Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Australas Emerg Care ; 25(4): 289-295, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-35153180

RESUMEN

BACKGROUND: Emergency Departments around the world are under increasing pressure due to rising demand. In the United Kingdom ambulatory emergency care has been demonstrated to deliver safe and effective care for emergency patients and has now become an accepted treatment modality. METHODS: This paper outlines a quality improvement project undertaken to evaluate an ambulatory emergency care centre implemented at a tertiary hospital in Perth, Western Australia, from February to August 2021. RESULTS: The findings demonstrated a 4% improvement in the Western Australian Emergency Access Target for a four week period, a 6.3% reduction in the number of patients admitted with a length of stay less than 24 h and that patient's attending the Ambulatory Emergency Care Centre were managed as safely as if they were seen in the Emergency Department.


Asunto(s)
Servicios Médicos de Urgencia , Servicio de Urgencia en Hospital , Australia , Humanos , Centros de Atención Terciaria , Australia Occidental
2.
Sci Rep ; 10(1): 1082, 2020 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-31974453

RESUMEN

Human cytomegalovirus (CMV) is the leading non-genetic cause of fetal malformation in developed countries. CMV placental infection is a pre-requisite for materno-fetal transmission of virus, and fetal infection. We investigated the roles of the viral pentameric complex gH/gL/pUL128-pUL131A, and cellular platelet-derived growth factor receptor-α (PDGFRα) for CMV infection in first trimester extravillous-derived (SGHPL-4) and villous-derived (HTR-8/SVneo) trophoblast cells. Infection with four CMV clinical and laboratory strains (Merlin, TB40E, Towne, AD169), and Merlin deletion mutants of UL128-, UL130-, and UL131A-genes, showed a cell type-dependent requirement of the viral pentameric complex for infection of trophoblast cells. The viral pentameric complex was essential for infection of villous trophoblasts, but non-essential for extravillous trophoblasts. Blocking of PDGFRα in extravillous trophoblasts, which naturally express PDGFRα, inhibited entry of pentameric complex-deficient CMV strains, but not the entry of pentameric positive CMV strains. Transient expression of PDGFRα in villous trophoblasts, which are naturally deficient in PDGFRα, promoted the entry of CMV strains lacking gH/gL/pUL128-pUL131A, but had no effect on entry of pentameric positive CMV strains. These results suggest PDGFRα is an important cell receptor for entry of CMV mutant strains lacking gH/gL/pUL128-pUL131A complexes in some placental cells, suggesting these entry pathways could be potential antiviral targets.


Asunto(s)
Infecciones por Citomegalovirus/metabolismo , Citomegalovirus/fisiología , Complicaciones Infecciosas del Embarazo/metabolismo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Trofoblastos/metabolismo , Internalización del Virus , Línea Celular , Citomegalovirus/genética , Infecciones por Citomegalovirus/genética , Infecciones por Citomegalovirus/virología , Femenino , Humanos , Placenta/citología , Placenta/metabolismo , Placenta/virología , Embarazo , Complicaciones Infecciosas del Embarazo/genética , Complicaciones Infecciosas del Embarazo/virología , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Trofoblastos/virología , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/metabolismo
3.
J ECT ; 34(2): 113-116, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29424757

RESUMEN

OBJECTIVES: The objective of the survey was to assess confidence in electroconvulsive therapy (ECT) in core psychiatry trainees across Scotland, looking at both theoretical and practical aspects of ECT. METHODS: A link to a 14-item electronic questionnaire was distributed to core trainees via deanery administrators. Most questions were based on the Royal College of Psychiatry's ECT guidelines. RESULTS: A total of 85 responses were analyzed from trainees at all 3 levels of core training and from all health boards across Scotland. Almost 90% of trainees felt that their ECT training was sufficient, with more senior trainees rating their training better than those in the first year of training. Trainees who had theoretical teaching before their practical sessions rated their training better than those with purely observational training. Most trainees felt confident delivering ECT under supervision, and nearly 75% of trainees felt confident preparing a patient for ECT. The areas in which trainees felt least confident were in practical aspects such as dosing protocols and electroencephalogram interpretation. CONCLUSIONS: While ECT training and trainee confidence in delivering ECT were generally good, there are variations in trainees' experience that could be addressed by having a standardized ECT training, including theoretical teaching and practical competencies, in line with current guidelines. Ideally, evidence of meeting the Royal College recommendations for ECT competencies could be made a compulsory aspect of core training in the United Kingdom.


Asunto(s)
Competencia Clínica/estadística & datos numéricos , Terapia Electroconvulsiva/estadística & datos numéricos , Internado y Residencia/estadística & datos numéricos , Psiquiatría/educación , Adulto , Femenino , Conocimientos, Actitudes y Práctica en Salud , Humanos , Masculino , Persona de Mediana Edad , Escocia , Encuestas y Cuestionarios
4.
J Gen Virol ; 97(1): 144-151, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26555090

RESUMEN

Cyclin-dependent kinases (CDKs) are multifaceted regulators involved in the replication of human cytomegalovirus. Recently, we demonstrated an interaction of CDK9-cyclin T1 as well as viral CDK orthologue pUL97 with the viral regulator pUL69, thereby leading to pUL69-activating phosphorylation. Here, we demonstrate that colocalization and direct pUL69-cyclin T1 interaction is independent of viral strains and host cell types. In vitro phosphorylation of pUL69 by CDK9 or pUL97 did not occur in a single site-specific manner, but at multiple sites. The previously described fine-speckled nuclear aggregation of pUL69 was assigned to the late phase of viral replication. CDK inhibitors, including a novel inhibitor of the CDK-activating kinase CDK7, massively intensified this fine-speckled accumulation. Interestingly, we also observed spontaneous pUL69 accumulation in the absence of inhibitors at a lower frequency. These findings provide new insight into pUL69 kinase interregulation and emphasize the importance of pUL69 phosphorylation for correct intranuclear localization.


Asunto(s)
Quinasa 9 Dependiente de la Ciclina/metabolismo , Citomegalovirus/fisiología , Interacciones Huésped-Patógeno , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Procesamiento Proteico-Postraduccional , Transactivadores/metabolismo , Humanos , Fosforilación , Transporte de Proteínas
5.
Aust N Z J Obstet Gynaecol ; 56(1): 9-18, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26391432

RESUMEN

Human cytomegalovirus (CMV) is under-recognised, despite being the leading infectious cause of congenital malformation, affecting ~0.3% of Australian live births. Approximately 11% of infants born with congenital CMV infection are symptomatic, resulting in clinical manifestations, including jaundice, hepatosplenomegaly, petechiae, microcephaly, intrauterine growth restriction and death. Congenital CMV infection may cause severe long-term sequelae, including progressive sensorineural hearing loss and developmental delay in 40-58% of symptomatic neonates, and ~14% of initially asymptomatic infected neonates. Up to 50% of maternal CMV infections have nonspecific clinical manifestations, and most remain undetected unless specific serological testing is undertaken. The combination of serology tests for CMV-specific IgM, IgG and IgG avidity provide improved distinction between primary and secondary maternal infections. In pregnancies with confirmed primary maternal CMV infection, amniocentesis with CMV-PCR performed on amniotic fluid, undertaken after 21-22 weeks gestation, may determine whether maternofetal virus transmission has occurred. Ultrasound and, to a lesser extent, magnetic resonance imaging are valuable tools to assess fetal structural and growth abnormalities, although the absence of fetal abnormalities does not exclude fetal damage. Diagnosis of congenital CMV infection at birth or in the first 3 weeks of an infant's life is crucial, as this should prompt interventions for prevention of delayed-onset hearing loss and neurodevelopmental delay in affected infants. Prevention strategies should also target mothers because increased awareness and hygiene measures may reduce maternal infection. Recognition of the importance of CMV in pregnancy and in neonates is increasingly needed, particularly as therapeutic and preventive interventions expand for this serious problem.


Asunto(s)
Infecciones por Citomegalovirus/congénito , Transmisión Vertical de Enfermedad Infecciosa/prevención & control , Complicaciones Infecciosas del Embarazo , Anomalías Congénitas/diagnóstico , Anomalías Congénitas/prevención & control , Anomalías Congénitas/virología , Infecciones por Citomegalovirus/diagnóstico , Infecciones por Citomegalovirus/prevención & control , Infecciones por Citomegalovirus/transmisión , Discapacidades del Desarrollo/diagnóstico , Discapacidades del Desarrollo/prevención & control , Discapacidades del Desarrollo/virología , Femenino , Enfermedades Fetales/diagnóstico , Enfermedades Fetales/prevención & control , Enfermedades Fetales/virología , Pérdida Auditiva Sensorineural/congénito , Pérdida Auditiva Sensorineural/diagnóstico , Pérdida Auditiva Sensorineural/prevención & control , Pérdida Auditiva Sensorineural/virología , Humanos , Recién Nacido , Embarazo , Complicaciones Infecciosas del Embarazo/diagnóstico , Complicaciones Infecciosas del Embarazo/prevención & control
6.
Trials ; 16: 148, 2015 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-25872928

RESUMEN

BACKGROUND: The need for structured education programmes for type 2 diabetes is a high priority for many governments around the world. One such national education programme in the United Kingdom is the DESMOND Programme, which has been shown to be robust and effective for patients in general. However, these programmes are not generally targeted to people with intellectual disabilities (ID), and robust evidence on their effects for this population is lacking. We have adapted the DESMOND Programme for people with ID and type 2 diabetes to produce an amended programme known as DESMOND-ID. This protocol is for a pilot trial to determine whether a large-scale randomised trial is feasible, to test if DESMOND-ID is more effective than usual care in adults with ID for self-management of their type 2 diabetes, in particular as a means to reduce glycated haemoglobin (Hb1Ac), improve psychological wellbeing and quality of life and promote a healthier lifestyle. This protocol describes the rationale, methods, proposed analysis plan and organisational and administrative details. METHODS/DESIGN: This trial is a two arm, individually randomised, pilot trial for adults with ID and type 2 diabetes, and their family and/or paid carers. It compares the DESMOND-ID programme with usual care. Approximately 36 adults with mild to moderate ID will be recruited from three countries in the United Kingdom. Family and/or paid carers may also participate in the study. Participants will be randomly assigned to one of two conditions using a secure computerised system with robust allocation concealment. A range of data will be collected from the adults with ID (biomedical, psychosocial and self-management strategies) and from their carers. Focus groups with all the participants will assess the acceptability of the intervention and the trial. DISCUSSION: The lack of appropriate structured education programmes and educational materials for this population leads to secondary health conditions and may lead to premature deaths. There are significant benefits to be gained globally, if structured education programmes are adapted and shown to be successful for people with ID and other cognitive impairments. TRIAL REGISTRATION: Registered with International Standard Randomised Controlled Trial (identifier: ISRCTN93185560 ) on 10 November 2014.


Asunto(s)
Diabetes Mellitus Tipo 2/terapia , Conocimientos, Actitudes y Práctica en Salud , Discapacidad Intelectual/psicología , Educación del Paciente como Asunto , Personas con Discapacidades Mentales/psicología , Autocuidado , Adolescente , Adulto , Factores de Edad , Biomarcadores/sangre , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/diagnóstico , Diabetes Mellitus Tipo 2/psicología , Estudios de Factibilidad , Femenino , Grupos Focales , Hemoglobina Glucada/metabolismo , Humanos , Discapacidad Intelectual/diagnóstico , Masculino , Cooperación del Paciente , Proyectos Piloto , Evaluación de Programas y Proyectos de Salud , Calidad de Vida , Proyectos de Investigación , Factores de Tiempo , Resultado del Tratamiento , Reino Unido , Adulto Joven
7.
J Gen Virol ; 96(Pt 7): 1855-62, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25711967

RESUMEN

Human cytomegalovirus (CMV) is the most common infectious cause of congenital birth defects in developed countries. Studies of infected amniotic fluid and placentae show CMV infection leads to a pro-inflammatory shift in cytokine profiles with implications for pathogenesis of foetal disease. ELISA, immunofluorescence and real-time-PCR assays were used to investigate CCL2 (monocyte chemotactic protein-1) and TNF-α changes following CMV infection of human fibroblasts, as well as following transient expression of CMV gene products in HeLa cells. Infection of human fibroblasts with CMV AD169 resulted in increased cytoplasmic and extracellular expression of CCL2 during early stages of infection, followed by marked downregulation of the chemokine at late times. Induction of CCL2 was not observed with CMV clinical strain Merlin, consistent with the postulated immune-evasion potential of this genetically intact WT strain. Comparison between live and UV-irradiated virus infections showed that changes in CCL2 levels were a direct response to active CMV replication. There were no significant changes in TNF-α expression during a parallel time-course of CMV infection. In transient transfection assays, overexpression of CMV tegument protein pp71 resulted in intracellular and extracellular upregulation of CCL2 protein. mRNA analysis showed that pp71-induced elevation in CCL2 was mediated through transcriptional upregulation. The data showed that CMV-induced upregulation of CCL2 during early stages of infection was mediated, at least in part, by stimulation of viral pp71, which may contribute to viral pathogenesis through enhanced virus dissemination.


Asunto(s)
Quimiocina CCL2/biosíntesis , Citomegalovirus/inmunología , Proteínas Virales/inmunología , Células Cultivadas , Ensayo de Inmunoadsorción Enzimática , Células Epiteliales/inmunología , Células Epiteliales/virología , Fibroblastos/inmunología , Fibroblastos/virología , Técnica del Anticuerpo Fluorescente , Perfilación de la Expresión Génica , Humanos , Reacción en Cadena en Tiempo Real de la Polimerasa , Factor de Necrosis Tumoral alfa/biosíntesis
9.
Rev Med Virol ; 24(6): 420-33, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25316174

RESUMEN

Human cytomegalovirus is the leading non-genetic cause of congenital malformation in developed countries. Congenital CMV may result in fetal and neonatal death or development of serious clinical sequelae. In this review, we identified evidence-based interventions for prevention of congenital CMV at the primary level (prevention of maternal infection), secondary level (risk reduction of fetal infection and disease) and tertiary level (risk reduction of infected neonates being affected by CMV). A systematic review of existing literature revealed 24 eligible studies that met the inclusion criteria. Prevention of maternal infection using hygiene and behavioural interventions reduced maternal seroconversion rates during pregnancy. However, evidence suggested maternal adherence to education on preventative behaviours was a limiting factor. Treatment of maternal CMV infection with hyperimmune globulin (HIG) showed some evidence for efficacy in prevention of fetal infection and fetal/neonatal morbidity with a reasonable safety profile. However, more robust clinical evidence is required before HIG therapy can be routinely recommended. Limited evidence also existed for the safety and efficacy of established CMV antivirals (valaciclovir, ganciclovir and valganciclovir) to treat neonatal consequences of CMV infection, but toxicity and lack of randomised clinical trial data remain major issues. In the absence of a licensed CMV vaccine or robust clinical evidence for anti-CMV therapeutics, patient education and behavioural interventions that emphasise adherence remain the best preventative strategies for congenital CMV. There is a strong need for further data on the use of HIG and other antivirals in pregnancy, as well as the development of less toxic, novel, antiviral agents.


Asunto(s)
Infecciones por Citomegalovirus/prevención & control , Infecciones por Citomegalovirus/transmisión , Citomegalovirus/fisiología , Transmisión Vertical de Enfermedad Infecciosa/prevención & control , Complicaciones Infecciosas del Embarazo/prevención & control , Adulto , Antivirales/uso terapéutico , Citomegalovirus/efectos de los fármacos , Infecciones por Citomegalovirus/congénito , Infecciones por Citomegalovirus/tratamiento farmacológico , Femenino , Humanos , Recién Nacido , Masculino , Embarazo , Complicaciones Infecciosas del Embarazo/tratamiento farmacológico , Complicaciones Infecciosas del Embarazo/virología
10.
J Gen Virol ; 94(Pt 11): 2495-2503, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23939977

RESUMEN

Human cytomegalovirus (CMV) infects monocytes and other haematopoietic progenitor cells which then act as reservoirs for latency and virus dissemination. The chemokine CCL2 (monocyte chemotactic protein-1 or MCP-1) exhibits potent chemotactic activity for monocytes and is a likely target for CMV-induced immunomodulation. In this study, we demonstrate CMV modulates CCL2 expression in MRC-5 fibroblasts with multiplicity-dependent kinetics, where CCL2 is upregulated during early stage infection, followed by CCL2 inhibition at late stage infection. This CMV-induced CCL2 modulation was dependent upon virus replication, as UV-inactivated virus did not elicit any changes in CCL2 levels. Dual immunofluorescence staining showed CMV strains AD169, purified AD169, Merlin, FIX WT (FLAG-US28/WT) and pUS28-deficient FIX (FIX-ΔUS28) all induced upregulation of CCL2 primarily within infected cells. Focal upregulation of CCL2 within FIX-ΔUS28-infected cells demonstrated intracellular CCL2 accumulation was independent of CCL2 sequestration by the CMV-encoded chemokine receptor US28. Infection with purified virus confirmed CMV-induced CCL2 upregulation was not due to any CCL2-inducing factors contained within non-purified virus stocks. The CMV-induced CCL2 expression kinetics occurred concurrently with modulation of the CCL2 transcriptional activators NF-κB, interferon regulatory factor 3 and cytokine IFN-ß, independent of virus strain, and with the establishment of viral replication compartments within infected cell nuclei. This is the first report to our knowledge to demonstrate CMV modulation of CCL2 expression within infected cells during viral replication. This immune modulation may facilitate virus dissemination, establishment of latency and pathogenesis of CMV-induced host disease.


Asunto(s)
Quimiocina CCL2/metabolismo , Citomegalovirus/patogenicidad , Fibroblastos/virología , Regulación de la Expresión Génica , Replicación Viral/fisiología , Línea Celular , Quimiocina CCL2/genética , Citomegalovirus/fisiología , Humanos , Inmunomodulación , Cinética
11.
J Paediatr Child Health ; 49(3): E221-6, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23432694

RESUMEN

AIM: Premature neonates represent a population highly vulnerable to infection. This study aims to profile viral colonisation of gut and the associated clinical manifestations among premature neonates admitted to a neonatal intensive care unit (NICU) in Australia. METHODS: In a cohort of 75 premature neonates born at less than 32 weeks gestation, who were followed for 4 weeks following admission to a NICU in Sydney, Australia, multiplex polymerase chain reaction assays were used to determine viral presence in stool, and clinical data were examined. RESULTS: Overall, viral RNA or DNA was detected in 24/419 (5.7%) of specimens, including norovirus genogroup 2 (1.9%), enterovirus (1.2%), herpes simplex virus-2 (1.2%), cytomegalovirus (0.7%), Epstein-Barr virus (0.5%) and rotavirus (0.2%). Viral infection was detected in 13/75 (17%) of premature neonates at some time point, including five (7%) neonates shedding more than one type of virus in stool. A higher rate of infection was observed among premature neonates with intrauterine growth restriction (56%) compared with those infants born appropriate for gestational age (12%. P = 0.006). CONCLUSION: The overall viral detection rate in stool of 5.7% (affecting 17% of neonates) indicates viral infections are an important health risk for premature infants in NICU.


Asunto(s)
Heces/virología , Enfermedades Gastrointestinales/virología , Enfermedades del Prematuro/epidemiología , Virosis/diagnóstico , Australia/epidemiología , Femenino , Enfermedades Gastrointestinales/diagnóstico , Enfermedades Gastrointestinales/epidemiología , Humanos , Lactante , Recién Nacido , Recien Nacido Prematuro , Enfermedades del Prematuro/virología , Unidades de Cuidado Intensivo Neonatal , Masculino , Reacción en Cadena de la Polimerasa , Prevalencia , Virosis/epidemiología
13.
J Med Virol ; 85(2): 315-9, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23171986

RESUMEN

Human cytomegalovirus (CMV) is the most common infectious cause of mental disability in newborns of developed countries. Transmission of CMV from mother to baby is more frequent in maternal primary infection, although CMV reactivation causes more congenital infections overall. Current diagnostic tests for distinguishing primary and reactivation CMV have problems with interpretation and immunoblots may assist with diagnosis. Sera from 60 pregnant women were analyzed using conventional serology in parallel with a commercial immunoblot assay (using Recomblot, Mikrogen Diagnostik). Comparison of detection of CMV IgG, IgM, IgG avidity in maternal primary infection showed the immunoblot relative to conventional serology had sensitivity and specificity of 100% for IgG identification. The detection of IgM on immunoblot showed sensitivity of 75%, specificity of 62.5%, positive predictive value (PPV) of 81.8% and negative predictive value (NPV) of 52.6%. The immunoblot IgG avidity assay had sensitivity of 94.1%, with a PPV of 100% when identifying low avidity serum samples, and sensitivity of 100% with a PPV of 97.1% for high avidity serum samples. Overall agreement between conventional serology (IgM, IgG avidity) and immunoblot (IgM, IgG avidity) for detection of primary CMV infection was 65%. Although the immunoblot is effective in detecting IgG and determining IgG avidity, it showed no significant benefits in performance or utility as a first line diagnostic technique for IgM or primary CMV infection in pregnant women.


Asunto(s)
Anticuerpos Antivirales/sangre , Infecciones por Citomegalovirus/diagnóstico , Citomegalovirus/inmunología , Immunoblotting/métodos , Complicaciones Infecciosas del Embarazo/diagnóstico , Adulto , Afinidad de Anticuerpos , Técnicas de Laboratorio Clínico/métodos , Femenino , Humanos , Inmunoglobulina G/sangre , Inmunoglobulina M/sangre , Recién Nacido , Persona de Mediana Edad , Valor Predictivo de las Pruebas , Embarazo , Sensibilidad y Especificidad , Virología/métodos , Adulto Joven
14.
J Infect Dis ; 205(8): 1305-10, 2012 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-22383678

RESUMEN

Congenital infection with cytomegalovirus (CMV) can induce immune responses and placental damage. By use of immunoassay panels, 27 cytokines were assessed in midtrimester amniotic fluid from 8 patients with congenital CMV, in midtrimester sera from 12 pregnant women with primary CMV infection, and in amniotic fluid and serum from uninfected maternal controls. Levels of the cytokines tumor necrosis factor α, interleukin 1ß, interleukin 12, and interleukin 17; the chemokines CCL2, CCL4, and CXCL10; and the growth factors granulocyte-macrophage colony-stimulating factor and platelet-derived growth factor bb were significantly elevated in amniotic fluid from congenital CMV patients (P < .01). Only CXCL10 was significantly higher in sera from CMV-infected pregnant women. CMV infection during pregnancy is associated with a shift in cytokine expression toward a proinflammatory state.


Asunto(s)
Líquido Amniótico/metabolismo , Citocinas/metabolismo , Infecciones por Citomegalovirus/transmisión , Transmisión Vertical de Enfermedad Infecciosa , Placenta/metabolismo , Complicaciones Infecciosas del Embarazo/patología , Adulto , Análisis por Conglomerados , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Recién Nacido , Embarazo
15.
PLoS One ; 7(12): e52899, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23300810

RESUMEN

Human cytomegalovirus (CMV) infection of the developing fetus can result in adverse pregnancy outcomes including death in utero. Fetal injury results from direct viral cytopathic damage to the CMV-infected fetus, although evidence suggests CMV placental infection may indirectly cause injury to the fetus, possibly via immune dysregulation with placental dysfunction. This study investigated the effects of CMV infection on expression of the chemokine MCP-1 (CCL2) and cytokine TNF-α in placentae from naturally infected stillborn babies, and compared these changes with those found in placental villous explant histocultures acutely infected with CMV ex vivo. Tissue cytokine protein levels were assessed using quantitative immunohistochemistry. CMV-infected placentae from stillborn babies had significantly elevated MCP-1 and TNF-α levels compared with uninfected placentae (p = 0.001 and p = 0.007), which was not observed in placentae infected with other microorganisms (p = 0.62 and p = 0.71) (n = 7 per group). Modelling acute clinical infection using ex vivo placental explant histocultures showed infection with CMV laboratory strain AD169 (0.2 pfu/ml) caused significantly elevated expression of MCP-1 and TNF-α compared with uninfected explants (p = 0.0003 and p<0.0001) (n = 25 per group). Explant infection with wild-type Merlin at a tenfold lower multiplicity of infection (0.02 pfu/ml), caused a significant positive correlation between increased explant infection and upregulation of MCP-1 and TNF-α expression (p = 0.0001 and p = 0.017). Cytokine dysregulation has been associated with adverse outcomes of pregnancy, and can negatively affect placental development and function. These novel findings demonstrate CMV infection modulates the placental immune environment in vivo and in a multicellular ex vivo model, suggesting CMV-induced cytokine modulation as a potential initiator and/or exacerbator of placental and fetal injury.


Asunto(s)
Quimiocina CCL2/metabolismo , Infecciones por Citomegalovirus/metabolismo , Citomegalovirus/inmunología , Placenta/metabolismo , Complicaciones Infecciosas del Embarazo/metabolismo , Mortinato , Factor de Necrosis Tumoral alfa/metabolismo , Células Cultivadas , Quimiocina CCL2/genética , Citocinas/metabolismo , Citomegalovirus/fisiología , Infecciones por Citomegalovirus/inmunología , Infecciones por Citomegalovirus/mortalidad , Infecciones por Citomegalovirus/virología , Femenino , Expresión Génica , Interacciones Huésped-Patógeno , Humanos , Placenta/patología , Placenta/virología , Embarazo , Complicaciones Infecciosas del Embarazo/inmunología , Complicaciones Infecciosas del Embarazo/mortalidad , Complicaciones Infecciosas del Embarazo/virología , Estudios Retrospectivos , Técnicas de Cultivo de Tejidos , Trofoblastos/virología , Factor de Necrosis Tumoral alfa/genética , Regulación hacia Arriba , Replicación Viral
16.
Cornea ; 30(9): 1054-7, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21673567

RESUMEN

PURPOSE: To report a case of recurrent corneal graft failure because of cytomegalovirus (CMV) infection and to demonstrate successful clearance of the virus with oral valganciclovir, confirmed by polymerase chain reaction (PCR) assay. This allowed for a successful corneal autograft to be performed. METHODS: Interventional case report. RESULTS: A 90-year-old white man with 4 previous corneal graft failures in his right eye is presented. His visual acuity was no light perception in the left eye subsequent to ocular trauma. His initial penetrating keratoplasty for pseudophakic bullous keratopathy was from a human leukocyte antigen-matched multiorgan donor who was CMV-seropositive. An anterior chamber paracentesis was performed to exclude an infective etiology. CMV was detected on PCR of aqueous humor. After a 12-week course of oral valganciclovir, a repeat aqueous PCR test confirmed the clearance of CMV. A corneal autograft from his left eye was subsequently performed with good outcome. CONCLUSIONS: We present a case of successful corneal autograft after clearance of CMV from the anterior chamber (PCR confirmed) in a patient treated with oral valganciclovir.


Asunto(s)
Antivirales/uso terapéutico , Humor Acuoso/virología , Infecciones por Citomegalovirus/cirugía , Infecciones Virales del Ojo/cirugía , Ganciclovir/análogos & derivados , Queratoplastia Penetrante , Administración Oral , Anciano de 80 o más Años , Citomegalovirus/genética , Citomegalovirus/aislamiento & purificación , Infecciones por Citomegalovirus/tratamiento farmacológico , Infecciones por Citomegalovirus/virología , ADN Viral/análisis , Infecciones Virales del Ojo/tratamiento farmacológico , Infecciones Virales del Ojo/virología , Ganciclovir/uso terapéutico , Rechazo de Injerto/cirugía , Supervivencia de Injerto , Humanos , Masculino , Reacción en Cadena de la Polimerasa , Recurrencia , Reoperación , Trasplante Autólogo , Trasplante Homólogo , Valganciclovir
17.
J Gen Virol ; 92(Pt 5): 1021-1031, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21307228

RESUMEN

Natural human cytomegalovirus (CMV) infection is characterized by a strain-specific neutralizing antibody response. This is particularly relevant in clinical settings such as transplantation and pregnancy where reinfection with heterologous strains occurs and the immune system does not mount an effective response against the infecting strain due to underlying immunosuppression. There is an emerging argument that a CMV vaccine that induces high titres of cross-neutralizing antibodies will be more effective in protecting individuals from infection with antigenically different CMV strains. In addition, induction of cell-mediated immunity offers the additional advantage of targeting virus-infected cells. This study presents a novel formulation of a CMV vaccine that, by combining recombinant soluble gB protein with a Toll-like receptor 9 agonist (CpG ODN1826) and immune-stimulating complexes (AbISCO 100), was able to elicit strong polyfunctional CMV-specific cellular and cross-neutralizing humoral immune responses. These data demonstrated that prime-boost immunization of human leukocyte antigen (HLA)-A2 mice with gB protein in combination with CpG ODN1826 and AbISCO 100 induced long-lasting CMV-specific CD4(+) and CD8(+) T-cell and humoral responses. Furthermore, these responses neutralized infection with multiple strains of CMV expressing different gB genotypes and afforded protection against challenge with recombinant vaccinia virus encoding the gB protein. These observations argue that this novel vaccine strategy, if applied to humans, should facilitate the generation of a robust, pluripotent immune response, which may be more effective in preventing infection with multiple strains of CMV.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Vacunas contra Citomegalovirus/inmunología , Oligodesoxirribonucleótidos/administración & dosificación , Proteínas del Envoltorio Viral/inmunología , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Complejo Antígeno-Anticuerpo/administración & dosificación , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Vacunas contra Citomegalovirus/administración & dosificación , Femenino , Humanos , Inmunización Secundaria/métodos , Ratones , Ratones Transgénicos , Receptor Toll-Like 9/agonistas , Vacunación/métodos , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/inmunología , Proteínas del Envoltorio Viral/administración & dosificación
18.
J Med Virol ; 83(3): 510-6, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21264873

RESUMEN

Influenza virus evolves continuously through mutations presumed to result from evolutionary pressure driving viral replication. This study examined the relationship between the genetic evolution and replication fidelity of influenza viruses. Analysis of influenza sequences from National Centre for Biotechnology Information (NCBI) database revealed a gradual decrease in the rate of genetic evolution of A/Fujian/411/02(H3N2)-like variants after the emergence and predominance of the A/H3N2 Fujian strain in 2002. This decrease may be related to an increase in replication fidelity, which was investigated by assessing mutation frequencies of reassortant viruses carrying the PB1 segment of Fujian variants isolated between 2003 and 2005 in a sequencing-based plaque assay. The data revealed a threefold decrease in substitution per site of the reassortant viruses carrying the Fujian PB1 segments isolated in 2004-2005 compared with those circulating in 2003. The decrease in mutation frequency paralleled a decrease in genetic evolution of the Fujian variants from the NCBI database. This correlation implicates changes in the polymerase replication fidelity as contributing to altered genetic evolution of influenza viruses.


Asunto(s)
Evolución Molecular , Subtipo H3N2 del Virus de la Influenza A/fisiología , Proteínas Virales/genética , Proteínas Virales/metabolismo , Replicación Viral/genética , Sustitución de Aminoácidos , Línea Celular , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Humanos , Subtipo H3N2 del Virus de la Influenza A/enzimología , Subtipo H3N2 del Virus de la Influenza A/genética , Mutación , Polimorfismo Genético , Virus Reordenados/fisiología , Proteínas no Estructurales Virales/genética
19.
J Virol ; 84(14): 7096-104, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20444889

RESUMEN

During human adenovirus 5 infection, a temporal cascade of gene expression leads ultimately to the production of large amounts of the proteins needed to construct progeny virions. However, the mechanism for the activation of the major late gene that encodes these viral structural proteins has not been well understood. We show here that two key positive regulators of the major late gene, L4-22K and L4-33K, previously thought to be expressed under the control of the major late promoter itself, initially are expressed from a novel promoter that is embedded within the major late gene and dedicated to their expression. This L4 promoter is required for late gene expression and is activated by a combination of viral protein activators produced during the infection, including E1A, E4 Orf3, and the intermediate-phase protein IVa2, and also by viral genome replication. This new understanding redraws the long-established view of how adenoviral gene expression patterns are controlled and offers new ways to manipulate that gene expression cascade for adenovirus vector applications.


Asunto(s)
Adenovirus Humanos/genética , Adenovirus Humanos/patogenicidad , Regulación Viral de la Expresión Génica , Regiones Promotoras Genéticas , Proteínas Virales/genética , Línea Celular , ADN Viral/genética , ADN Viral/metabolismo , Células HeLa , Humanos , Proteínas Virales/metabolismo , Replicación Viral/genética
20.
J Biol Chem ; 284(13): 8605-13, 2009 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-19179338

RESUMEN

Replication of human cytomegalovirus (HCMV) is subject to regulation by cellular protein kinases. Recently, we and others reported that inhibition of cyclin-dependent protein kinases (CDKs) or the viral CDK ortholog pUL97 can induce intranuclear speckled aggregation of the viral mRNA export factor, pUL69. Here we provide the first evidence for a direct regulatory role of CDKs on pUL69 functionality. Although replication of all HCMV strains was dependent on CDK activity, we found strain-specific differences in the amount of CDK inhibitor-induced pUL69 aggregate formation. In all cases analyzed, the inhibitor-induced pUL69 aggregates were clearly localized within viral replication centers but not subnuclear splicing, pore complex, or aggresome structures. The CDK9 and cyclin T1 proteins colocalized with these pUL69 aggregates, whereas other CDKs behaved differently. Phosphorylation analyses in vivo and in vitro demonstrated pUL69 was strongly phosphorylated in HCMV-infected fibroblasts and that CDKs represent a novel class of pUL69-phosphorylating kinases. Moreover, the analysis of CDK inhibitors in a pUL69-dependent nuclear mRNA export assay provided evidence for functional impairment of pUL69 under suppression of CDK activity. Thus, our data underline the crucial importance of CDKs for HCMV replication, and indicate a direct impact of CDK9-cyclin T1 on the nuclear localization and activity of the viral regulator pUL69.


Asunto(s)
Núcleo Celular/metabolismo , Quinasa 9 Dependiente de la Ciclina/metabolismo , Ciclinas/metabolismo , Infecciones por Citomegalovirus/metabolismo , Citomegalovirus/fisiología , Fibroblastos/metabolismo , Transactivadores/metabolismo , Transporte Activo de Núcleo Celular/fisiología , Núcleo Celular/virología , Ciclina T , Fibroblastos/virología , Células HeLa , Humanos , Fosforilación , ARN Mensajero/metabolismo , ARN Viral/metabolismo , Replicación Viral/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...