Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Infect Genet Evol ; 30: 120-127, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25528137

RESUMEN

Infectious bursal disease virus (IBDV) causes an economically significant disease of young chickens worldwide. The emergence of very virulent IBDV (vvIBDV) strains has brought more challenges for effective prevention and control of this disease. The aim of the present study was to characterize four IBDV isolates from various regions of China between late 1990s and recent years and to compare them with previously isolated European IBDV strains. In this study, one Chinese vvIBDV strain isolated in 1999 and three strains isolated between 2005 and 2011 were analyzed at the genetic, antigenic and pathogenic levels. Strain SH99 was closely related and clustered in the same genetic lineage as the typical vvIBDV based on the genomic sequences of segments A and B. However, the three more recent Chinese vvIBDV (HLJ0504, HeB10 and HuN11) showed several genetic changes in both segments and clustered in a distinct lineage from the typical vvIBDV and the previously known Chinese vvIBDV. Based on the binding to a panel of neutralizing monoclonal antibodies in antigen capture enzyme-linked immunosorbent assays, all Chinese vvIBDVs exhibited similar antigenicity with the European typical vvIBDV strains. Nonetheless, the pathogenicity caused by the recent Chinese vvIBDV was higher than that induced by the European typical vvIBDV. This study calls for a sustained surveillance of IBD situation in China in order to support a better prevention and control of the disease.


Asunto(s)
Infecciones por Birnaviridae/virología , Virus de la Enfermedad Infecciosa de la Bolsa/clasificación , Virus de la Enfermedad Infecciosa de la Bolsa/genética , Animales , Anticuerpos Neutralizantes/inmunología , Antígenos Virales/química , Infecciones por Birnaviridae/veterinaria , Embrión de Pollo , Pollos/virología , China , Virus de la Enfermedad Infecciosa de la Bolsa/inmunología , Virus de la Enfermedad Infecciosa de la Bolsa/patogenicidad
2.
PLoS One ; 9(7): e102740, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25036224

RESUMEN

Four avian metapneumovirus (AMPV) subgroups (A-D) have been reported previously based on genetic and antigenic differences. However, until now full length sequences of the only known isolates of European subgroup C and subgroup D viruses (duck and turkey origin, respectively) have been unavailable. These full length sequences were determined and compared with other full length AMPV and human metapneumoviruses (HMPV) sequences reported previously, using phylogenetics, comparisons of nucleic and amino acid sequences and study of codon usage bias. Results confirmed that subgroup C viruses were more closely related to HMPV than they were to the other AMPV subgroups in the study. This was consistent with previous findings using partial genome sequences. Closer relationships between AMPV-A, B and D were also evident throughout the majority of results. Three metapneumovirus "clusters" HMPV, AMPV-C and AMPV-A, B and D were further supported by codon bias and phylogenetics. The data presented here together with those of previous studies describing antigenic relationships also between AMPV-A, B and D and between AMPV-C and HMPV may call for a subclassification of metapneumoviruses similar to that used for avian paramyxoviruses, grouping AMPV-A, B and D as type I metapneumoviruses and AMPV-C and HMPV as type II.


Asunto(s)
Genoma Viral/genética , Metapneumovirus/genética , Secuencia de Aminoácidos , Secuencia de Bases , Codón/genética , Datos de Secuencia Molecular , Filogenia , Alineación de Secuencia
3.
Virol J ; 11: 74, 2014 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-24762011

RESUMEN

BACKGROUND: H5 low pathogenic avian influenza virus (LPAIV) infection in domestic ducks is a major problem in duck producing countries. Their silent circulation is an ongoing source of potential highly pathogenic or zoonotic emerging strains. To prevent such events, vaccination of domestic ducks might be attempted but remains challenging. Currently licensed vector vaccines derived from H5N1 HPAIV possess clade 0, clade 2.2 or clade 2.3.4 HA sequences: selection of the best HA candidate inducing the largest cross protection is a key issue. For this purpose, DNA immunization of specific pathogen free Muscovy ducks was performed using different synthetic codon optimized (opt) or native HA genes from H5N2 LPAIV and several H5N1 HPAIV clade 2.1, 2.2.1 and 2.3.4. Humoral cross-immunity was assessed 3 weeks after boost by hemagglutination inhibition (HI) and virus neutralization (VN) against three French H5 LPAIV antigens. FINDINGS: Vaccination with LP H5N2 HA induced the highest VN antibody titre against the homologous antigen; however, the corresponding HI titre was lower and comparable to HI titres obtained after immunization with opt HA derived from clades 2.3.4 or 2.1. Compared to the other HPAIV-derived constructs, vaccination with clade 2.3.4 opt HA consistently induced the highest antibody titres in HI and VN, when tested against all three H5 LPAIV antigens and H5N2 LPAIV, respectively: differences in titres against this last strain were statistically significant. CONCLUSION: The present study provides a standardized method to assess cross-immunity based on HA immunogenicity alone, and suggests that clade 2.3.4-derived recombinant vaccines might be the optimal candidates for further challenge testing to vaccinate domestic Muscovy ducks against H5 LPAIV.


Asunto(s)
Anticuerpos Antivirales/sangre , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Subtipo H5N1 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/inmunología , Gripe Aviar/prevención & control , Enfermedades de las Aves de Corral/prevención & control , Vacunas de ADN/inmunología , Animales , Protección Cruzada , Patos , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Subtipo H5N1 del Virus de la Influenza A/genética , Subtipo H5N2 del Virus de la Influenza A/genética , Subtipo H5N2 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/administración & dosificación , Vacunas contra la Influenza/genética , Gripe Aviar/inmunología , Enfermedades de las Aves de Corral/inmunología , Vacunas de ADN/administración & dosificación , Vacunas de ADN/genética
4.
Vaccine ; 31(38): 4121-8, 2013 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-23845804

RESUMEN

Vaccination protocols were evaluated in one-day old Muscovy ducklings, using an experimental Newcastle disease recombinant vaccine (vNDV-H5) encoding an optimized synthetic haemagglutinin gene from a clade 2.2.1 H5N1 highly pathogenic (HP) avian influenza virus (AIV), either as a single administration or as a boost following a prime inoculation with a fowlpox vectored vaccine (vFP89) encoding a different H5 HP haemagglutinin from an Irish H5N8 strain. These vaccination schemes did not induce detectable levels of serum antibodies in HI test using a clade 2.2.1 H5N1 antigen, and only induced H5 ELISA positive response in less than 10% of vaccinated ducks. However, following challenge against a clade 2.2.1 HPAIV, both protocols afforded full clinical protection at six weeks of age, and full protection against mortality at nine weeks. Only the prime-boost vaccination (vFP89+vNDV-H5) was still fully protecting Muscovy ducks against disease and mortality at 12 weeks of age. Reduction of oropharyngeal shedding levels was also constantly observed from the onset of the follow-up at 2.5 or three days post-infection in vaccinated ducks compared to unvaccinated controls, and was significantly more important for vFP89+vNDV-H5 vaccination than for vNDV-H5 alone. Although the latter vaccine is shown immunogenic in one-day old Muscovy ducks, the present work is original in demonstrating the high efficacy of the successive administration of two different vector vaccines encoding two different H5 in inducing lasting protection (at least similar to the one induced by an inactivated reassortant vaccine, Re-5). In addition, such a prime-boost schedule allows implementation of a DIVA strategy (to differentiate vaccinated from infected ducks) contrary to Re-5, involves easy practice on the field (with injection at the hatchery and mass vaccination later on), and should avoid eventual interference with NDV maternally derived antibodies. Last, the HA insert could be updated according to the epidemiological situation.


Asunto(s)
Patos/virología , Viruela Aviar/genética , Inmunización Secundaria/métodos , Subtipo H5N1 del Virus de la Influenza A/patogenicidad , Vacunas contra la Influenza/farmacología , Gripe Aviar/prevención & control , Virus de la Enfermedad de Newcastle/genética , Animales , Encéfalo/virología , Viruela Aviar/inmunología , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Inmunidad Humoral , Vacunas contra la Influenza/administración & dosificación , Vacunas contra la Influenza/genética , Vacunas contra la Influenza/inmunología , Gripe Aviar/mortalidad , Gripe Aviar/virología , Virus de la Enfermedad de Newcastle/inmunología , Resultado del Tratamiento , Vacunas Sintéticas , Esparcimiento de Virus/inmunología
5.
J Virol Methods ; 193(1): 121-7, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23707921

RESUMEN

Determining the sequence of non-segmented, negative sense RNA viral genomes is far from routine and often requires the application of several techniques. In this study, an existing method used currently just for determination of the genomic 3' extremity was used to determine both the 3' and 5' sequence extremities of a Newcastle disease virus and an avian metapneumovirus. This was achieved with a single 3' nucleotide tailing reaction of both the genomic RNA and the full length, positive sense, antigenomic RNA, followed by a single reverse transcription reaction targeted to the common polynucleotide tails, and then individual PCRs specific for each extremity using PCR primers derived from the sequence of the RT primer or from neighbouring virus sequences known previously. For each virus the method was employed separately. Sequences from both viruses were in agreement with those reported previously for other paramyxoviruses, yet one extra base at the 3' and one extra base at the 5' were identified for the avian metapneumovirus. In this study, importantly, the newly determined extremities maintained the complementarity known to exist between the extremities of these viruses. The method was equally successful with both viruses and can be tailored easily to function with other non-segmented, negative sense viruses through minor modification of only the primer sequences.


Asunto(s)
Regiones no Traducidas 5' , Metapneumovirus/genética , Virus de la Enfermedad de Newcastle/genética , ARN Viral/genética , Análisis de Secuencia de ADN/métodos , Reacción en Cadena de la Polimerasa , ARN Viral/química , Transcripción Reversa
6.
Avian Pathol ; 42(3): 248-52, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23607441

RESUMEN

Turkey coronavirus (TCoV) is a gammacoronavirus (Coronaviridae, Nidovirales) responsible for digestive disorders in young turkeys. TCoV has been associated with poult enteritis complex, a syndrome that severely affects turkey production. No medical prophylaxis exists to control TCoV, therefore sanitary measures such as cleaning and disinfection are essential. It is thus important to evaluate temperatures that allow persistence of TCoV in the environment. Two series of aliquots of a suspension of a French isolate of TCoV (Fr TCoV) were stored at room temperature or +4°C for 0 to 40 days. As TCoV does not grow in cell culture, the presence of residual infectious TCoV in the stored samples was tested by inoculating embryonated specific pathogen free turkey eggs. As TCoV does not induce lesions in the embryo, virus replication in the jejunum and ileum of the embryos was detected 4 days post inoculation, using RNA extraction and a real-time reverse transcriptase-polymerase chain reaction based on the nucleocapsid gene. No surviving virus was detected after 10 days storage at +21.6±1.4°C or after 40 days storage at +4.1±1.6°C, these temperatures being representative of the mean summer and winter temperatures, respectively, in the major French poultry-producing region. The relatively short survival of the virus at room temperature should contribute to limited virus survival during summer months. However, infectious virus was still detected after 20 days storage at the cooler temperatures, a finding that suggests prolonged survival of Fr TCoV and easier transmission between poultry farms in a cool environment are possible.


Asunto(s)
Coronavirus del Pavo/fisiología , Temperatura , Pavos/virología , Replicación Viral/fisiología , Animales , Coronavirus del Pavo/genética , Genoma Viral/genética , Proteínas de la Nucleocápside/genética , Óvulo/virología , Reacción en Cadena en Tiempo Real de la Polimerasa/veterinaria , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/veterinaria , Análisis de Supervivencia
7.
J Virol ; 87(5): 2767-80, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23269788

RESUMEN

Infectious bursal disease virus (IBDV) causes an economically significant disease of chickens worldwide. Very virulent IBDV (vvIBDV) strains have emerged and induce as much as 60% mortality. The molecular basis for vvIBDV pathogenicity is not understood, and the relative contributions of the two genome segments, A and B, to this phenomenon are not known. Isolate 94432 has been shown previously to be genetically related to vvIBDVs but exhibits atypical antigenicity and does not cause mortality. Here the full-length genome of 94432 was determined, and a reverse genetics system was established. The molecular clone was rescued and exhibited the same antigenicity and reduced pathogenicity as isolate 94432. Genetically modified viruses derived from 94432, whose vvIBDV consensus nucleotide sequence was restored in segment A and/or B, were produced, and their pathogenicity was assessed in specific-pathogen-free chickens. We found that a valine (position 321) that modifies the most exposed part of the capsid protein VP2 critically modified the antigenicity and partially reduced the pathogenicity of 94432. However, a threonine (position 276) located in the finger domain of the virus polymerase (VP1) contributed even more significantly to attenuation. This threonine is partially exposed in a hydrophobic groove on the VP1 surface, suggesting possible interactions between VP1 and another, as yet unidentified molecule at this amino acid position. The restored vvIBDV-like pathogenicity was associated with increased replication and lesions in the thymus and spleen. These results demonstrate that both genome segments influence vvIBDV pathogenicity and may provide new targets for the attenuation of vvIBDVs.


Asunto(s)
Infecciones por Birnaviridae/veterinaria , Virus de la Enfermedad Infecciosa de la Bolsa/genética , Virus de la Enfermedad Infecciosa de la Bolsa/patogenicidad , Enfermedades de las Aves de Corral/virología , Proteínas Estructurales Virales/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Infecciones por Birnaviridae/mortalidad , Infecciones por Birnaviridae/patología , Infecciones por Birnaviridae/virología , Embrión de Pollo , Pollos/virología , Simulación de Dinámica Molecular , Enfermedades de las Aves de Corral/patología , Análisis de Secuencia de ARN , Bazo/virología , Timo/virología , Proteínas Estructurales Virales/química
8.
Avian Dis ; 54(1 Suppl): 232-8, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20521637

RESUMEN

Fowlpox (FP)-vectored avian influenza (FP-AI) vaccines are used in 1-day-old chickens, but they have also recently been shown to be immunogenic in ducks. The objectives of this work were 1) to evaluate safety and to compare the immunogenicity in ducks of three poxvirus vectors (fowlpox, canarypox, and vaccinia) expressing the same hemagglutinin gene from an H5N1 isolate, 2) to study the effect of the dose of the FP-AI and the presence of an adjuvant in 1-day-old Pekin ducks on antibody response after a boost with inactivated vaccine given 3 wk later, and 3) to confirm the immunogenicity of such a heterologous prime-boost vaccination scheme in 1-day-old Muscovy ducks. Immunogenicity induced by the three poxvirus vectors was comparable, and the FP vector was selected for the other studies. As published previously, there was a strong dose effect of the FP-AI priming on the hemagglutination inhibition (HI) titers induced after the boost with an inactivated vaccine. In contrast, the two tested adjuvants did not significantly increase the activity of FP-AI priming. The heterologous prime-boost regimen given to both Muscovy and Pekin ducklings at 1 and 14 or 21 days of age, respectively, was shown to be at least as immunogenic as two administrations of inactivated vaccines given at 2 and 5 wk of age. However, HI antibody titers were of short duration for both vaccine schemes, and their persistence was heterogeneous among individual birds.


Asunto(s)
Avipoxvirus , Patos/genética , Vacunas contra la Influenza/inmunología , Gripe Aviar/prevención & control , Animales , Esquema de Medicación , Femenino , Subtipo H5N1 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/administración & dosificación , Factores de Tiempo , Vacunas de Productos Inactivados/administración & dosificación , Vacunas de Productos Inactivados/inmunología , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/inmunología
9.
Avian Dis ; 54(1 Suppl): 502-8, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20521685

RESUMEN

Highly pathogenic (HP) avian influenza A viruses (AIVs) subtype H5N1 (subclade 2.2) were detected in wild birds during outbreaks in France during winter 2006 and summer 2007 in la Dombes wetlands (eastern France) and in Moselle wetlands (northeastern France), respectively. Blood samples from apparently healthy wild birds were collected in 2006 and 2007 from the end of the outbreak to several weeks after the influenza A outbreak inside and outside the contaminated areas, and in 2008 outside the contaminated areas. The samples were tested for the presence and/or quantitation of serum antibodies to influenza A subtypes H5 and N1 using hemagglutination inhibition tests (HITs), a commercial N1-specific enzyme-linked immunosorbent assay kit, and virus neutralization assay. In the HIT, low pathogenicity (LP) and HP H5 AIVs (belonging to H5N1, H5N2, and H5N3 subtypes) were used as antigens. One hundred mute swans were bled in the la Dombes outbreak area in 2006. During 2007, 46 mallards, 69 common pochards, and 59 mute swans were sampled in the Moselle outbreak area. For comparison, blood samples were also collected in 2007 from 60 mute swans from the Marne department where no HP H5N1 influenza A cases have been reported, and in 2008 from 111 sacred ibises in western France where no HP H5N1 influenza A infections in wild birds have been reported either. Mute swans (irrespective of their origin and time of sampling) and sacred ibises (from an area with no known outbreaks) had the highest prevalence of positive sera in the H5 HIT (49-69% and 64%, respectively). The prevalence of anti-H5 antibodies in mallards and common pochards was lower (28% and 27%, respectively). Positive H5- and N1-antibody responses were also significantly associated in swans (irrespective of their origin and time of sampling) and in sacred ibises. However, in swans from the area without outbreaks, the HIT titer against an H5N1 LPAIV was significantly higher than against an H5N1 2.2.1 HPAIV, whereas no difference could be shown for swans from the outbreak areas sampled in 2006 and 2007. These results suggest that ibises and swans from areas without declared outbreaks had acquired humoral immunity after AIV infections with subtypes H5 and N1 but independently from HP H5N1 infection. However, for swans living in outbreak areas, it cannot be excluded that this immunity might result from either a subclinical or a nonlethal infection by HP H5N1.


Asunto(s)
Anticuerpos Antivirales/sangre , Aves/sangre , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Subtipo H5N1 del Virus de la Influenza A/clasificación , Gripe Aviar/sangre , Neuraminidasa/inmunología , Animales , Francia/epidemiología , Gripe Aviar/epidemiología , Neuraminidasa/clasificación , Pruebas de Neutralización/veterinaria , Humedales
10.
Avian Dis ; 54(1 Suppl): 538-47, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20521691

RESUMEN

Highly pathogenic (HP) H5N1 avian influenza (AI) is enzootic in several countries of Asia and Africa and constitutes a major threat, at the world level, for both animal and public health. Ducks play an important role in the epidemiology of AI, including HP H5N1 AI. Although vaccination can be a useful tool to control AI, duck vaccination has not proved very efficient in the field, indicating a need to develop new vaccines and a challenge model to evaluate the protection for duck species. Although Muscovy duck is the duck species most often reared in France, the primary duck-producing country in Europe, and is also produced in Asia, it is rarely studied. Our team recently demonstrated a good cross-reactivity with hemagglutinin from clade 2.2 and inferred that this could be a good vaccine candidate for ducks. Two challenges using two French H5N1 HP strains, 1) A/mute swan/France/06299/06 (Swan/06299), clade 2.2.1, and 2) A/mute swan/France/070203/07 (Swan/070203), clade 2.2 (but different from subclade 2.2.1), were performed (each) on 20 Muscovy ducks (including five contacts) inoculated by oculo-nasal route (6 log10 median egg infectious doses per duck). Clinical signs were recorded daily, and cloacal and oropharyngeal swabs were collected throughout the assay. Autopsies were done on all dead ducks, and organs were taken for analyses. Virus was measured by quantitative reverse transcriptase-PCR based on the M gene AI virus. Ducks presented severe nervous signs in both challenges. Swan/070203 strain led to 80% morbidity (12/15 sick ducks) and 73% mortality (11/15 ducks) at 13.5 days postinfection (dpi), whereas Swan/06299 strain produced 100% mortality at 6.5 dpi. Viral RNA load was significantly lower via the cloacal route than via the oropharyngeal route in both trials, presenting a peak in the first challenge at 3.5 dpi and being more stable in the second challenge. The brain was the organ containing the highest viral RNA load in both challenges. Viral RNA load in a given organ was similar or statistically significantly higher in ducks challenged with Swan/06299 strain. Thus, the Swan/06299 strain was more virulent and could be used as a putative challenge model. Moreover, challenged ducks and contacts contained the same amounts of viral RNA load, demonstrating the rapid and efficient transmission of H5N1 HP in Muscovy ducks in our experimental conditions.


Asunto(s)
Patos , Subtipo H5N1 del Virus de la Influenza A/clasificación , Gripe Aviar/virología , Animales , Encéfalo/virología , Subtipo H5N1 del Virus de la Influenza A/patogenicidad , Gripe Aviar/mortalidad , Gripe Aviar/patología , Riñón/virología , Páncreas/virología , ARN Viral/análisis , Tráquea/virología , Carga Viral , Esparcimiento de Virus
11.
Avian Pathol ; 37(1): 15-23, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18202945

RESUMEN

Highly pathogenic avian influenza (HPAI) viruses of subtype H5N1 have spread since late 2003 in East and Southeast Asia. In April 2005, a large-scale outbreak of H5N1 infection that occurred in migratory waterfowl in Qinghai Lake nature reserve in western China, killing more than 6000 wild birds, appeared to be the beginning of a epizootic that caused outbreaks in domestic and wild birds in nearly 60 countries from Central Asia, the Middle East, Europe and Africa. The first case of Asian lineage HPAI H5N1 virus in France was described in dead wild ducks (Common pochard) in the east of France in mid-February 2006. Up to the end of April, 42 HPAI H5N1 viruses were identified from about 60 wild birds belonging to different species and one outbreak occurred in commercial turkeys. To establish genetic relationships with other HPAI H5N1 viruses, 12 selected viruses were subjected to phylogenetic analysis. Genotyping and genetic analyses revealed that the French viruses were very similar to those of the 'Qinghai-like' sublineage and belonged to clade 2.2. However, two related but distinct genetic subgroups were identified, indicating that two different viruses were circulating in France at the same time and in the same area. Viruses of one subgroup were highly similar to one identified in Bavaria in Germany (A/mallard/Bavaria/1/2006). More surprisingly, French viruses belonging to the other subgroup retained the cleavage motif PQGERKRKKR/G, which is unique among the known HPAI H5N1 viruses. Our results confirmed that multiple H5N1 genogroups were present in Western Europe in early 2006.


Asunto(s)
Anseriformes/virología , Subtipo H5N1 del Virus de la Influenza A/patogenicidad , Gripe Aviar/epidemiología , Gripe Aviar/virología , Pavos/virología , Secuencias de Aminoácidos , Animales , Francia/epidemiología , Hemaglutininas Virales/química , Hemaglutininas Virales/genética , Subtipo H5N1 del Virus de la Influenza A/genética , Filogenia
12.
J Gen Virol ; 89(Pt 2): 525-533, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18198384

RESUMEN

Subgroup A avian metapneumoviruses lacking either the SH or G gene or the M2-2 open reading frame were generated by using a reverse-genetics approach. The growth properties of these viruses were studied in vitro and in vivo in their natural host. Deletion of the SH gene alone resulted in the generation of a syncytial-plaque phenotype and this was reversed by the introduction of the SH gene from a subgroup B, but not a subgroup C, virus. Infected turkeys were assessed for antibody production and the presence of viral genomic RNA in tracheal swabs. The virus with a deleted SH gene also showed the greatest impairment of replication both in cell culture and in infected turkeys. This contrasts with the situation with other pneumoviruses in culture and in model animals, where deletion of the SH gene results in little effect upon viral yield and a good antibody response. Replication of the G- and M2-2-deleted viruses was impaired more severely in turkeys than in cell culture, with only some animals showing evidence of virus growth and antibody production. There was no correlation between virus replication and antibody response, suggesting that replication sites other than the trachea may be important for induction of antibody responses.


Asunto(s)
Eliminación de Gen , Metapneumovirus/genética , Infecciones por Paramyxoviridae/inmunología , Proteínas del Envoltorio Viral/inmunología , Proteínas Virales/genética , Animales , Anticuerpos Antivirales , Línea Celular , Genes Virales/genética , Metapneumovirus/inmunología , Metapneumovirus/patogenicidad , Metapneumovirus/fisiología , Sistemas de Lectura Abierta , Infecciones por Paramyxoviridae/virología
13.
FEBS Lett ; 551(1-3): 159-64, 2003 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-12965222

RESUMEN

The liver endoplasmic reticulum glucose-6-phosphatase catalytic subunit (G6PC1) catalyses glucose 6-phosphate hydrolysis during gluconeogenesis and glycogenolysis. The highest glucose-6-phosphatase activities are found in the liver and the kidney; there have been many reports of glucose 6-phosphate hydrolysis in other tissues. We cloned a new G6Pase isoform (G6PC3) from human brain encoded by a six-exon gene (chromosome 17q21). G6PC3 protein was able to hydrolyse glucose 6-phosphate in transfected Chinese hamster ovary cells. The optimal pH for glucose 6-phosphate hydrolysis was lower and the K(m) higher relative to G6PC1. G6PC3 preferentially hydrolyzed other substrates including pNPP and 2-deoxy-glucose-6-phosphate compared to the liver enzyme.


Asunto(s)
Glucosa-6-Fosfatasa/genética , Glucosa-6-Fosfatasa/metabolismo , Secuencia de Aminoácidos , Animales , Células CHO , Clonación Molecular , Cricetinae , Componentes del Gen , Glucosa-6-Fosfatasa/química , Humanos , Isoenzimas/genética , Isoenzimas/metabolismo , Datos de Secuencia Molecular , ARN Mensajero/metabolismo , Alineación de Secuencia , Análisis de Secuencia de Proteína , Especificidad por Sustrato , Distribución Tisular
14.
Biochim Biophys Acta ; 1623(1): 21-8, 2003 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-12957713

RESUMEN

Previous studies have described the isolation of a new metalloprotease with a strict specificity for the amide bonds of peptide substrates having a threonine residue at the P1' position [Biochem. Biophys. Res. Commun. 256 (1999) 307]. The present work reports the physico-chemical properties of the enzyme which enable the optimal conditions for the digestion of proteins by the protease to be determined. At pH 8.2 and up to 37 degrees C, the enzyme possesses a good proteolytic activity and is stable for at least 12 h. The protease is sensitive to detergents and dithiol-reducing agents so that these chemicals must be eliminated after treatment of the protein substrate when this needs to be denatured and reduced before its hydrolysis by the enzyme. An increase in the enzymatic activity is observed in the presence of urea up to a 2.0 M concentration, beyond which the activity decreases. The enzyme can also be used in the presence of organic solvents such as acetonitrile, isopropanol or dioxane (10%, v/v) without loss of activity. Studies performed with antibodies raised against the purified endoprotease Thr-N indicated the absence of cross-immunoinactivation and cross-immunoprecipitation with all tested proteases. Also, no homology of sequence was found with the proteases indexed in the databases. Thus, our results show that endoprotease Thr-N not only represents an original protease by its unique specificity but also by its immunological and molecular properties.


Asunto(s)
Endopeptidasas/química , Endopeptidasas/inmunología , Metaloproteasas/química , Metaloproteasas/inmunología , Secuencia de Aminoácidos , Animales , Endopeptidasas/genética , Endopeptidasas/aislamiento & purificación , Activación Enzimática , Estabilidad de Enzimas , Concentración de Iones de Hidrógeno , Metaloproteasas/genética , Metaloproteasas/aislamiento & purificación , Datos de Secuencia Molecular , Peso Molecular , Conejos , Análisis de Secuencia de Proteína , Caracoles/química , Caracoles/enzimología , Temperatura
15.
Comp Biochem Physiol B Biochem Mol Biol ; 135(3): 503-10, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12831770

RESUMEN

The profile of sedimentation on a 4-20% (w/v) linear sucrose gradient of the digestive juice of the mollusk Archachatina ventricosa revealed the presence of at least four specific proteases. A first peak, corresponding to a sedimentation coefficient of 3.9 S, contained two endoproteases that could be assayed, one with Leu-pNA and the other with Met-pNA. Their activity was maximal at pH 8.0 and increased in the presence of Ca(2+) ions. Both enzymes were inhibited by the chelating agent 1,10-phenanthroline but their thermal inactivation kinetics were different. A second protease peak was observed at 6.8 S and corresponded to a metallo-endoprotease that hydrolyzed with a maximal activity at pH 8.0 only the amide bonds of peptide substrates having a threonine residue at the P1' position. A last protease peak identified at 9.0 S contained a protease that preferentially acted on tripeptides, such as Val-Pro-Leu (diprotin B) and Thr-Val-Leu, releasing the C-terminal residue. Unlike the proteases identified in the two other peaks, its activity was maximal at acid pH (5.0) and was inhibited by the serine protease inhibitors. Together these results show the potential of A. ventricosa as a source of specific proteases.


Asunto(s)
Sistema Digestivo/enzimología , Endopeptidasas/metabolismo , Moluscos/enzimología , Animales , Endopeptidasas/análisis , Calor , Concentración de Iones de Hidrógeno , Hidrólisis , Cinética , Desnaturalización Proteica , Especificidad por Sustrato
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...