Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
1.
Diagn Microbiol Infect Dis ; 99(3): 115244, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33253961

RESUMO

Nosocomial virus infections cause significant morbidity and mortality. Besides influenza viruses, the disease burden of parainfluenza virus type 3 (PIV-3) is comparatively high among hospitalized patients and severe disease courses can occur. PIV-3 showed the highest rates of nosocomial infections of a panel of respiratory viruses. Therefore, a retrospective observational study was conducted among patients with either PIV-3 or influenza viruses, which served as reference pathogen. The aim was to compare the seasonal dynamics and clinical characteristics of nosocomial infections with these highly transmittable viruses. Nosocomial infection occurred in 15.8% (n = 177) of all influenza cases, mainly in the first half of a season. About 24.3% (n = 104) of the PIV-3 cases were nosocomial and occurred mainly in the second half of a season. Both nosocomial rates of influenza and nosocomial rates of PIV-3 varied between the seasons. Community acquired and nosocomial cases differed in underlying medical conditions and immunosuppression. Knowledge of the baseline rates of nosocomial infections could contribute to the implementation of appropriate infection control measures.


Assuntos
Infecção Hospitalar/epidemiologia , Infecção Hospitalar/virologia , Hospitais Universitários/estatística & dados numéricos , Influenza Humana/epidemiologia , Infecções por Respirovirus/epidemiologia , Adolescente , Adulto , Criança , Pré-Escolar , Infecção Hospitalar/transmissão , Feminino , Alemanha/epidemiologia , Humanos , Lactente , Influenza Humana/transmissão , Masculino , Pessoa de Meia-Idade , Orthomyxoviridae/patogenicidade , Vírus da Parainfluenza 3 Humana/patogenicidade , Infecções Respiratórias/virologia , Infecções por Respirovirus/transmissão , Estudos Retrospectivos , Estações do Ano , Adulto Jovem
2.
J Hosp Infect ; 103(3): 349-353, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31356855

RESUMO

A hospital outbreak of human parainfluenza virus type 3 (HPIV-3) in haematologic oncology patients is described in 12 patients over a four-week period. Exposure histories and molecular analysis of HPIV-3 isolates suggest that both community-acquired and nosocomially transmitted infections occurred during this outbreak. Molecular analysis of HPIV-3 isolates indicated that a chain of transmission occurred among multiple patients in an oncology ward. This transmission was later determined to be associated with the movement of fomites, visitors, and activities in the unit. The infection prevention team stopped nosocomial spread of HPIV-3 through interventions including advanced cleaning procedures.


Assuntos
Infecção Hospitalar/epidemiologia , Infecção Hospitalar/virologia , Surtos de Doenças , Vírus da Parainfluenza 3 Humana/classificação , Vírus da Parainfluenza 3 Humana/genética , Infecções por Respirovirus/epidemiologia , Infecções por Respirovirus/virologia , Infecção Hospitalar/transmissão , Transmissão de Doença Infecciosa/prevenção & controle , Técnicas de Genotipagem , Neoplasias Hematológicas/complicações , Humanos , Controle de Infecções/métodos , Epidemiologia Molecular , Vírus da Parainfluenza 3 Humana/isolamento & purificação , Infecções por Respirovirus/transmissão
3.
Food Environ Virol ; 10(2): 133-140, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29196954

RESUMO

The aim of this study was to evaluate the potential role of office fomites in respiratory (human parainfluenza virus 1-HPIV1, human parainfluenza virus 3-HPIV3) and enteric (norovirus GI-NoV GI, norovirus GII-NoV GII) viruses transmission by assessing the occurrence of these viruses on surfaces in office buildings. Between 2016 and 2017, a total of 130 surfaces from open-space and non-open-space rooms in office buildings located in one city were evaluated for HPIV1, HPIV3, NoV GI, and NoV GII viral RNA presence. Detection of viruses was performed by RT-qPCR method. Study revealed 27 positive samples, among them 59.3% were HPIV3-positive, 25.9% HPIV1-positive, and 14.8% NoV GII-positive. All tested surfaces were NoV GI-negative. Statistical analysis of obtained data showed that the surfaces of office equipment including computer keyboards and mice, telephones, and desktops were significantly more contaminated with respiratory viruses than the surfaces of building equipment elements such as door handles, light switches, or ventilation tracts (χ 2 p = 0.006; Fisher's Exact p = 0.004). All examined surfaces were significantly more contaminated with HPIVs than NoVs (χ 2 p = 0.002; Fisher's Exact p = 0.003). Office fomites in open-space rooms were more often contaminated with HPIVs than with NoVs (χ 2 p = 0.016; Fisher's Exact p = 0.013). The highest average concentration of HPIVs RNA copies was observed on telephones (1.66 × 102 copies/100 cm2), while NoVs on the light switches (1.40 × 102 copies/100 cm2). However, the Kruskal-Wallis test did not show statistically significant differences in concentration levels of viral RNA copies on surfaces between the all tested samples. This study unequivocally showed that individuals in office environment may have contact with both respiratory and enteric viral particles present on frequently touched surfaces.


Assuntos
Infecções por Caliciviridae/virologia , Fômites/virologia , Norovirus/isolamento & purificação , Vírus da Parainfluenza 1 Humana/isolamento & purificação , Vírus da Parainfluenza 3 Humana/isolamento & purificação , Infecções por Respirovirus/virologia , Infecções por Caliciviridae/transmissão , Genoma Viral/genética , Humanos , Norovirus/genética , Vírus da Parainfluenza 1 Humana/genética , Vírus da Parainfluenza 3 Humana/genética , Prevalência , RNA Viral/genética , RNA Viral/isolamento & purificação , Infecções por Respirovirus/transmissão , Reação em Cadeia da Polimerase Via Transcriptase Reversa
4.
J Clin Virol ; 92: 53-55, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28531552

RESUMO

BACKGROUND: Respiratory viral infections are a significant problem in patients with hematologic malignancies. We report a cluster of HPIV 3 infections in our myeloma patients, and describe the utility of next generation sequencing (NGS) to identify transmission linkages which can assist in infection prevention. OBJECTIVES: To evaluate the utility of NGS to track respiratory viral infection outbreaks and delineate between community acquired and nosocomial infections in our cancer units. STUDY DESIGN: Retrospective chart review conducted at a single site. All patients diagnosed with multiple myeloma who developed symptoms suggestive of upper respiratory tract infection (URTI) or lower respiratory tract infection (LRTI) along with a respiratory viral panel (RVP) test positive for HPIV 3 between April 1, 2016, to June 30, 2016, were included. Sequencing was performed on the Illumina MiSeq™. To gain understanding regarding community strains of HPIV 3 during the same season, we also performed NGS on HPIV3 strains isolated from pediatric cases. RESULTS: We saw a cluster of 13 cases of HPIV3 infections in the myeloma unit. Using standard epidemiologic criteria, 3 cases were considered community acquired, 7 cases developed infection during treatment in the cancer infusion center, while an additional 3 developed infections during hospital stay. Seven patients required hospitalization for a median duration of 20days. NGS enabled sensitive discrimination of the relatedness of the isolates obtained during the outbreak and provided evidence for source of transmission. Two hospital onset infections could be tracked to an index case; the genome sequences of HPIV 3 strains from these 3 patients only differed by a single nucleotide. CONCLUSIONS: NGS offers a significantly higher discriminatory value as an epidemiologic tool, and can be used to gather real-time information and identification of transmission linkages to assist in infection prevention in immunocompromised patients.


Assuntos
Sequenciamento de Nucleotídeos em Larga Escala , Hospedeiro Imunocomprometido , Mieloma Múltiplo/complicações , Vírus da Parainfluenza 3 Humana/genética , Infecções por Respirovirus/prevenção & controle , Criança , Infecção Hospitalar/epidemiologia , Infecção Hospitalar/prevenção & controle , Infecção Hospitalar/virologia , Feminino , Genoma Viral , Humanos , Masculino , Infecções Respiratórias/epidemiologia , Infecções Respiratórias/virologia , Infecções por Respirovirus/epidemiologia , Infecções por Respirovirus/transmissão , Infecções por Respirovirus/virologia , Estudos Retrospectivos
5.
Transpl Infect Dis ; 16(1): 165-9, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24289829

RESUMO

Respiratory viral infections are frequent causes of morbidity in transplant patients. We screened symptomatic adult transplant recipients for respiratory viruses in a cohort of patients attending a referral medical center in Brazil. The duration of viral shedding and the prevalence of viral codetections were also determined. During a 1-year period (2011-2012), swabs were obtained from 50 patients. An in-house polymerase chain reaction panel designed to detect 10 viruses was used. Viruses were identified in 19 (38%) patients, particularly parainfluenza III (32%) and the respiratory syncytial virus (20%); multiple viruses were identified in 26% of patients. Prolonged viral shedding was observed with 60% of individuals excreting viruses for >10 days. The clinical and epidemiologic relevance of prolonged viral shedding remains to be determined.


Assuntos
Rejeição de Enxerto/prevenção & controle , Hospedeiro Imunocomprometido , Imunossupressores/uso terapêutico , Transplante de Órgãos , Infecções Respiratórias/transmissão , Viroses/transmissão , Eliminação de Partículas Virais , Adulto , Idoso , Estudos de Coortes , Coinfecção , Feminino , Humanos , Vírus da Influenza A/genética , Vírus da Influenza B/genética , Influenza Humana/imunologia , Influenza Humana/transmissão , Transplante de Rim , Transplante de Fígado , Transplante de Pulmão , Masculino , Pessoa de Meia-Idade , Vírus da Parainfluenza 1 Humana/genética , Vírus da Parainfluenza 2 Humana/genética , Vírus da Parainfluenza 3 Humana/genética , Estudos Prospectivos , Reação em Cadeia da Polimerase em Tempo Real , Infecções por Vírus Respiratório Sincicial/imunologia , Infecções por Vírus Respiratório Sincicial/transmissão , Vírus Sinciciais Respiratórios/genética , Infecções Respiratórias/imunologia , Infecções Respiratórias/virologia , Infecções por Respirovirus/imunologia , Infecções por Respirovirus/transmissão , Fatores de Tempo , Viroses/imunologia , Viroses/virologia , Adulto Jovem
6.
PLoS Pathog ; 9(11): e1003786, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24278024

RESUMO

Little is known about how the mode of respiratory virus transmission determines the dynamics of primary infection and protection from reinfection. Using non-invasive imaging of murine parainfluenza virus 1 (Sendai virus) in living mice, we determined the frequency, timing, dynamics, and virulence of primary infection after contact and airborne transmission, as well as the tropism and magnitude of reinfection after subsequent challenge. Contact transmission of Sendai virus was 100% efficient, phenotypically uniform, initiated and grew to robust levels in the upper respiratory tract (URT), later spread to the lungs, grew to a lower level in the lungs than the URT, and protected from reinfection completely in the URT yet only partially in the lungs. Airborne transmission through 7.6-cm and 15.2-cm separations between donor and recipient mice was 86%-100% efficient. The dynamics of primary infection after airborne transmission varied between individual mice and included the following categories: (a) non-productive transmission, (b) tracheal dominant, (c) tracheal initiated yet respiratory disseminated, and (d) nasopharyngeal initiated yet respiratory disseminated. Any previous exposure to Sendai virus infection protected from mortality and severe morbidity after lethal challenge. Furthermore, a higher level of primary infection in a given respiratory tissue (nasopharynx, trachea, or lungs) was inversely correlated with the level of reinfection in that same tissue. Overall, the mode of transmission determined the dynamics and tropism of primary infection, which in turn governed the level of seroconversion and protection from reinfection. These data are the first description of the dynamics of respiratory virus infection and protection from reinfection throughout the respiratory tracts of living animals after airborne transmission. This work provides a basis for understanding parainfluenza virus transmission and protective immunity and for developing novel vaccines and non-pharmaceutical interventions.


Assuntos
Sistema Respiratório , Infecções por Respirovirus , Vírus Sendai , Tropismo Viral/imunologia , Animais , Masculino , Camundongos , Sistema Respiratório/imunologia , Sistema Respiratório/patologia , Sistema Respiratório/virologia , Infecções por Respirovirus/imunologia , Infecções por Respirovirus/patologia , Infecções por Respirovirus/prevenção & controle , Infecções por Respirovirus/transmissão , Vírus Sendai/imunologia , Vírus Sendai/metabolismo , Vírus Sendai/patogenicidade
7.
PLoS Pathog ; 7(7): e1002134, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21750677

RESUMO

The parainfluenza viruses (PIVs) are highly contagious respiratory paramyxoviruses and a leading cause of lower respiratory tract (LRT) disease. Since no vaccines or antivirals exist, non-pharmaceutical interventions are the only means of control for these pathogens. Here we used bioluminescence imaging to visualize the spatial and temporal progression of murine PIV1 (Sendai virus) infection in living mice after intranasal inoculation or exposure by contact. A non-attenuated luciferase reporter virus (rSeV-luc(M-F*)) that expressed high levels of luciferase yet was phenotypically similar to wild-type Sendai virus in vitro and in vivo was generated to allow visualization. After direct intranasal inoculation, we unexpectedly observed that the upper respiratory tract (URT) and trachea supported robust infection under conditions that result in little infection or pathology in the lungs including a low inoculum of virus, an attenuated virus, and strains of mice genetically resistant to lung infection. The high permissivity of the URT and trachea to infection resulted in 100% transmission to naïve contact recipients, even after low-dose (70 PFU) inoculation of genetically resistant BALB/c donor mice. The timing of transmission was consistent with the timing of high viral titers in the URT and trachea of donor animals but was independent of the levels of infection in the lungs of donors. The data therefore reveals a disconnect between transmissibility, which is associated with infection in the URT, and pathogenesis, which arises from infection in the lungs and the immune response. Natural infection after transmission was universally robust in the URT and trachea yet limited in the lungs, inducing protective immunity without weight loss even in genetically susceptible 129/SvJ mice. Overall, these results reveal a dichotomy between PIV infection in the URT and trachea versus the lungs and define a new model for studies of pathogenesis, development of live virus vaccines, and testing of antiviral therapies.


Assuntos
Pulmão/virologia , Infecções por Respirovirus/transmissão , Doenças dos Roedores/transmissão , Vírus Sendai/fisiologia , Traqueia/virologia , Administração Intranasal , Animais , Líquido da Lavagem Broncoalveolar/citologia , Líquido da Lavagem Broncoalveolar/virologia , Linhagem Celular , Progressão da Doença , Luciferases/metabolismo , Medições Luminescentes , Pulmão/patologia , Camundongos , Camundongos Endogâmicos BALB C , Sistema Respiratório , Infecções por Respirovirus/imunologia , Infecções por Respirovirus/patologia , Doenças dos Roedores/imunologia , Doenças dos Roedores/patologia , Traqueia/patologia
8.
Transpl Infect Dis ; 13(4): 433-7, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21466639
9.
Transbound Emerg Dis ; 58(1): 37-43, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21062425

RESUMO

A longitudinal study was conducted to investigate the presence of antibodies (Ab) to Rotavirus (RV), Parainfluenza-3 virus (PI-3), Bovine Herpesvirus-1 (BoHV-1), Bovine Viral Diarrhoea virus (BVDV-1) and Bluetongue virus (BTV) in eleven guanaco's crias (chulengos) relocated from Rio Negro to Buenos Aires Province (Argentina) and reared in captivity for a year in an experimental field. Serum samples were collected periodically to detect the evidence of viral infections. Faecal samples were collected to investigate RV shedding. We detected the evidence of Ab to RV from the beginning of the experience, suggesting the presence of maternal Ab against the virus. RV infection was detected in seven of the eleven chulengos, by seroconversion (4), virus shedding in stools (1) or both (2). In all cases, the RV strain was typed as [P1]G8, the same G/P type combination detected in captive chulengos with acute diarrhoea sampled in Rio Negro, in 2001. In contrast, we could not detect antibodies against PI-3, BoHV-1, BVDV or BT in any of initial samples. No Abs against BoHV-1, BVDV or BTV were detected in the chulengos throughout the study. However, all the chulengos became asymptomatically seropositive to PI-3 by the 7 month after arrival. This study suggest that wild-born guanacos raised in captivity can be relatively susceptible to common livestock viral infections, such as RV and PI-3, which are easily spread among chulengos.


Assuntos
Anticorpos Antivirais/sangue , Camelídeos Americanos/virologia , Vírus da Parainfluenza 3 Bovina/isolamento & purificação , Infecções por Respirovirus/veterinária , Infecções por Rotavirus/veterinária , Rotavirus/isolamento & purificação , Animais , Anticorpos Antivirais/análise , Argentina/epidemiologia , Camelídeos Americanos/sangue , Camelídeos Americanos/imunologia , Diarreia/epidemiologia , Diarreia/veterinária , Diarreia/virologia , Suscetibilidade a Doenças , Fezes/virologia , Estudos Longitudinais , Masculino , Vírus da Parainfluenza 3 Bovina/imunologia , Infecções por Respirovirus/epidemiologia , Infecções por Respirovirus/transmissão , Rotavirus/classificação , Rotavirus/imunologia , Infecções por Rotavirus/sangue , Infecções por Rotavirus/epidemiologia , Infecções por Rotavirus/transmissão , Estudos Soroepidemiológicos , Eliminação de Partículas Virais
11.
Biochem Biophys Res Commun ; 355(1): 129-35, 2007 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-17292867

RESUMO

Dendritic cell (DC)-based immunotherapy has been clinically evaluated, however, still requires modification to improve its outcomes. We previously demonstrated that DCs activated by replication competent recombinant Sendai virus (SeV) showed dramatic efficacy over that seen in use of current DC vaccine for immunotherapy against malignancies; however, application of replication-deficient vector is more relevant in clinical setting. We here show that F-gene-deleted non-transmissible Sendai virus (SeV/dF)-activated DCs (DCs/SeV/dF) has strong antitumor effects against murine SCCVII tumor, that was well-known as a less immunogenic cell line. SeV/dF shows high transfection efficiency to DCs and leads them to upregulate costimulatory molecules. Intratumoral injection of DCs/SeV/dF resulted in a marked and representative inhibition of the tumor, even when the tumors were well-vascularized. This is the first demonstration that non-transmissible SeV vector, SeV/dF, could be a DC-activator; DC/SeV/dF-based cancer immunotherapy may, therefore, warrant further investigation.


Assuntos
Adjuvantes Imunológicos/uso terapêutico , Células Dendríticas/imunologia , Células Dendríticas/virologia , Imunoterapia/métodos , Infecções por Respirovirus/imunologia , Vírus Sendai/imunologia , Animais , Células da Medula Óssea/imunologia , Carcinoma de Células Escamosas/imunologia , Feminino , Camundongos , Camundongos Endogâmicos C3H , Infecções por Respirovirus/transmissão
12.
Am J Trop Med Hyg ; 75(4): 716-9, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17038700

RESUMO

Performing monkeys, a common phenomena in Asia, occupy a unique urban niche that comprises a number of factors influencing the likelihood of cross-species transmission of pathogens. Here we present the first documented evidence of exposure to measles, rubella, and parainfluenza in a population of performing monkeys. Evidence of exposure to these endemic human respiratory viruses in the performing monkeys confirms human-to-primate transmission and suggests the possibility of primate-to-human transmission. Urban animal markets, the likely source of these performing monkeys, may represent an environment conducive to the mixing of animals and pathogens, making these monkeys a potential conduit for infectious agents passing from a variety of animals found in animal markets to humans. The potential significance of these results to human public health and the unique contexts of disease transmission associated with the urban ecology of the performance monkeys are discussed. Given the level of overseas travel, this potential threat is not confined solely to Asia.


Assuntos
Macaca fascicularis , Doenças dos Macacos/transmissão , Infecções por Paramyxoviridae/transmissão , Rubéola (Sarampo Alemão)/transmissão , Zoonoses/transmissão , Animais , Animais Domésticos , Anticorpos Antivirais/sangue , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Indonésia , Masculino , Sarampo/imunologia , Sarampo/transmissão , Vírus do Sarampo/imunologia , Doenças dos Macacos/imunologia , Vírus da Parainfluenza 2 Humana/imunologia , Vírus da Parainfluenza 3 Humana/imunologia , Infecções por Paramyxoviridae/imunologia , Infecções por Respirovirus/imunologia , Infecções por Respirovirus/transmissão , Rubéola (Sarampo Alemão)/imunologia , Vírus da Rubéola/imunologia , Saúde da População Urbana
13.
Vaccine ; 24(13): 2432-9, 2006 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-16406170

RESUMO

BACKGROUND: This study examined the transmissibility between young children of an intranasally administered live attenuated human parainfluenza virus type 3 (HPIV3)-cp45 vaccine candidate. METHODS: Eighty subjects were enrolled in playgroups among whom there was at least one infected vaccinee in close contact with a seronegative placebo recipient over 21 days without a confounding infection with wtHPIV3. Following vaccination viral cultures were obtained on nine occasions to detect shedding and transmission of HPIV3cp45. Serum antibody titers were measured before and 7 weeks after vaccination. RESULTS: No child fulfilled the criteria for transmission of HPIV3cp45 giving a risk of transmission of 0.04 (95% CI 0.01-0.19), hence establishing that HPIV3cp45 is less infectious than wtHPIV3 and risk of transmission is not a limitation to further clinical development of this vaccine candidate.


Assuntos
Vírus da Parainfluenza 3 Humana/imunologia , Infecções por Respirovirus/transmissão , Vacinas Virais/imunologia , Anticorpos Antivirais/sangue , Pré-Escolar , Método Duplo-Cego , Ensaio de Imunoadsorção Enzimática , Humanos , Lactente , Vírus da Parainfluenza 3 Humana/patogenicidade , Vacinação , Vacinas Atenuadas/imunologia
15.
Biol Blood Marrow Transplant ; 10(1): 58-64, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14752780

RESUMO

Human parainfluenza virus type 3 (hPIV3) infections cause considerable morbidity and mortality after stem cell transplantation, and inpatient nosocomial outbreaks are common. From September 1998 to July 1999, 93 stem cell transplantation recipients at our institution contracted hPIV3, of which 66 (71%) were being followed up in our outpatient department (OPD). The peak incidence was in September and October, when 39 cases were identified; thereafter, hPIV3 incidence decreased to approximately 5 cases per month. Nucleotide sequences (778 nucleotides from variable regions of the hemagglutinin-neuraminidase gene) from 46 patient and 8 community hPIV3 isolates were compared to determine epidemiologic relatedness. Sequence analysis of OPD isolates revealed that 18 of 19 isolates from September and October and 11 of 15 isolates from November 1998 to July 1999 were genetically similar. In contrast, 2 of 3 community isolates from September and October and 0 of 5 from November to July were linked to this cluster. Symptomatic surveillance and isolation were ineffective in terminating the outbreak, suggesting asymptomatic shedding among patients, staff, or visitors or viral persistence on environmental surfaces as possible explanations. The concept of nosocomial transmission should be expanded to include the OPD for immunosuppressed patients.


Assuntos
Surtos de Doenças , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Vírus da Parainfluenza 3 Humana , Infecções por Respirovirus/epidemiologia , Infecção Hospitalar , Departamentos Hospitalares , Humanos , Hospedeiro Imunocomprometido , Incidência , Epidemiologia Molecular , Vírus da Parainfluenza 3 Humana/isolamento & purificação , RNA Viral/análise , Infecções por Respirovirus/etiologia , Infecções por Respirovirus/transmissão , Análise de Sequência de RNA , Washington/epidemiologia
16.
Curr Top Microbiol Immunol ; 278: 125-83, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12934944

RESUMO

Porcine organs, cells and tissues provide a viable source of transplants in humans, though there is some concern of public health risk from adaptation of swine infectious agents in humans. Limited information is available on the public health risk of many exogenous swine viruses, and reliable and rapid diagnostic tests are available for only a few of these. The ability of several porcine viruses to cause transplacental fetal infection (parvoviruses, circoviruses, and arteriviruses), emergence or recognition of several new porcine viruses during the last two decades (porcine circovirus, arterivirus, paramyxoviruses, herpesviruses, and porcine respiratory coronavirus) and the immunosuppressed state of the transplant recipients increases the xenozoonoses risk of humans to porcine viruses through transplantation. Much of this risk can be eliminated with vigilance and sustained monitoring along with a better understanding of pathogenesis and development of better diagnostic tests. In this review we present information on selected exogenous viruses, highlighting their characteristics, pathogenesis of viral infections in swine, methods for their detection, and the potential xenozoonoses risk they present. Emphasis has been given in this review to swine influenza virus, paramyxovirus (Nipah virus, Menagle virus, LaPiedad paramyxovirus, porcine paramyxovirus), arterivirus (porcine reproductive and respiratory syndrome virus) and circovirus as either they represent new swine viruses or present the greatest risk. We have also presented information on porcine parvovirus, Japanese encephalitis virus, encephalomyocarditis virus, herpesviruses (pseudorabies virus, porcine lymphotropic herpesvirus, porcine cytomegalovirus), coronaviruses (TGEV, PRCV, HEV, PEDV) and adenovirus. The potential of swine viruses to infect humans needs to be assessed in vitro and in vivo and rapid and more reliable diagnostic methods need to be developed to assure safe supply of porcine tissues and cells for xenotransplantation.


Assuntos
Doenças dos Suínos/transmissão , Suínos/virologia , Transplante Heterólogo/efeitos adversos , Viroses/veterinária , Zoonoses/transmissão , Animais , Infecções por Arterivirus/transmissão , Infecções por Arterivirus/veterinária , Infecções por Circoviridae/transmissão , Infecções por Circoviridae/veterinária , Infecções por Herpesviridae/transmissão , Infecções por Herpesviridae/veterinária , Humanos , Infecções por Orthomyxoviridae/transmissão , Infecções por Orthomyxoviridae/veterinária , Infecções por Respirovirus/transmissão , Infecções por Respirovirus/veterinária , Doenças dos Suínos/virologia , Viroses/transmissão
17.
Comp Med ; 53(2): 173-7, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12784851

RESUMO

Sendai virus may induce acute respiratory tract disease in laboratory mice and is a common contaminant of biological materials. Pneumonia virus of mice (PVM) also infects the respiratory tract and, like Sendai virus, may induce a persistent wasting disease syndrome in immunodeficient mice. Reverse transcriptase-polymerase chain reaction (RT-PCR) assays have proven useful for detection of Sendai virus and PVM immunodeficient animals and contaminated biomaterials. Fluorogenic nuclease RT-PCR assays (fnRT-PCR) combine RT-PCR with an internal fluorogenic hybridization probe, thereby potentially enhancing specificity and eliminating post-PCR processing. Therefore, fnRT-PCR assays specific for Sendai virus and PVM were developed by targeting primer andprobe sequences to unique regions of the Sendai virus nucleocapsid (NP) gene and the PVM attachment (G) gene, respectively. The Sendai virus and PVM fnRT-PCR assays detected only Sendai virusand PVM , respectively. Neither assay detected other viruses of the family Paramyxoviridae or other RNA viruses that naturally infect rodents. The fnRT-PCR assays detected as little as 10 fg of Sendai virus RNA and one picogram of PVM RNA, respectively, andthe Sendai virus fnRT-PCR assay had comparable sensitivity when directly compared with the mouse antibody production test. The fnRT-PCR assays were also able to detect viral RNA in respiratory tract tissues and cage swipe specimens collected from experimentally inoculated C.B-17 severe combined immunodeficient mice, but did not detect viral RNA in age- and strain-matched mock-infected mice. In conclusion, these fnRT-PCR assays offer potentially high-throughput diagnostic assays to detect Sendai virus and PVM in immunodeficient mice, and to detect Sendai virus in contaminated biological materials.


Assuntos
Vírus da Pneumonia Murina/isolamento & purificação , Infecções por Pneumovirus/veterinária , Infecções por Respirovirus/veterinária , Reação em Cadeia da Polimerase Via Transcriptase Reversa/veterinária , Vírus Sendai/isolamento & purificação , Animais , Corantes Fluorescentes , Hospedeiro Imunocomprometido , Camundongos , Camundongos Endogâmicos ICR , Camundongos SCID , Vírus da Pneumonia Murina/genética , Vírus da Pneumonia Murina/patogenicidade , Infecções por Pneumovirus/transmissão , Infecções por Pneumovirus/virologia , RNA Viral/análise , Infecções por Respirovirus/transmissão , Infecções por Respirovirus/virologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Vírus Sendai/genética , Vírus Sendai/patogenicidade , Sensibilidade e Especificidade , Imunodeficiência Combinada Severa/veterinária , Imunodeficiência Combinada Severa/virologia
18.
Contemp Top Lab Anim Sci ; 41(4): 53-7, 2002 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12109899

RESUMO

In searching for the cause of experimental variations in respiratory research data, serology revealed the prevalence of antibodies against parainfluenza virus type 3 (PIV 3) in guinea pigs. The aim of the present study was to explore the transmission rate, course, and kinetics of enzootic PIV 3 infection in guinea pig breeding units. In the first part of the study, blood samples to be analyzed for PIV 3 antibodies were collected from guinea pigs of a PIV 3-positive breeding colony at different times after birth. In the same breeding unit, 6 of 12 2-week-old guinea pigs were relocated and separately housed. The PIV 3 serum antibody titers of the two groups were compared at various times from birth to 13 weeks after birth. In the second part of the study, the spread of infectious virus and virus persistence were explored by housing seronegative sentinel animals together with 2- to 3-week-old guinea pigs from three different PIV 3-positive breeding units. The guinea pigs remaining in the breeding colony as well as those removed and housed separately showed declining serum antibody titers for about 1 month after birth, thereafter the titers were stable until about 8 weeks after birth. Five weeks later, the mean antibody titer of the guinea pigs remaining in the breeding colony had increased to a markedly higher level than that of the relocated, separately housed guinea pigs. Seroconversion was demonstrated in 7 of the 14 sentinels housed with the 2- to 3-week-old guinea pigs from PIV 3-positive breeding units. Sentinels housed together with PIV 3-positive guinea pigs 24 weeks after the start of the experiment did not seroconvert. We conclude that young guinea pigs born to PIV 3-positive mothers were protected by maternal immunity against infection with PIV 3 during their first 14 days of life. The guinea pig offspring became infected during the period from about 2 weeks until 8 weeks after birth, as demonstrated by seroconversion of sentinel animals and an increasing mean antibody titer seen beyond 8 weeks of age. The study did not reveal any indication of virus persistence or prolonged carrier status.


Assuntos
Criação de Animais Domésticos/métodos , Troca Materno-Fetal , Vírus da Parainfluenza 3 Humana , Hipersensibilidade Respiratória/veterinária , Infecções por Respirovirus/veterinária , Animais , Animais Recém-Nascidos , Anticorpos Antivirais/sangue , Feminino , Cobaias , Masculino , Troca Materno-Fetal/imunologia , Gravidez , Hipersensibilidade Respiratória/sangue , Infecções por Respirovirus/sangue , Infecções por Respirovirus/transmissão , Organismos Livres de Patógenos Específicos , Fatores de Tempo
19.
J Gen Virol ; 83(Pt 6): 1377-1386, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12029153

RESUMO

An efficient antigen expression system using a recombinant Sendai virus (SeV) has been established recently and its potential to induce resistance against immunodeficiency virus infections in macaques has been shown. SeV replication has been well characterized in mice, the natural host, but not in primates, including humans. Here, primary SeV replication was investigated in macaques. After intranasal immunization with a recombinant SeV expressing simian immunodeficiency virus Gag protein, SeV-Gag, robust gag expression was observed in the nasal mucosa and much lower but significant levels of gag expression were observed in the local retropharyngeal and submandibular lymph nodes (LN). Expression peaked within a week and lasted at least up to 13 days after immunization. SeV-Gag was isolated from nasal swabs consistently at day 4 but not at all at day 13. Gag expression was undetectable in the lung as well as in remote lymphoid tissues, such as the thymus, spleen and inguinal LN, indicating that the spread of the virus was more restricted in macaques than in mice. SeV-specific T cells were detectable in SeV-immunized macaques at day 7. Finally, no naive macaques showed significant levels of anti-SeV antibodies in the plasma, even after living in a cage together with an acutely SeV-infected macaque for 5 weeks, indicating that SeV transmission from SeV-infected macaques to naive ones was inefficient. None of the SeV-immunized macaques displayed appreciable clinical manifestations. These results support the idea that this system may be used safely in primates, including humans.


Assuntos
Vetores Genéticos/fisiologia , Macaca/virologia , Vírus Sendai/fisiologia , Replicação Viral , Animais , Anticorpos Antivirais/sangue , Produtos do Gene gag/análise , Produtos do Gene gag/biossíntese , Produtos do Gene gag/genética , Vetores Genéticos/genética , Imunização , Linfonodos/metabolismo , Macaca/sangue , Macaca fascicularis , Macaca mulatta , Mucosa Nasal/virologia , Recombinação Genética , Infecções por Respirovirus/transmissão , Vírus Sendai/genética , Vírus Sendai/imunologia , Linfócitos T/imunologia , Vacinas Sintéticas/virologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...