Herpesvirus Humano 1/fisiología , Animales , Herpes Genital/prevención & control , Infecciones por Herpesviridae/epidemiología , Infecciones por Herpesviridae/prevención & control , Herpesvirus Humano 2/inmunología , Herpesvirus Humano 2/fisiología , Medicina Preventiva/métodos , Sigmodontinae/genética
12.
J Virol ; 79(15): 9515-26, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16014914

RESUMEN

The outcome of a viral infection or of immunization with a vaccine can be influenced by the local cytokine environment. In studies of experimental vaccines against respiratory syncytial virus (RSV), an increased stimulation of Th2 (T helper 2) lymphocytes was associated with increased immunopathology upon subsequent RSV infection. For this study, we investigated the effect of increased local expression of the Th2 cytokine interleukin-4 (IL-4) from the genome of a recombinant RSV following primary infection and after a challenge with wild-type (wt) RSV. Mice infected with RSV/IL-4 exhibited an accelerated pulmonary inflammatory response compared to those infected with wt RSV, although the wt RSV group caught up by day 8. In the first few days postinfection, RSV/IL-4 was associated with a small but significant acceleration in the expansion of pulmonary T lymphocytes specific for an RSV CD8(+) cytotoxic T-lymphocyte (CTL) epitope presented as a major histocompatibility complex class I tetramer. However, by day 7 the response of tetramer-positive T lymphocytes in the wt RSV group caught up and exceeded that of the RSV/IL-4 group. At all times, the CTL response of the RSV/IL-4 group was deficient in the production of gamma interferon and was nonfunctional for in vitro cell killing. The accelerated inflammatory response coincided with an accelerated accumulation and activation of pulmonary dendritic cells early in infection, but thereafter the dendritic cells were deficient in the expression of B7-1, which governs the acquisition of cytolytic activity by CTL. Following a challenge with wt RSV, there was an increase in Th2 cytokines in the animals that had previously been infected with RSV/IL-4 compared to those previously infected with wt RSV, but the CD8(+) CTL response and the amount of pulmonary inflammation were not significantly different. Thus, a strong Th2 environment during primary pulmonary immunization with live RSV resulted in early inflammation and a largely nonfunctional primary CTL response but had a minimal effect on the secondary response.


Asunto(s)
Inflamación/patología , Interleucina-4/biosíntesis , Virus Reordenados/inmunología , Infecciones por Virus Sincitial Respiratorio/inmunología , Infecciones por Virus Sincitial Respiratorio/patología , Virus Sincitiales Respiratorios/inmunología , Linfocitos T Citotóxicos/inmunología , Animales , Linfocitos T CD8-positivos/inmunología , Proliferación Celular , Células Dendríticas/citología , Antígenos de Histocompatibilidad Clase I/inmunología , Interleucina-4/genética , Pulmón/inmunología , Pulmón/patología , Activación de Linfocitos , Ratones , Ratones Endogámicos BALB C , Infecciones por Virus Sincitial Respiratorio/genética , Factores de Tiempo
13.
J Virol ; 77(24): 13156-60, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-14645572

RESUMEN

CpG oligodeoxynucleotides (ODN) were identified that stimulated immunoglobulin production and cell proliferation in cotton rat cells in vitro. Three of these ODN were used as a mucosal adjuvant in the noses of cotton rats immunized via this route with respiratory syncytial virus fusion (F) protein. The CpG ODN markedly increased the cotton rat humoral neutralizing-antibody response to respiratory syncytial virus. Such immunized animals had a marked reduction in the production of infectious virus after a live-virus challenge. Animals immunized with the combination of F protein and CpG developed enhanced pulmonary pathology consisting of alveolitis and interstitial pneumonitis after a live-virus challenge. Similar enhanced disease has been seen in cotton rats and children immunized with formalin-inactivated respiratory syncytial virus.


Asunto(s)
Adyuvantes Inmunológicos , Islas de CpG/inmunología , Oligodesoxirribonucleótidos/inmunología , Vacunas contra Virus Sincitial Respiratorio/efectos adversos , Vacunas contra Virus Sincitial Respiratorio/inmunología , Proteínas Virales/inmunología , Administración Intranasal , Animales , Anticuerpos Antivirales/sangre , Modelos Animales de Enfermedad , Femenino , Humanos , Inmunización , Pulmón/patología , Masculino , Membrana Mucosa , Oligodesoxirribonucleótidos/administración & dosificación , Infecciones por Virus Sincitial Respiratorio/inmunología , Infecciones por Virus Sincitial Respiratorio/patología , Infecciones por Virus Sincitial Respiratorio/prevención & control , Infecciones por Virus Sincitial Respiratorio/virología , Vacunas contra Virus Sincitial Respiratorio/administración & dosificación , Virus Sincitiales Respiratorios/inmunología , Sigmodontinae , Proteínas Virales de Fusión/genética , Proteínas Virales de Fusión/inmunología , Proteínas Virales/genética
14.
J Gen Virol ; 82(Pt 12): 2881-2888, 2001 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11714962

RESUMEN

A formalin-inactivated respiratory syncytial virus vaccine was used to immunize infants in the mid-1960s; when these children later were naturally infected by the virus they developed markedly accentuated disease, and two died. For the present work, a new batch of vaccine was prepared using the original formula. Administration of either the old or new vaccines resulted in enhanced lesions in immunized cotton rats subsequently challenged with live virus, although administration of the vaccine reduced virus replication by 90%. Animals primed with formalin-inactivated virus and challenged 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 appear to be specific markers for the vaccine enhancement. The newly prepared vaccine appears suitable as a reference standard for studying the mechanism of vaccine-enhanced disease caused by this virus. Additionally, we reviewed the lesions in the lungs of the two humans who died with the vaccine-enhanced disease in 1967, and found that they were similar to, but more severe than those seen in the cotton rats.


Asunto(s)
Infecciones por Virus Sincitial Respiratorio/prevención & control , Virus Sincitiales Respiratorios/inmunología , Vacunación , Vacunas Virales/administración & dosificación , Animales , Autopsia , Bronquiolitis/fisiopatología , Modelos Animales de Enfermedad , Femenino , Formaldehído , Humanos , Pulmón/patología , Pulmón/virología , Masculino , Alveolos Pulmonares/patología , Infecciones por Virus Sincitial Respiratorio/fisiopatología , Sigmodontinae , Factores de Tiempo , Vacunas de Productos Inactivados/administración & dosificación , Vacunas de Productos Inactivados/efectos adversos , Vacunas Virales/efectos adversos , Vacunas Virales/normas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA