Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
J Virol ; 95(14): e0013021, 2021 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-33893170

RESUMEN

The nasal mucosa constitutes the primary entry site for respiratory viruses, including severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). While the imbalanced innate immune response of end-stage coronavirus disease 2019 (COVID-19) has been extensively studied, the earliest stages of SARS-CoV-2 infection at the mucosal entry site have remained unexplored. Here, we employed SARS-CoV-2 and influenza virus infection in native multi-cell-type human nasal turbinate and lung tissues ex vivo, coupled with genome-wide transcriptional analysis, to investigate viral susceptibility and early patterns of local mucosal innate immune response in the authentic milieu of the human respiratory tract. SARS-CoV-2 productively infected the nasal turbinate tissues, predominantly targeting respiratory epithelial cells, with a rapid increase in tissue-associated viral subgenomic mRNA and secretion of infectious viral progeny. Importantly, SARS-CoV-2 infection triggered robust antiviral and inflammatory innate immune responses in the nasal mucosa. The upregulation of interferon-stimulated genes, cytokines, and chemokines, related to interferon signaling and immune-cell activation pathways, was broader than that triggered by influenza virus infection. Conversely, lung tissues exhibited a restricted innate immune response to SARS-CoV-2, with a conspicuous lack of type I and III interferon upregulation, contrasting with their vigorous innate immune response to influenza virus. Our findings reveal differential tissue-specific innate immune responses in the upper and lower respiratory tracts that are specific to SARS-CoV-2. The studies shed light on the role of the nasal mucosa in active viral transmission and immune defense, implying a window of opportunity for early interventions, whereas the restricted innate immune response in early-SARS-CoV-2-infected lung tissues could underlie the unique uncontrolled late-phase lung damage of advanced COVID-19. IMPORTANCE In order to reduce the late-phase morbidity and mortality of COVID-19, there is a need to better understand and target the earliest stages of SARS-CoV-2 infection in the human respiratory tract. Here, we have studied the initial steps of SARS-CoV-2 infection and the consequent innate immune responses within the natural multicellular complexity of human nasal mucosal and lung tissues. Comparing the global innate response patterns of nasal and lung tissues infected in parallel with SARS-CoV-2 and influenza virus, we found distinct virus-host interactions in the upper and lower respiratory tract, which could determine the outcome and unique pathogenesis of SARS-CoV-2 infection. Studies in the nasal mucosal infection model can be employed to assess the impact of viral evolutionary changes and evaluate new therapeutic and preventive measures against SARS-CoV-2 and other human respiratory pathogens.


Asunto(s)
COVID-19/inmunología , Inmunidad Innata , Pulmón/inmunología , Mucosa Nasal/inmunología , SARS-CoV-2/inmunología , Animales , COVID-19/patología , Chlorocebus aethiops , Perros , Humanos , Gripe Humana/inmunología , Gripe Humana/patología , Pulmón/patología , Células de Riñón Canino Madin Darby , Mucosa Nasal/patología , Mucosa Nasal/virología , Especificidad de Órganos/inmunología , ARN Mensajero/inmunología , ARN Viral/inmunología , Células Vero
2.
J Virol ; 94(19)2020 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-32727881

RESUMEN

The initial events of viral infection at the primary mucosal entry site following horizontal person-to-person transmission have remained ill defined. Our limited understanding is further underscored by the absence of animal models in the case of human-restricted viruses, such as human cytomegalovirus (HCMV), a leading cause of congenital infection and a major pathogen in immunocompromised individuals. Here, we established a novel ex vivo model of HCMV infection in native human nasal turbinate tissues. Nasal turbinate tissue viability and physiological functionality were preserved for at least 7 days in culture. We found that nasal mucosal tissues were susceptible to HCMV infection, with predominant infection of ciliated respiratory epithelial cells. A limited viral spread was demonstrated, involving mainly stromal and vascular endothelial cells within the tissue. Importantly, functional antiviral and proleukocyte chemotactic signaling pathways were significantly upregulated in the nasal mucosa in response to infection. Conversely, HCMV downregulated the expression of nasal epithelial cell-related genes. We further revealed tissue-specific innate immune response patterns to HCMV, comparing infected human nasal mucosal and placental tissues, representing the viral entry and the maternal-to-fetal transmission sites, respectively. Taken together, our studies provide insights into the earliest stages of HCMV infection. Studies in this model could help evaluate new interventions against the horizontal transmission of HCMV.IMPORTANCE HCMV is a ubiquitous human pathogen causing neurodevelopmental disabilities in congenitally infected children and severe disease in immunocompromised patients. The earliest stages of HCMV infection in the human host have remained elusive in the absence of a model for the viral entry site. Here, we describe the establishment and use of a novel nasal turbinate organ culture to study the initial steps of viral infection and the consequent innate immune responses within the natural complexity and the full cellular repertoire of human nasal mucosal tissues. This model can be applied to examine new antiviral interventions against the horizontal transmission of HCMV and potentially that of other viruses.


Asunto(s)
Infecciones por Citomegalovirus/virología , Citomegalovirus/fisiología , Cornetes Nasales/virología , Internalización del Virus , Línea Celular , Infecciones por Citomegalovirus/patología , Infecciones por Citomegalovirus/transmisión , Células Endoteliales , Femenino , Fibroblastos , Prepucio , Humanos , Inmunidad Innata , Transmisión Vertical de Enfermedad Infecciosa , Masculino , Membrana Mucosa , Técnicas de Cultivo de Órganos , Embarazo
3.
J Virol ; 91(23)2017 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-28956761

RESUMEN

Human cytomegalovirus (HCMV) is the leading cause of congenital infection and is associated with a wide range of neurodevelopmental disabilities and intrauterine growth restriction. Yet our current understanding of the mechanisms modulating transplacental HCMV transmission is poor. The placenta, given its critical function in protecting the fetus, has evolved effective yet largely uncharacterized innate immune barriers against invading pathogens. Here we show that the intrinsic cellular restriction factor apolipoprotein B editing catalytic subunit-like 3A (APOBEC3A [A3A]) is profoundly upregulated following ex vivo HCMV infection in human decidual tissues-constituting the maternal aspect of the placenta. We directly demonstrated that A3A severely restricted HCMV replication upon controlled overexpression in epithelial cells, acting by a cytidine deamination mechanism to introduce hypermutations into the viral genome. Importantly, we further found that A3 editing of HCMV DNA occurs both ex vivo in HCMV-infected decidual organ cultures and in vivo in amniotic fluid samples obtained during natural congenital infection. Our results reveal a previously unexplored role for A3A as an innate anti-HCMV effector, activated by HCMV infection in the maternal-fetal interface. These findings pave the way to new insights into the potential impact of APOBEC proteins on HCMV pathogenesis.IMPORTANCE In view of the grave outcomes associated with congenital HCMV infection, there is an urgent need to better understand the innate mechanisms acting to limit transplacental viral transmission. Toward this goal, our findings reveal the role of the intrinsic cellular restriction factor A3A (which has never before been studied in the context of HCMV infection and vertical viral transmission) as a potent anti-HCMV innate barrier, activated by HCMV infection in the authentic tissues of the maternal-fetal interface. The detection of naturally occurring hypermutations in clinical amniotic fluid samples of congenitally infected fetuses further supports the idea of the occurrence of A3 editing of the viral genome in the setting of congenital HCMV infection. Given the widely differential tissue distribution characteristics and biological functions of the members of the A3 protein family, our findings should pave the way to future studies examining the potential impact of A3A as well as of other A3s on HCMV pathogenesis.


Asunto(s)
Citidina Desaminasa/genética , Citidina Desaminasa/metabolismo , Infecciones por Citomegalovirus/virología , Decidua/inmunología , Inmunidad Innata , Placenta/inmunología , Proteínas/genética , Proteínas/metabolismo , Líquido Amniótico/inmunología , Líquido Amniótico/virología , Citidina Desaminasa/inmunología , Citomegalovirus/genética , Citomegalovirus/inmunología , Citomegalovirus/patogenicidad , Infecciones por Citomegalovirus/congénito , Infecciones por Citomegalovirus/inmunología , Decidua/citología , Decidua/virología , Femenino , Edición Génica , Genoma Viral , Humanos , Recién Nacido , Transmisión Vertical de Enfermedad Infecciosa , Técnicas de Cultivo de Órganos , Placenta/citología , Placenta/virología , Embarazo , Complicaciones Infecciosas del Embarazo/inmunología , Complicaciones Infecciosas del Embarazo/virología , Proteínas/inmunología , Regulación hacia Arriba , Replicación Viral
4.
J Virol ; 91(4)2017 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-27974560

RESUMEN

Zika virus (ZIKV) has emerged as a cause of congenital brain anomalies and a range of placenta-related abnormalities, highlighting the need to unveil the modes of maternal-fetal transmission. The most likely route of vertical ZIKV transmission is via the placenta. The earliest events of ZIKV transmission in the maternal decidua, representing the maternal uterine aspect of the chimeric placenta, have remained unexplored. Here, we show that ZIKV replicates in first-trimester human maternal-decidual tissues grown ex vivo as three-dimensional (3D) organ cultures. An efficient viral spread in the decidual tissues was demonstrated by the rapid upsurge and continued increase of tissue-associated ZIKV load and titers of infectious cell-free virus progeny, released from the infected tissues. Notably, maternal decidual tissues obtained at midgestation remained similarly susceptible to ZIKV, whereas fetus-derived chorionic villi demonstrated reduced ZIKV replication with increasing gestational age. A genome-wide transcriptome analysis revealed that ZIKV substantially upregulated the decidual tissue innate immune responses. Further comparison of the innate tissue response patterns following parallel infections with ZIKV and human cytomegalovirus (HCMV) revealed that unlike HCMV, ZIKV did not induce immune cell activation or trafficking responses in the maternal-fetal interface but rather upregulated placental apoptosis and cell death molecular functions. The data identify the maternal uterine aspect of the human placenta as a likely site of ZIKV transmission to the fetus and further reveal distinct patterns of innate tissue responses to ZIKV. Our unique experimental model and findings could further serve to study the initial stages of congenital ZIKV transmission and pathogenesis and evaluate the effect of new therapeutic interventions. IMPORTANCE: In view of the rapid spread of the current ZIKV epidemic and the severe manifestations of congenital ZIKV infection, it is crucial to learn the fundamental mechanisms of viral transmission from the mother to the fetus. Our studies of ZIKV infection in the authentic tissues of the human maternal-fetal interface unveil a route of transmission whereby virus originating from the mother could reach the fetal compartment via efficient replication within the maternal decidual aspect of the placenta, coinhabited by maternal and fetal cells. The identified distinct placental tissue innate immune responses and damage pathways could provide a mechanistic basis for some of the placental developmental abnormalities associated with ZIKV infection. The findings in the unique model of the human decidua should pave the way to future studies examining the interaction of ZIKV with decidual immune cells and to evaluation of therapeutic interventions aimed at the earliest stages of transmission.


Asunto(s)
Decidua/virología , Inmunidad Innata , Placenta/virología , Complicaciones Infecciosas del Embarazo , Infección por el Virus Zika/inmunología , Infección por el Virus Zika/virología , Virus Zika/fisiología , Animales , Línea Celular , Vellosidades Coriónicas/virología , Citomegalovirus/inmunología , Infecciones por Citomegalovirus/inmunología , Infecciones por Citomegalovirus/transmisión , Infecciones por Citomegalovirus/virología , Susceptibilidad a Enfermedades , Femenino , Expresión Génica , Edad Gestacional , Humanos , Transmisión Vertical de Enfermedad Infecciosa , Interferones/genética , Interferones/metabolismo , Embarazo , Transducción de Señal , Infección por el Virus Zika/metabolismo , Infección por el Virus Zika/transmisión
6.
Cell Rep ; 43(2): 113698, 2024 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-38265934

RESUMEN

Congenital cytomegalovirus (cCMV) is the most common intrauterine infection, leading to infant neurodevelopmental disabilities. An improved knowledge of correlates of protection against cCMV is needed to guide prevention strategies. Here, we employ an ex vivo model of human CMV (HCMV) infection in decidual tissues of women with and without preconception immunity against CMV, recapitulating nonprimary vs. primary infection at the authentic maternofetal transmission site. We show that decidual tissues of women with preconception immunity against CMV exhibit intrinsic resistance to HCMV, mounting a rapid activation of tissue-resident memory CD8+ and CD4+ T cells upon HCMV reinfection. We further reveal the role of HCMV-specific decidual-tissue-resident CD8+ T cells in local protection against nonprimary HCMV infection. The findings could inform the development of a vaccine against cCMV and provide insights for further studies of the integrity of immune defense against HCMV and other pathogens at the human maternal-fetal interface.


Asunto(s)
Infecciones por Citomegalovirus , Citomegalovirus , Lactante , Humanos , Femenino , Linfocitos T CD8-positivos , Células T de Memoria , Feto
7.
J Gen Virol ; 93(Pt 8): 1664-1672, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22622327

RESUMEN

Advanced melanoma cells, characterized by resistance to chemotherapy, have been shown to be highly sensitive to oncolysis by Newcastle disease virus (NDV). In the present study, we investigated the capacity of NDV to specifically infect and spread into solid tissues of human melanoma and lung carcinoma, in vivo and ex vivo. For this purpose a new model of SCID-beige mice implanted with human melanoma was developed. Surprisingly, the replication competent NDV-MTH and the attenuated, single-cycle replication NDV-HUJ strains, demonstrated a similar oncolytic activity in the melanoma-implanted mice. Further, ex vivo analysis, using organ cultures derived from the melanoma tissues indicated a limited spread of the two NDV strains in the tissue. Extracellular matrix (ECM) molecules, notably heparin sulfate and collagen, were found to limit viral spread in the tissue. This observation was validated with yet another solid tumour of human lung carcinoma. Taken together, the results indicate that the ECM acts as a barrier to virus spread within solid tumour tissues and that this restriction must be overcome to achieve effective oncolysis with NDV.


Asunto(s)
Carcinoma/metabolismo , Carcinoma/virología , Matriz Extracelular/metabolismo , Melanoma/metabolismo , Melanoma/virología , Virus de la Enfermedad de Newcastle/fisiología , Animales , Humanos , Neoplasias Pulmonares/metabolismo , Ratones , Ratones SCID , Trasplante de Neoplasias , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo , Técnicas de Cultivo de Tejidos , Replicación Viral
8.
J Virol ; 85(24): 13204-13, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21976654

RESUMEN

Human cytomegalovirus (HCMV) is the leading cause of congenital infection, associated with severe birth defects and intrauterine growth retardation. The mechanism of HCMV transmission via the maternal-fetal interface is largely unknown, and there are no animal models for HCMV. The initial stages of infection are believed to occur in the maternal decidua. Here we employed a novel decidual organ culture, using both clinically derived and laboratory-derived viral strains, for the ex vivo modeling of HCMV transmission in the maternal-fetal interface. Viral spread in the tissue was demonstrated by the progression of infected-cell foci, with a 1.3- to 2-log increase in HCMV DNA and RNA levels between days 2 and 9 postinfection, the expression of immediate-early and late proteins, the appearance of typical histopathological features of natural infection, and dose-dependent inhibition of infection by ganciclovir and acyclovir. HCMV infected a wide range of cells in the decidua, including invasive cytotrophoblasts, macrophages, and endothelial, decidual, and dendritic cells. Cell-to-cell viral spread was revealed by focal extension of infected-cell clusters, inability to recover infectious extracellular virus, and high relative proportions (88 to 93%) of cell-associated viral DNA. Intriguingly, neutralizing HCMV hyperimmune globulins exhibited inhibitory activity against viral spread in the decidua even when added at 24 h postinfection-providing a mechanistic basis for their clinical use in prenatal prevention. The ex vivo-infected decidual cultures offer unique insight into patterns of viral tropism and spread, defining initial stages of congenital HCMV transmission, and can facilitate evaluation of the effects of new antiviral interventions within the maternal-fetal interface milieu.


Asunto(s)
Infecciones por Citomegalovirus/transmisión , Decidua/virología , Transmisión Vertical de Enfermedad Infecciosa , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , ADN Viral/genética , ADN Viral/aislamiento & purificación , Femenino , Expresión Génica , Humanos , Modelos Biológicos , Técnicas de Cultivo de Órganos/métodos , Embarazo , ARN Viral/genética , ARN Viral/aislamiento & purificación , Factores de Tiempo , Proteínas Virales/biosíntesis
9.
Planta Med ; 78(10): 962-7, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22588835

RESUMEN

Cranberry juice contains high molecular weight non-dialyzable material (NDM) which was found to inhibit hemagglutination induced by the influenza virus (IV) as well as to neutralize the cytotoxicity of IV in cell cultures. Because influenza virus surface glycoproteins hemagglutinin (HA) and neuraminidase (NA) are involved in viral replication and in the infectious process, we sought in the present study to examine the effect of NDM on neuraminidases which are the target of most anti-influenza drugs today. NDM inhibited the NA enzymatic activity of influenza A and B strains as well as that of Streptococcus pneumoniae. This finding is of importance considering the emergence of influenza isolates resistant to antiviral drugs, reaching 90 % in some places. The anti-NA activity of NDM, evaluated by the MUNANA method and expressed as the concentration required for 50 % inhibition (IC50), was most potent against N1 (IC50, 192 µg/mL), less active against BN and N2 (IC50, 509 µg/mL and 1128 µg/mL, respectively), and moderately active against Streptococcus pneumoniae NA (IC50, 594 µg/mL). The in vitro findings of the present study suggest that cranberry constituents may have a therapeutic potential against both A and B influenza virus infections and might also interfere with the development of secondary bacterial complications.


Asunto(s)
Antivirales/farmacología , Inhibidores Enzimáticos/farmacología , Subtipo H1N1 del Virus de la Influenza A/efectos de los fármacos , Virus de la Influenza B/efectos de los fármacos , Extractos Vegetales/farmacología , Vaccinium macrocarpon/química , Animales , Antibacterianos/farmacología , Línea Celular , Perros , Evaluación Preclínica de Medicamentos , Activación Enzimática , Eritrocitos , Pruebas de Hemaglutinación/métodos , Subtipo H1N1 del Virus de la Influenza A/enzimología , Subtipo H3N2 del Virus de la Influenza A/efectos de los fármacos , Subtipo H3N2 del Virus de la Influenza A/enzimología , Virus de la Influenza B/enzimología , Concentración 50 Inhibidora , Pruebas de Sensibilidad Microbiana/métodos , Peso Molecular , Neuraminidasa/antagonistas & inhibidores , Extractos Vegetales/química , Streptococcus/efectos de los fármacos , Streptococcus/enzimología , Proteínas Virales/antagonistas & inhibidores , Cultivo de Virus/métodos
10.
Viruses ; 14(7)2022 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-35891570

RESUMEN

SARS-CoV-2 Omicron variant has been characterized by decreased clinical severity, raising the question of whether early variant-specific interactions within the mucosal surfaces of the respiratory tract could mediate its attenuated pathogenicity. Here, we employed ex vivo infection of native human nasal and lung tissues to investigate the local-mucosal susceptibility and innate immune response to Omicron compared to Delta and earlier SARS-CoV-2 variants of concern (VOC). We show that the replication of Omicron in lung tissues is highly restricted compared to other VOC, whereas it remains relatively unchanged in nasal tissues. Mechanistically, Omicron induced a much stronger antiviral interferon response in infected tissues compared to Delta and earlier VOC-a difference, which was most striking in the lung tissues, where the innate immune response to all other SARS-CoV-2 VOC was blunted. Notably, blocking the innate immune signaling restored Omicron replication in the lung tissues. Our data provide new insights to the reduced lung involvement and clinical severity of Omicron.


Asunto(s)
COVID-19 , Interferones , Pulmón , COVID-19/inmunología , Humanos , Interferones/inmunología , Pulmón/inmunología , Pulmón/virología , SARS-CoV-2/fisiología , Replicación Viral
11.
J Clin Invest ; 132(11)2022 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-35439172

RESUMEN

BACKGROUNDCytomegalovirus (CMV) is the most common intrauterine infection, leading to infant brain damage. Prognostic assessment of CMV-infected fetuses has remained an ongoing challenge in prenatal care, in the absence of established prenatal biomarkers of congenital CMV (cCMV) infection severity. We aimed to identify prognostic biomarkers of cCMV-related fetal brain injury.METHODSWe performed global proteome analysis of mid-gestation amniotic fluid samples, comparing amniotic fluid of fetuses with severe cCMV with that of asymptomatic CMV-infected fetuses. The levels of selected differentially excreted proteins were further determined by specific immunoassays.RESULTSUsing unbiased proteome analysis in a discovery cohort, we identified amniotic fluid proteins related to inflammation and neurological disease pathways, which demonstrated distinct abundance in fetuses with severe cCMV. Amniotic fluid levels of 2 of these proteins - the immunomodulatory proteins retinoic acid receptor responder 2 (chemerin) and galectin-3-binding protein (Gal-3BP) - were highly predictive of the severity of cCMV in an independent validation cohort, differentiating between fetuses with severe (n = 17) and asymptomatic (n = 26) cCMV, with 100%-93.8% positive predictive value, and 92.9%-92.6% negative predictive value (for chemerin and Gal-3BP, respectively). CONCLUSIONAnalysis of chemerin and Gal-3BP levels in mid-gestation amniotic fluids could be used in the clinical setting to profoundly improve the prognostic assessment of CMV-infected fetuses.FUNDINGIsrael Science Foundation (530/18 and IPMP 3432/19); Research Fund - Hadassah Medical Organization.


Asunto(s)
Infecciones por Citomegalovirus , Complicaciones Infecciosas del Embarazo , Líquido Amniótico , Biomarcadores , Citomegalovirus , Infecciones por Citomegalovirus/diagnóstico , Femenino , Humanos , Lactante , Embarazo , Proteoma
12.
J Virol ; 84(1): 639-46, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19864394

RESUMEN

Patients with advanced melanoma usually do not benefit from conventional chemotherapy treatment. There is therefore a true need for a new kind of therapy for melanoma. One factor responsible for the poor prognosis of melanoma is the inhibitor of apoptosis protein (IAP) family member Livin. In this study, we applied a novel approach for the treatment of melanoma, using a unique strain of the oncolytic Newcastle disease virus (NDV-HUJ). We found that, unlike chemotherapeutic drugs, NDV-HUJ, a one-cycle replicating virus, overcomes the resistance to apoptosis of melanoma primary cultures that over express the Livin protein. In contrast, melanoma tumor cells that do not express Livin are relatively resistant to NDV-HUJ treatment. Furthermore, we show that NDV-HUJ-induced oncolysis is attributed to the dual function of Livin: although Livin inhibits apoptosis through the inhibition of caspases, under the robust apoptotic stimulation of NDV-HUJ, caspases can cleave Livin to create a truncated protein with a paradoxical proapoptotic activity. Thus, NDV-HUJ is a potent inducer of apoptosis that can overcome the antiapoptotic effect of Livin and allow cleavage of Livin into the proapoptotic tLivin protein. Moreover, the results indicate that the interferon system, which is functional in melanoma, is not involved in NDV-induced oncolysis. Taken together, our data offer the possibility of a new viral oncolytic treatment for chemoresistant melanoma.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Resistencia a Antineoplásicos , Proteínas Inhibidoras de la Apoptosis/fisiología , Melanoma/terapia , Proteínas de Neoplasias/fisiología , Virus de la Enfermedad de Newcastle/patogenicidad , Virus Oncolíticos/patogenicidad , Apoptosis , Inhibidores de Caspasas , Caspasas/metabolismo , Humanos , Melanoma/patología , Viroterapia Oncolítica/métodos , Células Tumorales Cultivadas
13.
Isr Med Assoc J ; 8(3): 155-8, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16599048

RESUMEN

BACKGROUND: The evaluation of influenza vaccine activity and potency are based on the immune response to hemagglutinin, and protection is indicated when the titer of hemagglutination inhibition serum antibody is > or = 1:40. Neuraminidase, the second surface glycoprotein, may also have a role in protection, but little information is available on the immunologic response to this component. OBJECTIVES: To determine whether any response to neuraminidase is evoked by intranasal immunization with a novel, whole, inactivated anti-influenza vaccine. METHODS: This study was part of a more comprehensive study of mucosal and serum immune response to this vaccine. Fifty-four young adults were immunized intranasally, 9 intramuscularly and 18 received a placebo. Twenty-three elderly people were immunized intramuscularly, and 21 elderly and 17 children were immunized intranasally. Serum and nasal antibodies to antigens N1 and N2 were determined by the lectin neuraminidase test. RESULTS: Serum response following intranasal vaccination was lower than after intramuscular vaccination, and ranged from 21.4 to 35.3% and 33.3 to 64.7% following intranasal vaccination and from 52.2 to 77.8% and 47.8 to 88.9% after intramuscular vaccination, to N1 and N2 respectively. Nasal antibody response was low and was found only after intranasal vaccination, and response to N2 was better than to the N1 antigen. CONCLUSIONS: It may be beneficial if future vaccines would include competent hemagglutinin and neuraminidase, which would afford a higher level of protection.


Asunto(s)
Anticuerpos/metabolismo , Virus de la Influenza A/inmunología , Vacunas contra la Influenza/uso terapéutico , Gripe Humana/prevención & control , Neuraminidasa/inmunología , Vacunación/métodos , Administración Intranasal , Adulto , Anciano , Niño , Ensayo de Inmunoadsorción Enzimática , Humanos , Vacunas contra la Influenza/administración & dosificación , Gripe Humana/inmunología , Gripe Humana/metabolismo , Inyecciones Intramusculares , Mucosa Nasal/inmunología , Vacunas de Productos Inactivados/administración & dosificación , Vacunas de Productos Inactivados/uso terapéutico
14.
Harefuah ; 144(10): 689-91, 752, 2005 Oct.
Artículo en Hebreo | MEDLINE | ID: mdl-16281758

RESUMEN

BACKGROUND: Influenza is a common disease, especially in the winter season. Influenza virus causes symptoms of acute viral illness, development of complications and worsening of chronic diseases. Vaccination may prevent influenza illness. OBJECTIVES: 1. To determine the rate of vaccination against influenza. Furthermore, in the immunized patients--the study aimed to determine the efficiency of the vaccination. 2. To determine the incidence of influenza illness among patients admitted to a Department of Internal Medicine during the winter season. METHODS: We performed a prospective screening of 124 consecutive patients hospitalized in the Department of Internal Medicine during February 1998. Demographic parameters (age, sex) and admission diagnosis were collected. All patients were asked about influenza vaccination in the last fall. Blood samples were collected on admission and 10-30 days later. Three relevant virus subtypes were examined. Virus subtype was determined in all patients with evidence of influenza illness. RESULTS: Eleven patients (8.9%) were found with evidence of acute influenza illness at the time of hospitalization. The rate of patients with chronic pulmonary disease was higher in this group than in the group without evidence of influenza (p = 0.03). Forty-four out of 124 patients (35.5%) had been vaccinated against influenza. In the vaccinated patients, the protective antibody level for all 3 subtypes developed in 27.3% of cases and for one or two subtypes in 50% of cases. In 22.7% of patients no protective level of antibody was detected for any subtype. CONCLUSIONS: It is necessary to encourage vaccination in the high-risk group patient, especially those with chronic pulmonary disease. It is essential to develop new vaccination strategies and prophylactic antiviral medications.


Asunto(s)
Gripe Humana/epidemiología , Departamentos de Hospitales/estadística & datos numéricos , Humanos , Medicina Interna/estadística & datos numéricos , Israel/epidemiología , Estaciones del Año
15.
Oncolytic Virother ; 4: 49-62, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-27512670

RESUMEN

Newcastle disease virus (NDV) features a natural preference for replication in many tumor cells compared with normal cells. The observed antitumor effect of NDV appears to be a result of both selective killing of tumor cells and induction of immune responses. Genetic manipulations to change viral tropism and arming the virus with genes encoding for cytokines improved the oncolytic capacity of NDV. Several intracellular proteins in tumor cells, including antiapoptotic proteins (Livin) and oncogenic proteins (H-Ras), are relevant for the oncolytic activity of NDV. Defects in the interferon system, found in some tumor cells, also contribute to the oncolytic selectivity of NDV. Notwithstanding, NDV displays effective oncolytic activity in many tumor types, despite having intact interferon signaling. Taken together, several cellular systems appear to dictate the selective oncolytic activity of NDV. Some barriers, such as neutralizing antibodies elicited during NDV treatment and the extracellular matrix in tumor tissue appear to interfere with spread of NDV and reduce oncolysis. To further understand the oncolytic activity of NDV, we compared two NDV strains, ie, an attenuated virus (NDV-HUJ) and a pathogenic virus (NDV-MTH-68/H). Significant differences in amino acid sequence were noted in several viral proteins, including the fusion precursor (F0) glycoprotein, an important determinant of replication and pathogenicity. However, no difference in the oncolytic activity of the two strains was noted using human tumor tissues maintained as organ cultures or in mouse tumor models. To optimize virotherapy in clinical trials, we describe here a unique organ culture methodology, using a biopsy taken from a patient's tumor before treatment for ex vivo infection with NDV to determine the oncolytic potential on an individual basis. In conclusion, oncolytic NDV is an excellent candidate for cancer therapy, but more knowledge is needed to ensure success in clinical trials.

16.
Virology ; 485: 289-96, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26318261

RESUMEN

The initial interplay between human cytomegalovirus (HCMV) and innate tissue response in the human maternal-fetal interface, though crucial for determining the outcome of congenital HCMV infection, has remained unknown. We studied the innate response to HCMV within the milieu of the human decidua, the maternal aspect of the maternal-fetal interface, maintained ex vivo as an integral tissue. HCMV infection triggered a rapid and robust decidual-tissue innate immune response predominated by interferon (IFN)γ and IP-10 induction, dysregulating the decidual cytokine/chemokine environment in a distinctive fashion. The decidual-tissue response was already elicited during viral-tissue contact, and was not affected by neutralizing HCMV antibodies. Of note, IFNγ induction, reflecting immune-cell activation, was distinctive to the maternal decidua, and was not observed in concomitantly-infected placental (fetal) villi. Our studies in a clinically-relevant surrogate human model, provide a novel insight into the first-line decidual tissue response which could affect the outcome of congenital infection.


Asunto(s)
Infecciones por Citomegalovirus/inmunología , Citomegalovirus/inmunología , Interacciones Huésped-Patógeno/inmunología , Inmunidad Innata , Placenta/inmunología , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Citocinas/genética , Citocinas/metabolismo , Infecciones por Citomegalovirus/genética , Infecciones por Citomegalovirus/metabolismo , Infecciones por Citomegalovirus/virología , Decidua/inmunología , Decidua/metabolismo , Decidua/patología , Decidua/virología , Femenino , Expresión Génica , Heparina/metabolismo , Humanos , Mediadores de Inflamación/metabolismo , Placenta/metabolismo , Placenta/patología , Placenta/virología , Embarazo , Virión/inmunología
17.
Clin Infect Dis ; 39(4): 595-7, 2004 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-15356830

RESUMEN

In the summer of 2003 in Jerusalem, Israel, 23 children were hospitalized with influenza A virus (A/Fujian/411/02-like virus) infection. The majority were neonates and infants. Clinical manifestations included neonatal fever, bronchitis, bronchiolitis, and pneumonia, and outcomes were favorable. Continued surveillance between epidemic seasons could allow early recognition of influenza strains that will appear in the following influenza season.


Asunto(s)
Brotes de Enfermedades , Virus de la Influenza A/aislamiento & purificación , Gripe Humana/epidemiología , Niño , Niño Hospitalizado , Preescolar , Infecciones Comunitarias Adquiridas/diagnóstico , Humanos , Lactante , Recién Nacido , Israel/epidemiología
18.
Exp Gerontol ; 38(10): 1199-203, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-14580873

RESUMEN

Intramuscular (IM) influenza vaccines are about 50% effective in preventing respiratory illness among the elderly. The aim of this study was to identify factors associated with immune response to influenza vaccination among nursing home and community-residing elderly. 114 nursing home (NHE) and 62 community residing elderly (CE) were vaccinated with a commercial IM vaccine. Serum antibodies were evaluated by HIA, and the impact of subjects' clinical characteristics on seroconversion was determined. Factors that were associated with low seroconversion among NHE, included: type II diabetes [for B/Harbin: p=0.044, OR 0.12, (CI: 0.015-0.94)], and antibody titer prior to vaccination A/(H1N1): p=0.03, OR 2.38, (CI: 1.09-5.22); A/(H3N2): p=0.015, OR 2.68 (CI: 1.22-5.92), B/Harbin: p=0.057, OR 4.46 (CI: 0.96-20.85)]. Factors that were associated with lower seroconversion CE elderly, included older age [A/(H1N1): p=0.008, OR 0.846, (CI 0.75-0.96), B/Harbin: p=0.016, OR 0.812, (CI:0.69-0.96)], and antibody titer prior to vaccination A/(H1N1): p=0.029, OR 4.08, (CI: 1.16-14.37); A/(H3N2): p<0.0001, OR 11.495 (CI: 3.18-41.55)]. There was no significant difference in seroconversion between nursing home residing elderly and community elderly. We conclude that Type-II diabetes and antibody titer>1:40 prior to vaccination are associated with reduced response to the influenza vaccination in nursing home elderly.


Asunto(s)
Anticuerpos Antivirales/biosíntesis , Virus de la Influenza A/inmunología , Virus de la Influenza B/inmunología , Vacunas contra la Influenza/inmunología , Anciano , Anciano de 80 o más Años , Servicios de Salud Comunitaria , Diabetes Mellitus Tipo 2/inmunología , Femenino , Evaluación Geriátrica , Pruebas de Inhibición de Hemaglutinación , Hogares para Ancianos , Humanos , Gripe Humana/prevención & control , Modelos Logísticos , Masculino , Persona de Mediana Edad , Casas de Salud , Características de la Residencia , Factores de Riesgo
19.
Br J Gen Pract ; 52(479): 481-2, 2002 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12051213

RESUMEN

Herpetic gingivo-stomatitis is a common, often painful affliction of young children. Recently, aciclovir treatment has been found to be effective in hospital-referred cases, an approach limited in general practice where laboratory diagnosis is invariably impractical. This study demonstrated that 49 out of 63 clinical diagnoses of herpetic gingivo-stomatitis [PPV = 78%] made by 27 GPs were subsequently validated by laboratory virus culture. This suggests that herpetic gingivo-stomatitis might be treated with aciclovir in general practice on the basis of a clinical diagnosis alone.


Asunto(s)
Competencia Clínica , Medicina Familiar y Comunitaria/normas , Estomatitis Herpética/diagnóstico , Femenino , Humanos , Lactante , Israel , Masculino , Estudios Prospectivos
20.
Pediatr Infect Dis J ; 30(6): 491-4, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21248658

RESUMEN

BACKGROUND: Data on the immunogenicity of the influenza vaccine in children after liver transplantation are sparse. Our study aims to evaluate the response of such patients to the trivalent influenza vaccine, administered by different protocols in 2 influenza seasons. METHODS: Children attending the Liver Transplantation Unit of a tertiary care medical center were prospectively recruited and immunized with the inactivated subvirion influenza vaccine during the influenza seasons of 2004/2005 (1 dose, n = 18) and 2005/2006 (2 doses 4-6 weeks apart, n = 32). Antibodies were measured by hemagglutination inhibition assay. Immunity was defined as a titer of ≥1:40, and response was defined as a ≥4-fold increase in antibody titer from baseline. RESULTS: In 2004/2005, the proportions of patients with protective antibodies were similar before and after 1 dose of vaccine. We found significant difference after the first dose for the A/H3N2 Wisconsin strain (43.2% vs. 70.3%, P = 0.003) and B/Malaysia strains (8.1% vs. 35.1%, P = 0.003) and for A/H1N1 New Caledonia strain (48.6% vs. 64.9% vs. 75%, P = 0.08, 0.005, respectively) after the second dose in 2005/2006 season. In 2004/2005, geometric mean titers rose significantly (P = 0.03) for the A/H3N2 New York strain; in 2005/2006, geometric mean titers for A/H3N2 New York and B/Malaysia increased after the first dose and for A/H1N1 New Caledonia after the second dose. Antibody titers were unrelated to age at transplantation, time from transplantation, and number of immunosuppressive drugs used. No serious vaccine-related events were documented. CONCLUSIONS: Liver-transplanted children respond to influenza vaccination. For some strains, the response is similar to that reported for healthy children. A second vaccine dose yielded no statistically significant benefit.


Asunto(s)
Anticuerpos Antivirales/sangre , Vacunas contra la Influenza/inmunología , Gripe Humana/prevención & control , Trasplante de Hígado/inmunología , Trasplante , Adolescente , Niño , Preescolar , Femenino , Pruebas de Inhibición de Hemaglutinación , Humanos , Lactante , Vacunas contra la Influenza/administración & dosificación , Malasia , Masculino , Estudios Prospectivos , Vacunas de Productos Inactivados/administración & dosificación , Vacunas de Productos Inactivados/inmunología , Vacunas de Subunidad/administración & dosificación , Vacunas de Subunidad/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA