Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
J Gen Virol ; 101(10): 1047-1055, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32667279

RESUMEN

Type I interferon receptor knockout mice (strain A129) were assessed as a disease model of hantavirus infection. A range of infection routes (intramuscular, intraperitoneal and intranasal) were assessed using minimally passaged Seoul virus (strain Humber). Dissemination of virus to the spleen, kidney and lung was observed at 5 days after intramuscular and intraperitoneal challenge, which was resolved by day 14. In contrast, intranasal challenge of A129 mice demonstrated virus tropism to the lung, which was maintained to day 14 post-challenge. These data support the use of the A129 mouse model for future infection studies and the in vivo evaluation of interventions.


Asunto(s)
Modelos Animales de Enfermedad , Infecciones por Hantavirus , Orthohantavirus/fisiología , Animales , Orthohantavirus/aislamiento & purificación , Orthohantavirus/patogenicidad , Infecciones por Hantavirus/patología , Infecciones por Hantavirus/virología , Fiebre Hemorrágica con Síndrome Renal/patología , Fiebre Hemorrágica con Síndrome Renal/virología , Riñón/virología , Hígado/patología , Pulmón/patología , Pulmón/virología , Masculino , Ratones , Ratones Noqueados , ARN Viral/análisis , ARN Viral/sangre , Receptor de Interferón alfa y beta/genética , Bazo/patología , Bazo/virología , Tropismo Viral
2.
Cytokine ; 125: 154864, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31577989

RESUMEN

Zika virus (ZIKV) is phylogenetically divided into two lineages comprising African (ZIKVAF) and Asian (ZIKVAS) genotypes. In the type-I interferon receptor deficient mouse model, ZIKVAF causes severe disease with all mice meeting humane endpoints with doses as low as 10 plaque-forming units (pfu) whereas a much milder infection is seen after challenge with ZIKVAS, including with doses as high as 106 pfu. Using this mouse model, the elucidation of cytokine, chemokine, growth factor and acute phase protein responses over the course of infection were studied to determine whether these analytes contributed to the stark difference in clinical outcome. Results demonstrated some significant differences, with the ZIKVAF infection being associated with increases in a higher number of biomarkers than ZIKVAS. When low (10 pfu) and high (106 pfu) challenge doses were compared, animals given the lower virus inoculum showed a wider range of responses, indicating a different disease progression compared to those challenged with high doses. These results aid with elucidating the different outcomes with the two lineages of ZIKV and with future work to assess pathogenicity of virus infection.


Asunto(s)
Proteínas de Fase Aguda/metabolismo , Quimiocinas/sangre , Citocinas/sangre , Péptidos y Proteínas de Señalización Intercelular/sangre , Infección por el Virus Zika/metabolismo , Virus Zika/patogenicidad , Animales , Quimiocinas/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Inflamación/metabolismo , Inflamación/virología , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Masculino , Ratones , Receptor de Interferón alfa y beta/deficiencia , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/metabolismo , Infección por el Virus Zika/fisiopatología , Infección por el Virus Zika/virología
3.
Methods ; 158: 17-21, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30771491

RESUMEN

In the UK, research on hazard group 4 (HG4) pathogens requires specialised Containment Level 4 (CL4) facilities. These differ from Biosafety Level 4 (BSL4) conditions in that work is conducted in class III microbiological safety cabinets for primary containment instead of using positive pressure suits. This presents unique challenges associated with the physical restrictions of working in a limited space, and prohibits the use of many techniques and specialist equipment. In consequence, detailed studies on the biology of HG4 pathogens and in particular their immunological relationships with the host are understudied in the UK; for example, the majority of immunological assays with which the immune system is interrogated require specialist equipment that is unsuitable for CL4. Multiplexing to simultaneously measure multiple analytes is increasingly being used in immunological studies. This assay is attractive for CL4 work because it reduces the time spent in the laboratory whilst maximising the use of valuable sample volume. The Luminex microsphere approach allows for the determination of many cytokines and chemokines, however, the detection system uses fixed aligned lasers and integrated computer systems which are unsuitable for use at CL4. Therefore, we have developed an approach in which the Luminex assay is conducted within the CL4 laboratory and a formalin-fixation stage is introduced to allow for analysis to be undertaken outside of containment. Quality control preparations allow the assay characteristics to be monitored and analysis of assay performance to be evaluated. Our data demonstrate that Luminex is an applicable tool for use at CL4 and that assays can be run reliably to generate reproducible standardised data across different plates and individual experiments.


Asunto(s)
Contención de Riesgos Biológicos/normas , Ensayos Analíticos de Alto Rendimiento/instrumentación , Laboratorios/normas , Microbiología/normas , Microesferas , Servicios de Laboratorio Clínico , Enfermedades Transmisibles/diagnóstico , Enfermedades Transmisibles/microbiología , Fijadores/química , Formaldehído/química , Ensayos Analíticos de Alto Rendimiento/métodos , Ensayos Analíticos de Alto Rendimiento/normas , Humanos , Control de Calidad , Reproducibilidad de los Resultados , Fijación del Tejido/métodos , Fijación del Tejido/normas
4.
J Virol ; 87(14): 7805-15, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23658452

RESUMEN

To support the licensure of a new and safer vaccine to protect people against smallpox, a monkeypox model of infection in cynomolgus macaques, which simulates smallpox in humans, was used to evaluate two vaccines, Acam2000 and Imvamune, for protection against disease. Animals vaccinated with a single immunization of Imvamune were not protected completely from severe and/or lethal infection, whereas those receiving either a prime and boost of Imvamune or a single immunization with Acam2000 were protected completely. Additional parameters, including clinical observations, radiographs, viral load in blood, throat swabs, and selected tissues, vaccinia virus-specific antibody responses, immunophenotyping, extracellular cytokine levels, and histopathology were assessed. There was no significant difference (P > 0.05) between the levels of neutralizing antibody in animals vaccinated with a single immunization of Acam2000 (132 U/ml) and the prime-boost Imvamune regime (69 U/ml) prior to challenge with monkeypox virus. After challenge, there was evidence of viral excretion from the throats of 2 of 6 animals in the prime-boost Imvamune group, whereas there was no confirmation of excreted live virus in the Acam2000 group. This evaluation of different human smallpox vaccines in cynomolgus macaques helps to provide information about optimal vaccine strategies in the absence of human challenge studies.


Asunto(s)
Inmunización/métodos , Orthopoxvirus/inmunología , Infecciones por Poxviridae/prevención & control , Vacuna contra Viruela/farmacología , Animales , Anticuerpos Neutralizantes/inmunología , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Macaca fascicularis , Masculino , Reacción en Cadena en Tiempo Real de la Polimerasa , Vacunas Atenuadas/farmacología , Esparcimiento de Virus/inmunología
5.
J Immunol Methods ; 512: 113405, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36496007

RESUMEN

Humanised antibodies targeting Crimean-Congo Haemorrhagic virus (CCHFV) are needed for the development and standardisation of serological assays. These assays are needed to address a shortfall in available tests that meet regulatory diagnostic standards and to aid surveillance activities to extend knowledge on the distribution of CCHFV. To generate a humanised monoclonal antibody against CCHFV, we have compared two methods: the traditional mouse hybridoma approach with subsequent sequencing and humanisation of antibodies versus a non-animal alternative using a human combinatorial antibody library (HuCAL). Our results demonstrated that the mouse hybridoma followed by humanisation protocol gave higher affinity antibodies. Whilst not yet able to demonstrate the generation of equivalent humanised antibodies without the use of animals, sequencing data enables the subsequent production of recombinant antibodies, thus providing a reduction in future animal usage for this application. Ultimately, our report provides information on development of a humanised standardised control, which can form an important positive control component of serological assays against CCHFV.


Asunto(s)
Virus de la Fiebre Hemorrágica de Crimea-Congo , Fiebre Hemorrágica de Crimea , Humanos , Animales , Ratones , Fiebre Hemorrágica de Crimea/diagnóstico , Fiebre Hemorrágica de Crimea/epidemiología , Hibridomas , Anticuerpos Antivirales , Inmunoglobulina G
6.
Viruses ; 15(3)2023 03 11.
Artículo en Inglés | MEDLINE | ID: mdl-36992434

RESUMEN

The emergence of severe acute respiratory syndrome coronavirus (SARS-CoV-2) and its expansion to a worldwide pandemic resulted in efforts to assess and develop interventions to reduce the disease burden. Despite the introduction of vaccine programmes against SARS-CoV-2, global incidence levels in early 2022 remained high, demonstrating a need for the development of physiologically relevant models, which are essential for the identification of alternative antiviral strategies. The hamster model of SARS-CoV-2 infection has been widely adopted due to similarities with humans in terms of host cell entry mechanism (via ACE2), and aspects of symptomology and virus shedding. We have previously described a natural transmission hamster model that better represents the natural course of infection. In the present study, we have conducted further testing of the model using the first-in-class antiviral Neumifil, which has previously shown promise against SARS-CoV-2 after a direct intranasal challenge. Neumifil is an intranasally delivered carbohydrate-binding module (CBM) which reduces the binding of viruses to their cellular receptor. By targeting the host cell, Neumifil has the potential to provide broad protection against multiple pathogens and variants. This study demonstrates that using a combination of a prophylactic and therapeutic delivery of Neumifil significantly reduces the severity of clinical signs in animals infected via a natural route of transmission and indicates a reduction of viral loads in the upper respiratory tract. Further refinements of the model are required in order to ensure the adequate transmission of the virus. However, our results provide additional data to the evidence base of Neumifil efficacy against respiratory virus infection and demonstrate that the transmission model is a potentially valuable tool for testing antiviral compounds against SARS-CoV-2.


Asunto(s)
COVID-19 , SARS-CoV-2 , Animales , Cricetinae , Humanos , SARS-CoV-2/metabolismo , Antivirales/farmacología , Antivirales/uso terapéutico , Antivirales/química , Carbohidratos
7.
EBioMedicine ; 90: 104523, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-36933409

RESUMEN

BACKGROUND: The tick-borne bunyavirus, Crimean-Congo Haemorrhagic Fever virus (CCHFV), can cause severe febrile illness in humans and has a wide geographic range that continues to expand due to tick migration. Currently, there are no licensed vaccines against CCHFV for widespread usage. METHODS: In this study, we describe the preclinical assessment of a chimpanzee adenoviral vectored vaccine (ChAdOx2 CCHF) which encodes the glycoprotein precursor (GPC) from CCHFV. FINDINGS: We demonstrate here that vaccination with ChAdOx2 CCHF induces both a humoral and cellular immune response in mice and 100% protection in a lethal CCHF challenge model. Delivery of the adenoviral vaccine in a heterologous vaccine regimen with a Modified Vaccinia Ankara vaccine (MVA CCHF) induces the highest levels of CCHFV-specific cell-mediated and antibody responses in mice. Histopathological examination and viral load analysis of the tissues of ChAdOx2 CCHF immunised mice reveals an absence of both microscopic changes and viral antigen associated with CCHF infection, further demonstrating protection against disease. INTERPRETATION: There is the continued need for an effective vaccine against CCHFV to protect humans from lethal haemorrhagic disease. Our findings support further development of the ChAd platform expressing the CCHFV GPC to seek an effective vaccine against CCHFV. FUNDING: This research was supported by funding from the Biotechnology and Biological Sciences Research Council (UKRI-BBSRC) [BB/R019991/1 and BB/T008784/1].


Asunto(s)
Virus de la Fiebre Hemorrágica de Crimea-Congo , Fiebre Hemorrágica de Crimea , Vacunas Virales , Humanos , Animales , Ratones , Fiebre Hemorrágica de Crimea/prevención & control , Virus de la Fiebre Hemorrágica de Crimea-Congo/genética , Vacunación , Vectores Genéticos/genética , Virus Vaccinia
8.
Viruses ; 15(12)2023 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-38140610

RESUMEN

Rift Valley fever virus (RVFV) is a mosquito-borne zoonotic pathogen causing disease in livestock and humans. Whilst initially restricted to the African continent, recent spread to the Arabian Peninsula has highlighted the likelihood of entry into new regions. Due to the absence of a regulatory-approved human vaccine, work is ongoing to develop and assess countermeasures. As such, small animal models play a pivotal role in providing information on disease pathogenesis and elucidating which intervention strategies confer protection. To develop and establish the BALB/c mouse model, we challenged mice with RVFV grown from two separate cell lines: one derived from mosquitoes (C6/36) and the other mammalian derived (Vero E6). Following infection, we assessed the clinical course of disease progression at days 1 and 3 post-challenge and evaluated viral tropism and immune analytes. The results demonstrated that RVFV infection was affected by the cell line used to propagate the challenge virus, with those grown in insect cells resulting in a more rapid disease progression. The lowest dose that caused uniform severe disease remained the same across both virus preparations. In addition, to demonstrate reproducibility, the lowest dose was used for a subsequent infection study using male and female animals. The results further demonstrated that male mice succumbed to infection more rapidly than their female counterparts. Our results establish an RVFV mouse model and key parameters that affect the course of disease progression in BALB/c mice.


Asunto(s)
Fiebre del Valle del Rift , Virus de la Fiebre del Valle del Rift , Masculino , Femenino , Humanos , Animales , Ratones , Ratones Endogámicos BALB C , Reproducibilidad de los Resultados , Progresión de la Enfermedad , Mamíferos
9.
Vaccine ; 38(2): 345-349, 2020 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-31668821

RESUMEN

Ebola virus (EBOV) represents a major concern to global health due to the unpredictable nature of outbreaks. Infection with EBOV can cause a severe viral haemorrhagic fever with no licensed vaccine or treatment, restricting work with live EBOV to Containment/Biosafety Level 4 facilities. Whilst the magnitude of recent outbreaks has provided an impetus for vaccine and antiviral development, establishing the efficacy of candidate vaccine materials relies on EBOV challenge models and advanced human trials should outbreaks occur and where logistics and funding allow. To address these hurdles in vaccine development, we investigated whether a recently established serological reference standard, the 1st WHO International Standard for Ebola virus antibody, could be used to provide a quantifiable correlate of immune protection in vivo. Dilutions of the International Standard were inoculated into naïve guinea pigs 24 h before challenge with a lethal dose of Ebola virus. Only subjects receiving the highest dose of the International Standard exhibited evidence of delayed progression. Due to it being a WHO established reagent and available globally upon request, this standard allows for effective comparisons of data between laboratories and may prove valuable to select the candidate vaccines that are most likely to confer humoral immune protection ensuring the most promising candidates progress into efficacy studies.


Asunto(s)
Formación de Anticuerpos/inmunología , Vacunas contra el Virus del Ébola/administración & dosificación , Ebolavirus/inmunología , Fiebre Hemorrágica Ebola/prevención & control , Animales , Desarrollo de Medicamentos , Vacunas contra el Virus del Ébola/inmunología , Femenino , Cobayas , Fiebre Hemorrágica Ebola/inmunología , Inmunización Pasiva/métodos
10.
Cell Rep ; 25(13): 3750-3758.e4, 2018 12 26.
Artículo en Inglés | MEDLINE | ID: mdl-30590046

RESUMEN

The Gn subcomponent of the Gn-Gc assembly that envelopes the human and animal pathogen, Rift Valley fever virus (RVFV), is a primary target of the neutralizing antibody response. To better understand the molecular basis for immune recognition, we raised a class of neutralizing monoclonal antibodies (nAbs) against RVFV Gn, which exhibited protective efficacy in a mouse infection model. Structural characterization revealed that these nAbs were directed to the membrane-distal domain of RVFV Gn and likely prevented virus entry into a host cell by blocking fusogenic rearrangements of the Gn-Gc lattice. Genome sequence analysis confirmed that this region of the RVFV Gn-Gc assembly was under selective pressure and constituted a site of vulnerability on the virion surface. These data provide a blueprint for the rational design of immunotherapeutics and vaccines capable of preventing RVFV infection and a model for understanding Ab-mediated neutralization of bunyaviruses more generally.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Anticuerpos Antivirales/farmacología , Virus de la Fiebre del Valle del Rift/inmunología , Secuencia de Aminoácidos , Animales , Anticuerpos Neutralizantes/farmacología , Chlorocebus aethiops , Femenino , Glicoproteínas/química , Glicoproteínas/metabolismo , Células HEK293 , Humanos , Inmunización , Inmunoglobulina G/metabolismo , Ratones Endogámicos BALB C , Modelos Biológicos , Pruebas de Neutralización , Dominios Proteicos , Conejos , Proteínas Recombinantes/farmacología , Virus de la Fiebre del Valle del Rift/efectos de los fármacos , Células Vero , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/metabolismo
11.
PLoS Negl Trop Dis ; 11(7): e0005704, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28672028

RESUMEN

Zika virus (ZIKV) falls into two lineages: African (ZIKVAF) and Asian (ZIKVAS). These lineages have not been tested comprehensively in parallel for disease progression using an animal model system. Here, using the established type-I interferon receptor knockout (A129) mouse model, it is first demonstrated that ZIKVAF causes lethal infection, with different kinetics of disease manifestations according to the challenge dose. Animals challenged with a low dose of 10 plaque-forming units (pfu) developed more neurological symptoms than those challenged with 5-log higher doses. By contrast, animals challenged with ZIKVAS displayed no clinical signs or mortality, even at doses of 106 pfu. However, viral RNA was detected in the tissues of animals infected with ZIKV strains from both lineages and similar histological changes were observed. The present study highlights strain specific virulence differences between the African and Asian lineages in a ZIKV mouse model.


Asunto(s)
Receptor de Interferón alfa y beta/deficiencia , Infección por el Virus Zika/patología , Virus Zika/patogenicidad , Estructuras Animales/patología , Estructuras Animales/virología , Animales , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Histocitoquímica , Masculino , Ratones Noqueados , ARN Viral/análisis , Análisis de Supervivencia , Virulencia , Virus Zika/clasificación
12.
PLoS Negl Trop Dis ; 10(5): e0004658, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-27149521

RESUMEN

Zika virus (ZIKV) is a mosquito-borne pathogen which has recently spread beyond Africa and into Pacific and South American regions. Despite first being detected in 1947, very little information is known about the virus, and its spread has been associated with increases in Guillain-Barre syndrome and microcephaly. There are currently no known vaccines or antivirals against ZIKV infection. Progress in assessing interventions will require the development of animal models to test efficacies; however, there are only limited reports on in vivo studies. The only susceptible murine models have involved intracerebral inoculations or juvenile animals, which do not replicate natural infection. Our report has studied the effect of ZIKV infection in type-I interferon receptor deficient (A129) mice and the parent strain (129Sv/Ev) after subcutaneous challenge in the lower leg to mimic a mosquito bite. A129 mice developed severe symptoms with widespread viral RNA detection in the blood, brain, spleen, liver and ovaries. Histological changes were also striking in these animals. 129Sv/Ev mice developed no clinical symptoms or histological changes, despite viral RNA being detectable in the blood, spleen and ovaries, albeit at lower levels than those seen in A129 mice. Our results identify A129 mice as being highly susceptible to ZIKV and thus A129 mice represent a suitable, and urgently required, small animal model for the testing of vaccines and antivirals.


Asunto(s)
Modelos Animales de Enfermedad , Receptor de Interferón alfa y beta/metabolismo , Infección por el Virus Zika/inmunología , Infección por el Virus Zika/virología , Virus Zika/clasificación , Animales , Encéfalo/patología , Susceptibilidad a Enfermedades , Femenino , Ratones , Ratones Noqueados , ARN Viral/genética , ARN Viral/metabolismo , Receptor de Interferón alfa y beta/deficiencia , Receptor de Interferón alfa y beta/genética , Virus Zika/patogenicidad , Infección por el Virus Zika/patología
13.
PLoS One ; 11(6): e0156637, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27272940

RESUMEN

Crimean-Congo Haemorrhagic Fever (CCHF) is a severe tick-borne disease, endemic in many countries in Africa, the Middle East, Eastern Europe and Asia. There is no approved vaccine currently available against CCHF. The most promising candidate, which has previously been shown to confer protection in the small animal model, is a modified Vaccinia Ankara virus vector expressing the CCHF viral glycoprotein (MVA-GP). It has been shown that MVA-GP induces both humoral and cellular immunogenicity. In the present study, sera and T-lymphocytes were passively and adoptively transferred into recipient mice prior to challenge with CCHF virus. Results demonstrated that mediators from both arms of the immune system were required to demonstrate protective effects against lethal challenge.


Asunto(s)
Glicoproteínas/inmunología , Virus de la Fiebre Hemorrágica de Crimea-Congo/inmunología , Fiebre Hemorrágica de Crimea/inmunología , Virus Vaccinia/inmunología , Proteínas Virales/inmunología , Vacunas Virales/inmunología , África/epidemiología , Animales , Asia/epidemiología , Línea Celular , Europa Oriental/epidemiología , Fiebre Hemorrágica de Crimea/epidemiología , Humanos , Inmunidad Celular , Inmunidad Humoral , Ratones , Medio Oriente/epidemiología
14.
Sci Rep ; 6: 30497, 2016 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-27465308

RESUMEN

Ebola virus (EBOV) is highly pathogenic, with a predisposition to cause outbreaks in human populations accompanied by significant mortality. An ovine polyclonal antibody therapy has been developed against EBOV, named EBOTAb. When tested in the stringent guinea pig model of EBOV disease, EBOTAb has been shown to confer protection at levels of 83.3%, 50% and 33.3% when treatment was first started on days 3, 4 and 5 post-challenge, respectively. These timepoints of when EBOTAb treatment was initiated correspond to when levels of EBOV are detectable in the circulation and thus mimic when treatment would likely be initiated in human infection. The effects of EBOTAb were compared with those of a monoclonal antibody cocktail, ZMapp, when delivered on day 3 post-challenge. Results showed ZMapp to confer complete protection against lethal EBOV challenge in the guinea pig model at this timepoint. The data reported demonstrate that EBOTAb is an effective treatment against EBOV disease, even when delivered late after infection.


Asunto(s)
Anticuerpos Antivirales/uso terapéutico , Ebolavirus/fisiología , Fiebre Hemorrágica Ebola/tratamiento farmacológico , Profilaxis Posexposición , Animales , Anticuerpos Monoclonales/sangre , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Antivirales/sangre , Especificidad de Anticuerpos/inmunología , Antígenos Virales/metabolismo , Ebolavirus/genética , Genoma Viral , Cobayas , Fiebre Hemorrágica Ebola/sangre , Fiebre Hemorrágica Ebola/patología , Hígado/patología , Hígado/virología , ARN Viral/sangre , Ovinos , Bazo/patología , Bazo/virología , Análisis de Supervivencia
15.
J Immunol Methods ; 348(1-2): 30-5, 2009 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-19560467

RESUMEN

Work with highly pathogenic material mandates the use of biological containment facilities, involving microbiological safety cabinets and specialist laboratory engineering structures typified by containment level 3 (CL3) and CL4 laboratories. Consequences of working in high containment are the practical difficulties associated with containing specialist assays and equipment often essential for experimental analyses. In an era of increased interest in biodefence pathogens and emerging diseases, immunological analysis has developed rapidly alongside traditional techniques in virology and molecular biology. For example, in order to maximise the use of small sample volumes, multiplexing has become a more popular and widespread approach to quantify multiple analytes simultaneously, such as cytokines and chemokines. The luminex microsphere system allows for the detection of many cytokines and chemokines in a single sample, but the detection method of using aligned lasers and fluidics means that samples often have to be analysed in low containment facilities. In order to perform cytokine analysis in materials from high containment (CL3 and CL4 laboratories), we have developed an appropriate inactivation methodology after staining steps, which although results in a reduction of median fluorescent intensity, produces statistically comparable outcomes when judged against non-inactivated samples. This methodology thus extends the use of luminex technology for material that contains highly pathogenic biological agents.


Asunto(s)
Contención de Riesgos Biológicos , Citocinas/análisis , Desinfectantes/farmacología , Formaldehído/farmacología , Inactivación de Virus , Biomarcadores/análisis , Desinfectantes/química , Formaldehído/química , Humanos , Mediciones Luminiscentes , Microesferas , Soluciones , Virus/efectos de los fármacos , Virus/inmunología , Virus/patogenicidad
16.
Eur J Immunol ; 37(4): 925-34, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17330818

RESUMEN

The role of CD44 in T cell biology remains incompletely understood. Although studies using anti-CD44 antibodies have implicated this cell adhesion molecule in a variety of important T cell processes, few T cell defects have been reported in CD44-deficient mice. We have assessed the requirement for CD44 in T cell development and mature T cell function by analyzing mice in which CD44(-/-) and WT cells were produced simultaneously. In mixed (CD44(-/-) + CD44(+/+)) bone marrow chimeras, production of CD44(-/-) T cells was shown to be reduced compared to WT cells due to inefficient intrathymic development. In addition, mature CD44(-/-) CD8(+) T cells generated a substantially lower response than WT T cells after infection of mice with lymphocytic choriomeningitis virus, with the reduction in response apparent in both lymphoid and non-lymphoid tissues. Overall, these results demonstrate a poor capacity of CD44(-/-) T lineage cells to compete with WT cells at multiple levels, implicating CD44 in normal T cell function.


Asunto(s)
Comunicación Celular/inmunología , Diferenciación Celular/inmunología , Receptores de Hialuranos/fisiología , Subgrupos de Linfocitos T/inmunología , Linfocitos T/inmunología , Animales , Infecciones por Arenaviridae/inmunología , Comunicación Celular/genética , Diferenciación Celular/genética , Receptores de Hialuranos/biosíntesis , Receptores de Hialuranos/genética , Virus de la Coriomeningitis Linfocítica/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Subgrupos de Linfocitos T/metabolismo , Linfocitos T/clasificación , Linfocitos T/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA