Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Sleep Breath ; 28(1): 123-131, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-37428352

RESUMEN

OBJECTIVE: To investigate the role of TWIK-related acid-sensitive potassium channels TASK-1 and TASK-3 in the mechanism of asthma combined with obstructive sleep apnea (OSA) in mice. METHOD: C57BL/6 mice were randomly divided into four groups: control group (NS-RA), asthma group (OVA-RA), OSA group (NS-IH), and asthma combined with OSA group (OVA-IH). After monitoring lung function in each group, the expression levels of TASK-1 and TASK-3 mRNA and protein in lung tissues were measured, and the correlation between the changes of both and lung function was analyzed. RESULTS: A total of 64 male mice were studied. Penh, serum IgE concentrations, and the percentage of eosinophils in bronchoalveolar lavage fluid (BALF) were higher in OVA-RA and OVA-IH mice compared with NS-RA (P < 0.05),while the above indexes were slightly elevated in NS-IH mice compared with NS-RA (P > 0.05), where the Penh and the percentage of eosinophils in BALF was higher in OVA-IH mice than NS-IH (P < 0.05).Increased TASK-3 mRNA expression (P < 0.05) as well as TASK-1 and TASK-3 protein expression (P > 0.05) in lung tissues of OVA-RA and NS-IH mice compared with NS-RA, and TASK-3 mRNA expression was slightly more in the OVA-IH group compared with NS-RA (P > 0.05), but less compared with OVA-RA (P < 0.05) or NS-IH (P > 0.05), while TASK-1 and TASK-3 protein expression was increased in the OVA-IH group compared with the remaining three groups, and TASK-3 protein expression was associated with lung function impairment was positively correlated with the degree of lung function impairment (P < 0.05). CONCLUSION: Task-1 and Task-3 may be involved in the pathogenesis of asthma with OSA by affecting lung function.


Asunto(s)
Asma , Animales , Masculino , Ratones , Líquido del Lavado Bronquioalveolar , Modelos Animales de Enfermedad , Pulmón/metabolismo , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , ARN Mensajero
2.
Aesthetic Plast Surg ; 42(5): 1298-1303, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30097671

RESUMEN

OBJECTIVE: The acellular dermal matrix (ADM) used in correcting the tear trough deformity has been reported, but there were only a few cases. The long-term effectiveness of ADM was not clear. We aim to discuss the technique and the effect of using ADM to correct the tear trough deformity through more cases. METHODS: A retrospective study was conducted from January 2012 to January 2017. Twenty-six patients who showed obvious tear trough deformity with moderate or severe orbital fat bulging and excess of lower eyelid skin were treated with ADM to improve the appearance of the midface. Follow-up was performed for 2-12 months in 26 cases. The level of postoperative satisfaction was assessed by interview during the follow-up and rated as very satisfied, satisfied, acceptable, or unacceptable. RESULT: Twenty patients were very satisfied for having achieved complete correction. Three patients were satisfied for having achieved obvious improvement. Three patients felt the results were just acceptable and were refilled because of the insufficiency of the filler. No one was unacceptable. There were no complications such as rapid resorption, rejection, or inflammation. CONCLUSION: The method of using ADM for the correction of tear trough deformity has the advantages of low absorption rate, good appearance, and high security. It provides a new choice for the treatment of tear trough deformity. LEVEL OF EVIDENCE IV: This journal requires that authors assign a level of evidence to each article. For a full description of these Evidence-Based Medicine ratings, please refer to the Table of Contents or the online Instructions to Authors www.springer.com/00266 .


Asunto(s)
Dermis Acelular , Tejido Adiposo/trasplante , Blefaroplastia/métodos , Párpados/cirugía , Conducto Nasolagrimal/cirugía , Adulto , Distribución de Chi-Cuadrado , China , Estudios de Cohortes , Estética , Párpados/anomalías , Femenino , Humanos , Masculino , Persona de Mediana Edad , Conducto Nasolagrimal/anomalías , Órbita/anomalías , Órbita/cirugía , Satisfacción del Paciente/estadística & datos numéricos , Estudios Retrospectivos , Medición de Riesgo , Cirugía Plástica/métodos
3.
J Clin Microbiol ; 53(5): 1727-30, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25694519

RESUMEN

For detection of infectious laryngotracheitis virus (ILTV) antibody, glycoprotein B-, C-, and D-based enzyme-linked immunosorbent assays (B-, C-, and D-ELISAs, respectively) were developed. The B- and D-ELISAs showed enhanced detection of anti-ILTV antibodies in infected chickens compared to that of the commercial ELISA. Furthermore, the D-ELISA was efficient in detecting seroconversion with vectored vaccine, using recombinant Newcastle disease virus (rNDV) expressing glycoprotein D (gD) as the vaccine vector.


Asunto(s)
Anticuerpos Antivirales/sangre , Infecciones por Herpesviridae/veterinaria , Herpesvirus Gallináceo 1/aislamiento & purificación , Enfermedades de las Aves de Corral/diagnóstico , Enfermedades de las Aves de Corral/virología , Pruebas Serológicas/métodos , Medicina Veterinaria/métodos , Animales , Pollos , Ensayo de Inmunoadsorción Enzimática/métodos , Infecciones por Herpesviridae/diagnóstico , Infecciones por Herpesviridae/virología , Herpesvirus Gallináceo 1/inmunología
5.
Proc Natl Acad Sci U S A ; 108(45): 18406-11, 2011 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-22042859

RESUMEN

IgG was traditionally thought to neutralize virions by blocking their attachment to or penetration into mucosal epithelial cells, a common site of exposure to viruses. However, we describe an intracellular neutralizing action for an influenza hemagglutinin-specific monoclonal antibody, Y8-10C2 (Y8), which has neutralizing activity only at an acidic pH. When Y8 was applied to the basolateral surface of Madin-Darby canine kidney cells expressing the rat neonatal Fc receptor for IgG (FcRn), it significantly reduced viral replication following apical exposure of the cell monolayer to influenza virus. Virus neutralization by Y8 mAb was dependent on FcRn expression and its transport of IgG. As both FcRn and Y8 mAb bind their partners only at acidic pH, the Y8 mAb is proposed to carry out its antiviral activity intracellularly. Furthermore, the virus, Y8 mAb, and FcRn colocalized within endosomes, possibly inhibiting the fusion of viral envelopes with endosomal membranes during primary uncoating, and preventing the accumulation of the neutralized viral nucleoprotein antigen in the nucleus. Prophylactic administration of Y8 mAb before viral challenge in WT mice, but not FcRn-KO mice, conferred protection from lethality, prevented weight loss, resulted in a significant reduction in pulmonary virus titers, and largely reduced virus-induced lung pathology. Thus, this study reveals an intracellular mechanism for viral neutralization in polarized epithelial cells that is dependent on FcRn-mediated transport of neutralizing IgG.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Inmunoglobulina G/metabolismo , Infecciones por Orthomyxoviridae/inmunología , Receptores Fc/fisiología , Animales , Animales Recién Nacidos , Perros , Ratones , Ratones Noqueados , Orthomyxoviridae/inmunología , Ratas , Receptores Fc/genética , Receptores Fc/inmunología
6.
PLoS Pathog ; 6(10): e1001145, 2010 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-20976194

RESUMEN

A novel, swine-origin influenza H1N1 virus (H1N1pdm) caused the first pandemic of the 21st century. This pandemic, although efficient in transmission, is mild in virulence. This atypical mild pandemic season has raised concerns regarding the potential of this virus to acquire additional virulence markers either through further adaptation or possibly by immune pressure in the human host. Using the mouse model we generated, within a single round of infection with A/California/04/09/H1N1 (Ca/04), a virus lethal in mice--herein referred to as mouse-adapted Ca/04 (ma-Ca/04). Five amino acid substitutions were found in the genome of ma-Ca/04: 3 in HA (D131E, S186P and A198E), 1 in PA (E298K) and 1 in NP (D101G). Reverse genetics analyses of these mutations indicate that all five mutations from ma-Ca/04 contributed to the lethal phenotype; however, the D131E and S186P mutations--which are also found in the 1918 and seasonal H1N1 viruses-in HA alone were sufficient to confer virulence of Ca/04 in mice. HI assays against H1N1pdm demonstrate that the D131E and S186P mutations caused minor antigenic changes and, likely, affected receptor binding. The rapid selection of ma-Ca/04 in mice suggests that a virus containing this constellation of amino acids might have already been present in Ca/04, likely as minor quasispecies.


Asunto(s)
Variación Genética , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Subtipo H1N1 del Virus de la Influenza A/genética , Gripe Humana/epidemiología , Virulencia/genética , Animales , Células Cultivadas , Perros , Femenino , Hurones , Estudios de Asociación Genética , Variación Genética/fisiología , Glicoproteínas Hemaglutininas del Virus de la Influenza/química , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Humanos , Subtipo H1N1 del Virus de la Influenza A/inmunología , Subtipo H1N1 del Virus de la Influenza A/patogenicidad , Gripe Humana/genética , Gripe Humana/inmunología , Gripe Humana/virología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos DBA , Modelos Moleculares , Pandemias , Fenotipo , Conformación Proteica
7.
Proc Natl Acad Sci U S A ; 106(18): 7565-70, 2009 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-19380727

RESUMEN

Pandemic influenza requires interspecies transmission of an influenza virus with a novel hemagglutinin (HA) subtytpe that can adapt to its new host through either reassortment or point mutations and transmit by aerosolized respiratory droplets. Two previous pandemics of 1957 and 1968 resulted from the reassortment of low pathogenic avian viruses and human subtypes of that period; however, conditions leading to a pandemic virus are still poorly understood. Given the endemic situation of avian H9N2 influenza with human-like receptor specificity in Eurasia and its occasional transmission to humans and pigs, we wanted to determine whether an avian-human H9N2 reassortant could gain respiratory transmission in a mammalian animal model, the ferret. Here we show that following adaptation in the ferret, a reassortant virus carrying the surface proteins of an avian H9N2 in a human H3N2 backbone can transmit efficiently via respiratory droplets, creating a clinical infection similar to human influenza infections. Minimal changes at the protein level were found in this virus capable of respiratory droplet transmission. A reassortant virus expressing only the HA and neuraminidase (NA) of the ferret-adapted virus was able to account for the transmissibility, suggesting that currently circulating avian H9N2 viruses require little adaptation in mammals following acquisition of all human virus internal genes through reassortment. Hemagglutinin inhibition (HI) analysis showed changes in the antigenic profile of the virus, which carries profound implications for vaccine seed stock preparation against avian H9N2 influenza. This report illustrates that aerosolized respiratory transmission is not exclusive to current human H1, H2, and H3 influenza subtypes.


Asunto(s)
Glicoproteínas Hemaglutininas del Virus de la Influenza/metabolismo , Subtipo H9N2 del Virus de la Influenza A/patogenicidad , Gripe Aviar/transmisión , Gripe Humana/transmisión , Virus Reordenados/patogenicidad , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Aves , Modelos Animales de Enfermedad , Brotes de Enfermedades , Hurones/virología , Glicoproteínas Hemaglutininas del Virus de la Influenza/química , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Humanos , Subtipo H9N2 del Virus de la Influenza A/genética , Subtipo H9N2 del Virus de la Influenza A/metabolismo , Gripe Aviar/epidemiología , Gripe Aviar/virología , Gripe Humana/epidemiología , Gripe Humana/virología , Pulmón/virología , Datos de Secuencia Molecular , Mutación , Conformación Proteica , Virus Reordenados/genética
8.
J Virol ; 84(22): 11831-40, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20826691

RESUMEN

The events and mechanisms that lead to interspecies transmission of, and host adaptation to, influenza A virus are unknown; however, both surface and internal proteins have been implicated. Our previous report highlighted the role that Japanese quail play as an intermediate host, expanding the host range of a mallard H2N2 virus, A/mallard/Potsdam/178-4/83 (H2N2), through viral adaptation. This quail-adapted virus supported transmission in quail and increased its host range to replicate and be transmitted efficiently in chickens. Here we report that of the six amino acid changes in the quail-adapted virus, a single change in the hemagglutinin (HA) was crucial for transmission in quail, while the changes in the polymerase genes favored replication at lower temperatures than those for the wild-type mallard virus. Reverse genetic analysis indicated that all adaptive mutations were necessary for transmission in chickens, further implicating quail in extending this virus to terrestrial poultry. Adaptation of the quail-adapted virus in chickens resulted in the alteration of viral tropism from intestinal shedding to shedding and transmission via the respiratory tract. Sequence analysis indicated that this chicken-adapted virus maintained all quail-adaptive mutations, as well as an additional change in the HA and, most notably, a 27-amino-acid deletion in the stalk region of neuraminidase (NA), a genotypic marker of influenza virus adaptation to chickens. This stalk deletion was shown to be responsible for the change in virus tropism from the intestine to the respiratory tract.


Asunto(s)
Subtipo H2N2 del Virus de la Influenza A/enzimología , Subtipo H2N2 del Virus de la Influenza A/genética , Gripe Aviar/virología , Neuraminidasa/genética , Enfermedades de las Aves de Corral/virología , Sistema Respiratorio/virología , Eliminación de Secuencia , Proteínas Virales/genética , Replicación Viral , Animales , Línea Celular , Pollos , Coturnix , Subtipo H2N2 del Virus de la Influenza A/fisiología , Datos de Secuencia Molecular , Neuraminidasa/metabolismo , Codorniz , Proteínas Virales/metabolismo
9.
Virol J ; 8: 31, 2011 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-21255403

RESUMEN

Mass in ovo vaccination with live attenuated viruses is widely used in the poultry industry to protect against various infectious diseases. The worldwide outbreaks of low pathogenic and highly pathogenic avian influenza highlight the pressing need for the development of similar mass vaccination strategies against avian influenza viruses. We have previously shown that a genetically modified live attenuated avian influenza virus (LAIV) was amenable for in ovo vaccination and provided optimal protection against H5 HPAI viruses. However, in ovo vaccination against other subtypes resulted in poor hatchability and, therefore, seemed impractical. In this study, we modified the H7 and H9 hemagglutinin (HA) proteins by substituting the amino acids at the cleavage site for those found in the H6 HA subtype. We found that with this modification, a single dose in ovo vaccination of 18-day old eggs provided complete protection against homologous challenge with low pathogenic virus in ≥ 70% of chickens at 2 or 6 weeks post-hatching. Further, inoculation of 19-day old egg embryos with 106 EID50 of LAIVs improved hatchability to ≥ 90% (equivalent to unvaccinated controls) with similar levels of protection. Our findings indicate that the strategy of modifying the HA cleavage site combined with the LAIV backbone could be used for in ovo vaccination against avian influenza. Importantly, with protection conferred as early as 2 weeks post-hatching, with this strategy birds would be protected prior to or at the time of delivery to a farm or commercial operation.


Asunto(s)
Subtipo H7N2 del Virus de la Influenza A/patogenicidad , Subtipo H9N2 del Virus de la Influenza A/patogenicidad , Vacunas contra la Influenza/efectos adversos , Vacunas contra la Influenza/inmunología , Gripe Aviar/inmunología , Gripe Aviar/prevención & control , Vacunación/métodos , Animales , Embrión de Pollo , Pollos , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Subtipo H7N2 del Virus de la Influenza A/genética , Subtipo H7N2 del Virus de la Influenza A/inmunología , Subtipo H9N2 del Virus de la Influenza A/genética , Subtipo H9N2 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/administración & dosificación , Vacunas contra la Influenza/genética , Gripe Aviar/virología , Análisis de Supervivencia , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/efectos adversos , Vacunas Atenuadas/genética , Vacunas Atenuadas/inmunología
10.
Avian Dis ; 54(1 Suppl): 513-21, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20521687

RESUMEN

The influenza A/Mallard/Pennsylvania/10218/1984 (H5N2) virus is unable to replicate in 3-wk-old immunocompetent specific-pathogen-free chickens when a dose of 5 x 10(6) 50% egg infectious dose/ml is used. In contrast, this mallard virus shows limited replication in 3-wk-old chickens that had been previously infected at 2 days of age with, and recovered from, the immunosuppressive agent infectious bursal disease virus (IBDV; herein referred to as IBDV chickens). This limited replication in IBDV chickens allowed for the serial passage of the mallard influenza virus in chickens. After 22 passages (P22) in IBDV chickens, the resulting chicken-adapted influenza virus replicated in both immunocompetent and IBDV chickens more efficiently than the mallard influenza virus. Analysis of the outcomes of infection and the lesions caused by the two viruses at the microscopic level in a time-point study showed that the P22 virus is more virulent than the parental mallard virus in both immunocompetent and IBDV chickens. Our studies provide evidence that a previous history of IBDV infection in chickens may render them more susceptible to avian influenza virus (AIV) infections, allowing for the potential introduction of AIVs in an otherwise resistant population.


Asunto(s)
Pollos , Virus de la Enfermedad Infecciosa de la Bolsa/inmunología , Subtipo H5N2 del Virus de la Influenza A/fisiología , Gripe Aviar/virología , Replicación Viral/fisiología , Adaptación Fisiológica , Animales , Infecciones por Birnaviridae/inmunología , Infecciones por Birnaviridae/veterinaria , Inmunocompetencia , Subtipo H5N2 del Virus de la Influenza A/patogenicidad , Organismos Libres de Patógenos Específicos , Virulencia
11.
Regen Med ; 15(12): 2341-2349, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33480804

RESUMEN

Aim: This work aims to explore the biological role of negative pressure wound therapy (NPWT) in the treatment of diabetic ulcer. Materials & methods: Full-thickness skin defects were created in diabetic (db/db) and non diabetic (db/m) mice to create wound models. The mice were received NPWT or rapamycin injection. Mouse macrophage cells (Raw264.7) were treated with lipopolysaccharide to induce inflammatory response, and then received negative pressure treatment. We observed the wound healing of mice and examined gene and protein expression and CD68+ macrophage levels. Results: NPWT notably enhanced the wound closure ratio, and inhibited the LC3-II/LC3-I ratio and Beclin-1 expression in diabetes mellitus (DM) mice. NPWT decreased CD68+ macrophage levels in wound tissues of DM mice. The influence conferred by NPWT was abolished by rapamycin treatment. Negative pressure repressed the LC3-II/LC3-I ratio and the expression of Beclin-1, TNF-α, IL-6 and IL-1ß in the Raw264.7 cells. Conclusion: NPWT promotes wound healing by suppressing autophagy and macrophage inflammation in DM.


Asunto(s)
Diabetes Mellitus , Terapia de Presión Negativa para Heridas , Animales , Inflamación , Macrófagos , Ratones , Úlcera , Cicatrización de Heridas
12.
Virol J ; 6: 126, 2009 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-19682381

RESUMEN

BACKGROUND: Avian influenza viruses of the H7 subtype have caused multiple outbreaks in domestic poultry and represent a significant threat to public health due to their propensity to occasionally transmit directly from birds to humans. In order to better understand the cross species transmission potential of H7 viruses in nature, we performed biological and molecular characterizations of an H7N3 virus isolated from mallards in Canada in 2001. RESULTS: Sequence analysis that the HA gene of the mallard H7N3 virus shares 97% identity with the highly pathogenic avian influenza (HPAI) H7N3 virus isolated from a human case in British Columbia, Canada in 2004. The mallard H7N3 virus was able to replicate in quail and chickens, and transmitted efficiently in quail but not in chickens. Interestingly, although this virus showed preferential binding to analogs of avian-like receptors with sialic acid (SA) linked to galactose in an alpha2-3 linkage (SAalpha2-3Gal), it replicated to high titers in cultures of primary human airway epithelial (HAE) cells, comparable to an avian H9N2 influenza virus with human-like alpha2-6 linkage receptors (SAalpha2-6Gal). In addition, the virus replicated in mice and ferrets without prior adaptation and was able to transmit partially among ferrets. CONCLUSION: Our findings highlight the importance and need for systematic in vitro and in vivo analysis of avian influenza viruses isolated from the natural reservoir in order to define their zoonotic potential.


Asunto(s)
Modelos Animales de Enfermedad , Hurones , Virus de la Influenza A/fisiología , Gripe Aviar/transmisión , Gripe Humana/transmisión , Receptores Virales/metabolismo , Animales , Aves , Canadá , Línea Celular , Femenino , Humanos , Virus de la Influenza A/genética , Virus de la Influenza A/aislamiento & purificación , Gripe Aviar/metabolismo , Gripe Aviar/virología , Gripe Humana/metabolismo , Gripe Humana/virología , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Unión Proteica , Receptores Virales/química , Especificidad de la Especie , Acoplamiento Viral , Replicación Viral
13.
Virus Res ; 122(1-2): 61-8, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16891025

RESUMEN

Infectious pancreatic necrosis virus (IPNV), a member of the Birnaviridae family, encodes a nonstructural VP5 protein from a small open reading frame (ORF), which overlaps with a major ORF encoding pVP2, VP4 and VP3 proteins. In majority of the Sp strains of IPNV sequenced to date, VP5 gene codes for a 15-kDa protein. However, we have shown that in highly virulent strains, there is a premature in-frame stop codon (UGA) at nucleotide (nt) position 427, (preceding the 15-kDa stop codon at nt position 511) which could encode a 12-kDa protein. Using reverse genetics, we recovered recombinant rNVI15, rNVI15-15K and rNVI15-DeltaVP5 viruses (which could encode 12 or 15-kDa VP5 or lack the expression of VP5, respectively) and demonstrated that VP5 is dispensable for viral replication in vivo but is not involved in virulence (Santi, N., Song, H., Vakharia, V. N., Evensen, Ø., 2005a. Infectious pancreatic necrosis virus VP5 is dispensable for virulence and persistence. J. Virol. 79, 9206-9216). Here, we utilized these viruses to investigate the gene expression of VP5 in vitro. Our results indicate that a 15-kDa VP5 is produced in rNVI15-infected cells, albeit at lower levels than in rNVI15-15K-infected cells, suggesting that the opal stop codon at nt 427 is suppressed. Furthermore, to examine translational suppression of the opal stop codon in VP5 gene, we constructed plasmids containing VP5-specific sequence and employed a yeast-based bicistronic dual-luciferase reporter system (Harger, J.W., Dinman, J.D., 2003. An in vivo dual-luciferase assay system for studying translational recoding in the yeast Saccharomyces cerevisiae. RNA 9, 1019-1024). Our results demonstrate that the VP5 sequence (with or without a stop codon) yielded approximately 13% termination suppression and the efficiency is directly related to the base immediately 3' of the termination codon, C>A>U>G.


Asunto(s)
Codón sin Sentido/genética , Expresión Génica , Virus de la Necrosis Pancreática Infecciosa/genética , Supresión Genética/fisiología , Proteínas no Estructurales Virales/biosíntesis , Animales , Western Blotting , Línea Celular , Genes Reporteros , Luciferasas/biosíntesis , Luciferasas/genética , Biosíntesis de Proteínas , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Salmón , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/fisiología
14.
J Plast Surg Hand Surg ; 48(6): 402-6, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24693868

RESUMEN

Treatment of cicatricial lower eyelid ectropion is often difficult and requires surgical intervention. Numerous techniques have been developed over the years to treat the cicatricial lower eyelid ectropion. This article describes an effective surgical technique using the free transplantation of autogenous palmaris longus tendon in the repair of cicatricial lower eyelid ectropion. The operation was performed with the patient under local anaesthesia. After the contracture of the lower eyelid skin or conjunctiva had been thoroughly released, a palmaris longus tendon graft was obtained through small transverse incisions along the non-dominant forearm. The graft is suspended between the medial canthal ligament and lateral orbital rim with proper tension. Then a local skin flap was transferred to cover the wound at the lower eyelid. In this study, 15 patients were treated using autogenous palmaris longus tendon between December 2008 and October 2012. At the 9-18 months of follow-up assessment, there were no major complications reported with any of the cases, and all have achieved good function and satisfactory appearance of the lower eyelid. However, five patients (33%) still had lagophthalmos, but it was in remission. In conclusion, free transplantation of autogenous palmaris longus tendon in the repair of cicatricial ectropion of the lower eyelid is an effective procedure for cicatricial ectropion, the functional and cosmetic results were satisfactory, the recurrence rate is low. Especially for the moderate and severe ectropion this method is a good choice.


Asunto(s)
Ectropión/cirugía , Tendones/trasplante , Adulto , Autoinjertos , Cicatriz/complicaciones , Femenino , Humanos , Masculino , Persona de Mediana Edad , Técnicas de Sutura , Adulto Joven
15.
Zhonghua Zheng Xing Wai Ke Za Zhi ; 28(6): 444-8, 2012 Nov.
Artículo en Zh | MEDLINE | ID: mdl-23520782

RESUMEN

OBJECTIVE: To determine the role of toll like receptor-4 signal pathways activation in ischemia-reperfusion injury of island skin flap. METHODS: A totol of 50 adult male SD rats were randomized into 3 groups: sham-operated group (n=10), ischemia/reperfusion group (n=20) and TLR4 inhibitor-eritoran tetrasodium (E5564)-treated group (n=20). The inguinal island skin flaps models were set up. A bolus of E5564 (5 mg/kg) was infused intravenously 60 min before reper fusionm. TLR4 binding activity in flap tissue was analyzed at 1, 2, 4 and 6 h of reperfusion by immunohistochemical technique and flaps were assessed histologically at 6 h of reperfusion. The viability of flaps was assessed 7 days postoperatively. RESULTS: Exprerssion TLR4 in skin flap tissue was significantly increased in I/R group, compared with E5564-treated group. Immunohistochemical exam showed TLR4 mainly expressed in skin flap vessel wall and PMN membrane. Marked neutrophil infiltration and edema was observed in I/R group, while less neutrophil infiltration was observed in E5564-treated group. In the E5564-treated group, the survival of flaps was (80.31 +/- 11.63)%, which was significantly greater than that in the I/R group (51.70 +/- 7.62)% (P < 0.01). CONCLUSION: After ischemia-reperfusion injury in rats, the expression of TLR4 increased in the skin flap tissue with excessive neutrophil infiltration. Administration of E5564 can significantly improve flap survival by regulating the early activation of TLR4 and suppressing neutrophil infiltration within the flap.


Asunto(s)
Isquemia/metabolismo , Daño por Reperfusión/metabolismo , Transducción de Señal , Colgajos Quirúrgicos/irrigación sanguínea , Receptor Toll-Like 4/metabolismo , Animales , Ingle , Lípido A/análogos & derivados , Lípido A/farmacología , Masculino , Distribución Aleatoria , Ratas , Receptor Toll-Like 4/antagonistas & inhibidores
16.
Clin Vaccine Immunol ; 18(3): 406-13, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21177911

RESUMEN

Lyme disease (LD) is a tick-borne infection caused by the bacterial pathogen Borrelia burgdorferi. Current diagnostic tests mostly use borrelial lysates or select antigens to detect serum antibodies against B. burgdorferi. These immunoassays are not entirely effective, especially for detection of early infection. We have recently characterized an in vivo-induced antigen, BBK07, as a serodiagnostic marker for LD. We now report that in a line blot assay, recombinant BBK07 protein-based detection is 90% sensitive and nearly 100% specific against B. burgdorferi infection in humans. Using an overlapping peptide library of 23 peptides encompassing full-length BBK07, we identified the immunodominant epitopes of BBK07 during human infection. We show that a select combination of amino-terminal peptides significantly enhanced BBK07-based diagnostic accuracy compared to that with the full-length protein. Although in enzyme-linked immunosorbent assay (ELISA) studies BBK07 peptides had overall lower sensitivity than established serodiagnostic peptides, such as the VlsE peptide C6 and OspC peptide pepC10, for the detection of early human LD, a subset of serum samples that failed to recognize either VlsE or OspC peptides were preferentially reactive to BBK07 peptides. These results highlight the fact that BBK07 peptides could be useful to complement the efficacy of VlsE and OspC peptide-based serodiagnostic assays. Finally, using a panel of canine sera, we show that BBK07 peptide is also effective for LD diagnosis in infected dogs. Together, our data show that peptides from the B. burgdorferi surface protein BBK07 are highly specific and sensitive serodiagnostic markers, and we suggest their future use in LD diagnostic assays.


Asunto(s)
Anticuerpos Antibacterianos/sangre , Antígenos Bacterianos , Borrelia burgdorferi/química , Técnicas de Laboratorio Clínico/métodos , Epítopos Inmunodominantes , Enfermedad de Lyme/diagnóstico , Enfermedad de Lyme/veterinaria , Animales , Biomarcadores/sangre , Borrelia burgdorferi/inmunología , Perros , Humanos , Sensibilidad y Especificidad , Pruebas Serológicas/métodos
17.
Clin Vaccine Immunol ; 17(9): 1363-70, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20668143

RESUMEN

Highly pathogenic avian H5N1 influenza viruses are endemic in poultry in Asia and pose a pandemic threat to humans. Since the deployment of vaccines against a pandemic strain may take several months, adequate antiviral alternatives are needed to minimize the effects and the spread of the disease. Passive immunotherapy is regarded as a viable alternative. Here, we show the development of an IgA monoclonal antibody (DPJY01 MAb) specific to H5 hemagglutinin. The DPJY01 MAb showed a broad hemagglutination inhibition (HI) profile against Asian H5N1 viruses of clades 0, 1.0, 2.1, 2.2, and 2.3 and also against H5 wild bird influenza viruses of the North American and Eurasian lineages. DPJY01 MAb displayed also high neutralization activity in vitro and in vivo. In mice, DPJY01 MAb provided protection via a single dose administered intranasally before or after inoculation with a sublethal dose of H5N1 viruses of clades 1.0 and 2.2. Pretreatment with 50 mg of DPJY01 MAb kg of body weight at either 24, 48, or 72 h before highly pathogenic H5N1 virus (A/Vietnam/1203/2004 [H5N1]) inoculation resulted in complete protection. Treatment with 50 mg/kg at either at 24, 48, or 72 h after H5N1 inoculation provided 100%, 80%, and 60% protection, respectively. These studies highlight the potential use of DPJY01 MAb as an intranasal antiviral treatment for H5N1 influenza virus infections.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Inmunoglobulina A/administración & dosificación , Inmunoterapia/métodos , Subtipo H5N1 del Virus de la Influenza A/inmunología , Infecciones por Orthomyxoviridae/prevención & control , Infecciones por Orthomyxoviridae/terapia , Administración Intranasal , Animales , Anticuerpos Monoclonales/aislamiento & purificación , Femenino , Pruebas de Inhibición de Hemaglutinación , Inmunoglobulina A/aislamiento & purificación , Ratones , Ratones Endogámicos BALB C , Pruebas de Neutralización , Análisis de Supervivencia , Factores de Tiempo
18.
J Gen Virol ; 89(Pt 11): 2682-2690, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18931063

RESUMEN

The unprecedented emergence in Asia of multiple avian influenza virus (AIV) subtypes with a broad host range poses a major challenge in the design of vaccination strategies that are both effective and available in a timely manner. The present study focused on the protective effects of a genetically modified AIV as a source for the preparation of vaccines for epidemic and pandemic influenza. It has previously been demonstrated that a live attenuated AIV based on the internal backbone of influenza A/Guinea fowl/Hong Kong/WF10/99 (H9N2), called WF10att, is effective at protecting poultry species against low- and high-pathogenicity influenza strains. More importantly, this live attenuated virus provided effective protection when administered in ovo. In order to characterize the WF10att backbone further for use in epidemic and pandemic influenza vaccines, this study evaluated its protective effects in mice. Intranasal inoculation of modified attenuated viruses in mice provided adequate protective immunity against homologous lethal challenges with both the wild-type influenza A/WSN/33 (H1N1) and A/Vietnam/1203/04 (H5N1) viruses. Adequate heterotypic immunity was also observed in mice vaccinated with modified attenuated viruses carrying H7N2 surface proteins. The results presented in this report suggest that the internal genes of a genetically modified AIV confer similar protection in a mouse model and thus could be used as a master donor strain for the generation of live attenuated vaccines for epidemic and pandemic influenza.


Asunto(s)
Brotes de Enfermedades/prevención & control , Brotes de Enfermedades/veterinaria , Virus de la Influenza A/inmunología , Vacunas contra la Influenza/uso terapéutico , Gripe Aviar/inmunología , Gripe Humana/epidemiología , Vacunas Atenuadas/uso terapéutico , Adulto , Animales , Aves , Línea Celular , Niño , Perros , Humanos , Gripe Aviar/epidemiología , Gripe Humana/inmunología , Gripe Humana/mortalidad , Gripe Humana/virología , Riñón/embriología , Pruebas de Neutralización , Vacunas Sintéticas/inmunología , Carga Viral , Proteínas Virales/genética , Proteínas Virales/inmunología
19.
PLoS One ; 3(8): e2923, 2008 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-18698430

RESUMEN

H9N2 avian influenza A viruses are endemic in poultry of many Eurasian countries and have caused repeated human infections in Asia since 1998. To evaluate the potential threat of H9N2 viruses to humans, we investigated the replication and transmission efficiency of H9N2 viruses in the ferret model. Five wild-type (WT) H9N2 viruses, isolated from different avian species from 1988 through 2003, were tested in vivo and found to replicate in ferrets. However these viruses achieved mild peak viral titers in nasal washes when compared to those observed with a human H3N2 virus. Two of these H9N2 viruses transmitted to direct contact ferrets, however no aerosol transmission was detected in the virus displaying the most efficient direct contact transmission. A leucine (Leu) residue at amino acid position 226 in the hemagglutinin (HA) receptor-binding site (RBS), responsible for human virus-like receptor specificity, was found to be important for the transmission of the H9N2 viruses in ferrets. In addition, an H9N2 avian-human reassortant virus, which contains the surface glycoprotein genes from an H9N2 virus and the six internal genes of a human H3N2 virus, showed enhanced replication and efficient transmission to direct contacts. Although no aerosol transmission was observed, the virus replicated in multiple respiratory tissues and induced clinical signs similar to those observed with the parental human H3N2 virus. Our results suggest that the establishment and prevalence of H9N2 viruses in poultry pose a significant threat for humans.


Asunto(s)
Hurones/virología , Subtipo H9N2 del Virus de la Influenza A/fisiología , Gripe Humana/epidemiología , Replicación Viral , Animales , Aves , Diagnóstico Diferencial , Modelos Animales de Enfermedad , Brotes de Enfermedades , Humanos , Subtipo H9N2 del Virus de la Influenza A/aislamiento & purificación , Gripe Aviar/epidemiología , Gripe Humana/diagnóstico , Modelos Biológicos , Pérdida de Peso
20.
Virology ; 378(2): 363-70, 2008 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-18632129

RESUMEN

Avian influenza (AI) viruses have been sporadically isolated in South America. The most recent reports are from an outbreak in commercial poultry in Chile in 2002 and its putative ancestor from a wild bird in Bolivia in 2001. Extensive surveillance in wild birds was carried out in Argentina during 2006-2007. Using RRT-PCR, 12 AI positive detections were made from cloacal swabs. One of those positive samples yielded an AI virus isolated from a wild kelp gull (Larus dominicanus) captured in the South Atlantic coastline of Argentina. Further characterization by nucleotide sequencing reveals that it belongs to the H13N9 subtype. Phylogenetic analysis of the 8 viral genes suggests that the 6 internal genes are related to the isolates from Chile and Bolivia. The analysis also indicates that a cluster of phylogenetically related AI viruses from South America may have evolved independently, with minimal gene exchange, from influenza viruses in other latitudes. The data produced from our investigations are valuable contributions to the study of AI viruses in South America.


Asunto(s)
Aves/virología , Virus de la Influenza A/clasificación , Virus de la Influenza A/aislamiento & purificación , Gripe Aviar/virología , Animales , Argentina , Cloaca/virología , Evolución Molecular , Virus de la Influenza A/genética , Datos de Secuencia Molecular , Filogenia , ARN Viral/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Análisis de Secuencia de ADN , Homología de Secuencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA