Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Biomolecules ; 13(10)2023 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-37892192

RESUMEN

The cytokine-inducible SH2 domain-containing (CISH) protein was the first member of the suppressor of cytokine signaling (SOCS) family of negative feedback regulators discovered, being identified in vitro as an inducible inhibitor of erythropoietin (EPO) signaling. However, understanding of the physiological role played by CISH in erythropoiesis has remained limited. To directly assess the function of CISH in this context, mice deficient in CISH were characterized with respect to developmental, steady-state, and EPO-induced erythropoiesis. CISH was strongly expressed in the fetal liver, but CISH knockout (KO) mice showed only minor disruption of primitive erythropoiesis. However, adults exhibited mild macrocytic anemia coincident with subtle perturbation particularly of bone marrow erythropoiesis, with EPO-induced erythropoiesis blunted in the bone marrow of KO mice but enhanced in the spleen. Cish was expressed basally in the bone marrow with induction following EPO stimulation in bone marrow and spleen. Overall, this study indicates that CISH participates in the control of both basal and EPO-induced erythropoiesis in vivo.


Asunto(s)
Eritropoyesis , Proteínas Supresoras de la Señalización de Citocinas , Animales , Ratones , Anemia/genética , Citocinas , Eritropoyesis/fisiología , Transducción de Señal/fisiología , Dominios Homologos src , Proteínas Supresoras de la Señalización de Citocinas/metabolismo
2.
PLoS One ; 16(11): e0259829, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34793507

RESUMEN

The current Mycobacterium bovis BCG vaccine provides inconsistent protection against pulmonary infection with Mycobacterium tuberculosis. Immunity induced by subcutaneous immunization with BCG wanes and does not promote early recruitment of T cell to the lungs after M. tuberculosis infection. Delivery of Tuberculosis (TB) vaccines to the lungs may increase and prolong immunity at the primary site of M. tuberculosis infection. Pulmonary immunization with recombinant influenza A viruses (rIAVs) expressing an immune-dominant M. tuberculosis CD4+ T cell epitope (PR8-p25 and X31-p25) stimulates protective immunity against lung TB infection. Here, we investigated the potential use of rIAVs to improve the efficacy of BCG using simultaneous immunization (SIM) and prime-boost strategies. SIM with parenteral BCG and intranasal PR8-p25 resulted in equivalent protection to BCG alone against early, acute and chronic M. tuberculosis infection. Boosting BCG with rIAVs increased the frequency of IFN-γ-secreting specific T cells (p<0.001) and polyfunctional CD4+ T cells (p<0.05) in the lungs compared to the BCG alone, however, this did not result in a significant increase in protection against M. tuberculosis compared to BCG alone. Therefore, sequential pulmonary immunization with these rIAVs after BCG increased M. tuberculosis-specific memory T cell responses in the lung, but not protection against M. tuberculosis infection.


Asunto(s)
Vacuna BCG/inmunología , Virus de la Influenza A/inmunología , Mycobacterium tuberculosis/inmunología , Linfocitos T/inmunología , Tuberculosis Pulmonar/prevención & control , Animales , Epítopos , Epítopos de Linfocito T/genética , Epítopos de Linfocito T/inmunología , Femenino , Inmunización Secundaria , Inmunogenicidad Vacunal , Pulmón/inmunología , Células T de Memoria/inmunología , Ratones , Ratones Endogámicos C57BL , Tuberculosis Pulmonar/inmunología , Vacunas Sintéticas/inmunología
3.
Front Mol Biosci ; 8: 703868, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34527702

RESUMEN

Remodelling of the extracellular matrix (ECM) by ECM metalloproteinases is increasingly being associated with regulation of immune cell function. ECM metalloproteinases, including Matrix Metalloproteinases (MMPs), A Disintegrin and Metalloproteinases (ADAMs) and ADAMs with Thombospondin-1 motifs (ADAMTS) play a vital role in pathogen defence and have been shown to influence migration of immune cells. This review provides a current summary of the role of ECM enzymes in immune cell migration and function and discusses opportunities and limitations for development of diagnostic and therapeutic strategies targeting metalloproteinase expression and activity in the context of infectious disease.

4.
Front Mol Biosci ; 8: 703456, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34291090

RESUMEN

Each year, hundreds of thousands of individuals succumb to influenza virus infection and its associated complications. Several preventative and therapeutic options may be applied in order to preserve life. These traditional approaches include administration of seasonal influenza vaccines, pharmacological interventions in the form of antiviral drug therapy and supportive clinical approaches including mechanical ventilation and extracorporeal membrane oxygenation. While these measures have shown varying degrees of success, antiviral therapies and vaccination are constrained due to ongoing antigenic drift. Moreover, clinical approaches can also be associated with complications and drawbacks. These factors have led to the exploration and development of more sophisticated and nuanced therapeutic approaches involving host proteins. Advances in immunotherapy in the cancer field or administration of steroids following virus infection have highlighted the therapeutic potential of targeting host immune responses. We have now reached a point where we can consider the contribution of other "non-traditional" host components such as the extracellular matrix in immunity. Herein, we will review current, established therapeutic interventions and consider novel therapeutic approaches involving the extracellular matrix.

5.
Viruses ; 13(4)2021 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-33917411

RESUMEN

Vaccines and therapeutics targeting viral surface glycoproteins are a major component of disease prevention for respiratory viral diseases. Over the years, vaccines have proven to be the most successful intervention for preventing disease. Technological advances in vaccine platforms that focus on viral surface glycoproteins have provided solutions for current and emerging pathogens like SARS-CoV-2, and our understanding of the structural basis for antibody neutralization is guiding the selection of other vaccine targets for respiratory viruses like RSV. This review discusses the role of viral surface glycoproteins in disease intervention approaches.


Asunto(s)
Anticuerpos Antivirales/inmunología , COVID-19/inmunología , Glicoproteínas de Membrana/inmunología , SARS-CoV-2/inmunología , Animales , Anticuerpos Neutralizantes , Vacunas contra la COVID-19 , Humanos , Glicoproteínas de Membrana/efectos de los fármacos , Virus Sincitiales Respiratorios , Estaciones del Año , Vacunas Atenuadas/inmunología , Vacunas Virales
6.
Expert Rev Respir Med ; 14(11): 1141-1147, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32762572

RESUMEN

INTRODUCTION: The innate immune response is the first line of defense and consists of physical, chemical and cellular defenses. The adaptive immune response is the second line of defense and is pathogen-specific. Innate immunity occurs immediately while adaptive immunity develops upon pathogen exposure, and is long-lasting, highly specific, and sustained by memory T cells. Respiratory virus infection typically induces effective immunity but over-exuberant responses are associated with pathophysiology. Cytokines expressed in response to viral infection can enhance biological responses, activate, and trigger signaling pathways leading to adaptive immunity Vaccines induce immunity, specifically B and T cell responses. Vaccination is generally efficacious, but for many viruses, our understanding of vaccination strategies and immunity is incomplete or in its infancy. Studies that examine innate and adaptive immune responses to respiratory virus infection will aid vaccine development and may reduce the burden of respiratory viral disease. AREAS COVERED: A literature search was performed using PubMed. The search covered: innate, adaptive, respiratory virus, vaccine development, B cell, and T cell. EXPERT OPINION: Immunity rests on two pillars, i.e. the innate and adaptive immune system, which function together on different tasks to maintain homeostasis. a better understanding of immunity is necessary for disease prevention and intervention.


Asunto(s)
Inmunidad Adaptativa/fisiología , Inmunidad Innata/fisiología , Infecciones del Sistema Respiratorio/inmunología , Virosis/inmunología , Animales , Humanos , Infecciones del Sistema Respiratorio/diagnóstico , Infecciones del Sistema Respiratorio/patología , Infecciones del Sistema Respiratorio/terapia , Vacunación/métodos , Vacunación/tendencias , Vacunas Virales/uso terapéutico , Virosis/diagnóstico , Virosis/epidemiología , Virosis/terapia , Virus/inmunología
8.
Cell Rep ; 29(11): 3539-3550.e4, 2019 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-31825834

RESUMEN

Interferons (IFN) are pleiotropic cytokines essential for defense against infection, but the identity and tissue distribution of IFN-responsive cells in vivo are poorly defined. In this study, we generate a mouse strain capable of reporting IFN-signaling activated by all three types of IFNs and investigate the spatio-temporal dynamics and identity of IFN-responding cells following IFN injection and influenza virus infection. Despite ubiquitous expression of IFN receptors, cellular responses to IFNs are highly heterogenous in vivo and are determined by anatomical site, cell type, cellular preference to individual IFNs, and activation status. Unexpectedly, type I and II pneumocytes, the primary target of influenza infection, exhibit striking differences in the strength and temporal dynamics of IFN signaling associated with differential susceptibility to the viral infection. Our findings suggest that time- and cell-type-dependent integration of distinct IFN signals govern the specificity and magnitude of IFN responses in vivo.


Asunto(s)
Interferones/metabolismo , Infecciones por Orthomyxoviridae/metabolismo , Transducción de Señal , Células Epiteliales Alveolares/metabolismo , Animales , Células Cultivadas , Femenino , Proteínas de Unión al GTP/genética , Proteínas de Unión al GTP/metabolismo , Células Madre Hematopoyéticas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL
10.
PLoS Biol ; 17(11): e3000558, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31693658

RESUMEN

[This corrects the article DOI: 10.1371/journal.pbio.1002580.].

11.
J Immunol ; 202(12): 3370-3380, 2019 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-31092636

RESUMEN

The importance of antiviral CD8+ T cell recognition of alternative reading frame (ARF)-derived peptides is uncertain. In this study, we describe an epitope (NS1-ARF21-8) present in a predicted 14-residue peptide encoded by the +1 register of NS1 mRNA in the influenza A virus (IAV). NS1-ARF21-8 elicits a robust, highly functional CD8+ T cell response in IAV-infected BALB/c mice. NS1-ARF21-8 is presented from unspliced NS mRNA, likely from downstream initiation on a Met residue that comprises the P1 position of NS1-ARF21-8 Derived from a 14-residue peptide with no apparent biological function and negligible impacts on IAV infection, infectivity, and pathogenicity, NS1-ARF21-8 provides a clear demonstration of how immunosurveillance exploits natural errors in protein translation to provide antiviral immunity. We further show that IAV infection enhances a model cellular ARF translation, which potentially has important implications for virus-induced autoimmunity.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Epítopos de Linfocito T/metabolismo , Virus de la Influenza A/fisiología , Gripe Humana/inmunología , Infecciones por Orthomyxoviridae/inmunología , Proteínas no Estructurales Virales/metabolismo , Empalme Alternativo , Animales , Modelos Animales de Enfermedad , Epítopos de Linfocito T/genética , Epítopos de Linfocito T/inmunología , Células HEK293 , Interacciones Huésped-Patógeno , Humanos , Vigilancia Inmunológica , Ratones , Ratones Endogámicos BALB C , Sistemas de Lectura Abierta/genética , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/inmunología
12.
Front Immunol ; 10: 339, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30899256

RESUMEN

T-lymphocytes are critical for protection against respiratory infections, such as Mycobacterium tuberculosis and influenza virus, with chemokine receptors playing an important role in directing these cells to the lungs. CXCR6 is expressed by activated T-lymphocytes and its ligand, CXCL16, is constitutively expressed by the bronchial epithelia, suggesting a role in T-lymphocyte recruitment and retention. However, it is unknown whether CXCR6 is required in responses to pulmonary infection, particularly on CD4+ T-lymphocytes. Analysis of CXCR6-reporter mice revealed that in naïve mice, lung leukocyte expression of CXCR6 was largely restricted to a small population of T-lymphocytes, but this population was highly upregulated after either infection. Nevertheless, pulmonary infection of CXCR6-deficient mice with M. tuberculosis or recombinant influenza A virus expressing P25 peptide (rIAV-P25), an I-Ab-restricted epitope from the immunodominant mycobacterial antigen, Ag85B, demonstrated that the receptor was redundant for recruitment of T-lymphocytes to the lungs. Interestingly, CXCR6-deficiency resulted in reduced bacterial burden in the lungs 6 weeks after M. tuberculosis infection, and reduced weight loss after rIAV-P25 infection compared to wild type controls. This was paradoxically associated with a decrease in Th1-cytokine responses in the lung parenchyma. Adoptive transfer of P25-specific CXCR6-deficient T-lymphocytes into WT mice revealed that this functional change in Th1-cytokine production was not due to a T-lymphocyte intrinsic mechanism. Moreover, there was no reduction in the number or function of CD4+ and CD8+ tissue resident memory cells in the lungs of CXCR6-deficient mice. Although CXCR6 was not required for T-lymphocyte recruitment or retention in the lungs, CXCR6 influenced the kinetics of the inflammatory response so that deficiency led to increased host control of M. tuberculosis and influenza virus.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Virus de la Influenza A/inmunología , Mycobacterium tuberculosis/inmunología , Infecciones por Orthomyxoviridae/inmunología , Receptores CXCR6/inmunología , Tuberculosis Pulmonar/inmunología , Traslado Adoptivo/métodos , Animales , Antígenos Bacterianos/inmunología , Citocinas/inmunología , Inflamación/inmunología , Inflamación/virología , Pulmón/inmunología , Pulmón/virología , Ratones , Ratones Endogámicos C57BL , Infecciones por Orthomyxoviridae/virología , Células TH1/inmunología
13.
Sci Rep ; 8(1): 15468, 2018 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-30341336

RESUMEN

Current prophylactic and therapeutic strategies targeting human influenza viruses include vaccines and antivirals. Given variable rates of vaccine efficacy and antiviral resistance, alternative strategies are urgently required to improve disease outcomes. Here we describe the use of HiSeq deep sequencing to analyze host gene expression in primary human alveolar epithelial type II cells infected with highly pathogenic avian influenza H5N1 virus. At 24 hours post-infection, 623 host genes were significantly upregulated, including the cell adhesion molecule CEACAM1. H5N1 virus infection stimulated significantly higher CEACAM1 protein expression when compared to influenza A PR8 (H1N1) virus, suggesting a key role for CEACAM1 in influenza virus pathogenicity. Furthermore, silencing of endogenous CEACAM1 resulted in reduced levels of proinflammatory cytokine/chemokine production, as well as reduced levels of virus replication following H5N1 infection. Our study provides evidence for the involvement of CEACAM1 in a clinically relevant model of H5N1 infection and may assist in the development of host-oriented antiviral strategies.


Asunto(s)
Células Epiteliales Alveolares/virología , Antígenos CD/metabolismo , Moléculas de Adhesión Celular/metabolismo , Interacciones Huésped-Patógeno , Subtipo H5N1 del Virus de la Influenza A/crecimiento & desarrollo , Células Cultivadas , Perfilación de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Subtipo H1N1 del Virus de la Influenza A/crecimiento & desarrollo
14.
Mucosal Immunol ; 11(6): 1743-1752, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30115996

RESUMEN

The lung is the primary site of infection with the major human pathogen, Mycobacterium tuberculosis. Effective vaccines against M. tuberculosis must stimulate memory T cells to provide early protection in the lung. Recently, tissue-resident memory T cells (TRM) were found to be phenotypically and transcriptional distinct from circulating memory T cells. Here, we identified M. tuberculosis-specific CD4+ T cells induced by recombinant influenza A viruses (rIAV) vaccines expressing M. tuberculosis peptides that persisted in the lung parenchyma with the phenotypic and transcriptional characteristics of TRMs. To determine if these rIAV-induced CD4+ TRM were protective independent of circulating memory T cells, mice previously immunized with the rIAV vaccine were treated with the sphingosine-1-phosphate receptor modulator, FTY720, prior to and during the first 17 days of M. tuberculosis challenge. This markedly reduced circulating T cells, but had no effect on the frequency of M. tuberculosis-specific CD4+ TRMs in the lung parenchyma or their cytokine response to infection. Importantly, mice immunized with the rIAV vaccine were protected against M. tuberculosis infection even when circulating T cells were profoundly depleted by the treatment. Therefore, pulmonary immunization with the rIAV vaccine stimulates lung-resident CD4+ memory T cells that are associated with early protection against tuberculosis infection.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Virus de la Influenza A/fisiología , Vacunas contra la Influenza/inmunología , Gripe Humana/inmunología , Pulmón/inmunología , Infecciones por Orthomyxoviridae/inmunología , Animales , Antígenos Bacterianos/metabolismo , Células Cultivadas , Femenino , Clorhidrato de Fingolimod/administración & dosificación , Humanos , Inmunización , Memoria Inmunológica , Pulmón/virología , Ratones , Ratones Endogámicos C57BL , Péptidos/metabolismo , Vacunas Sintéticas
15.
Sci Rep ; 8(1): 11518, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-30068964

RESUMEN

Low pathogenicity avian influenza viruses (LPAIVs) are generally asymptomatic in their natural avian hosts. LPAIVs can evolve into highly pathogenic forms, which can affect avian and human populations with devastating consequences. The switch to highly pathogenic avian influenza virus (HPAIV) from LPAIV precursors requires the acquisition of multiple basic amino acids in the haemagglutinin cleavage site (HACS) motif. Through reverse genetics of an H5N1 HPAIV, and experimental infection of chickens, we determined that viruses containing five or more basic amino acids in the HACS motif were preferentially selected over those with three to four basic amino acids, leading to rapid replacement with virus types containing extended HACS motifs. Conversely, viruses harbouring low pathogenicity motifs containing two basic amino acids did not readily evolve to extended forms, suggesting that a single insertion of a basic amino acid into the cleavage site motif of low-pathogenic viruses may lead to escalating selection for extended motifs. Our results may explain why mid-length forms are rarely detected in nature. The stability of the short motif suggests that pathogenicity switching may require specific conditions of intense selection pressure (such as with high host density) to boost selection of the initial mid-length HACS forms.


Asunto(s)
Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Glicoproteínas Hemaglutininas del Virus de la Influenza/metabolismo , Subtipo H5N1 del Virus de la Influenza A/patogenicidad , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Animales , Pollos , Subtipo H5N1 del Virus de la Influenza A/genética , Subtipo H5N1 del Virus de la Influenza A/aislamiento & purificación , Gripe Aviar/virología , Enfermedades de las Aves de Corral , Proteolisis , Genética Inversa , Selección Genética
16.
Virus Res ; 235: 115-120, 2017 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-28392443

RESUMEN

Influenza A vaccine efficacy in the elderly is generally poor and so identification of novel molecular adjuvants to improve immunogenicity is important to reduce the overall burden of disease. Short non-coding RNAs, known as microRNAs (miRNAs) are known to regulate gene expression and have the potential to influence immune responses. One such miRNA, miR-155, has been shown to modulate T and B cell development and function. We incorporated miR-155 into the influenza A virus (IAV) genome creating a self-adjuvanting 'live vaccine' with the ability to modify immunogenicity. Infection of mice with a recombinant influenza virus encoding miR-155 in the NS gene segment altered epitope-specific expansion of influenza-specific CD8+ T cells and induced significantly higher levels of neutralising antibody.


Asunto(s)
Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Linfocitos T CD8-positivos/inmunología , Genoma Viral , Virus de la Influenza A/genética , Virus de la Influenza A/inmunología , MicroARNs/genética , Animales , Ratones , Proteínas no Estructurales Virales/genética
17.
PLoS Biol ; 14(11): e1002580, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27855162

RESUMEN

The extracellular matrix (ECM) provides physical scaffolding for cellular constituents and initiates biochemical and biomechanical cues that are required for physiological activity of living tissues. The ECM enzyme ADAMTS5, a member of the ADAMTS (A Disintegrin-like and Metalloproteinase with Thrombospondin-1 motifs) protein family, cleaves large proteoglycans such as aggrecan, leading to the destruction of cartilage and osteoarthritis. However, its contribution to viral pathogenesis and immunity is currently undefined. Here, we use a combination of in vitro and in vivo models to show that ADAMTS5 enzymatic activity plays a key role in the development of influenza-specific immunity. Influenza virus infection of Adamts5-/- mice resulted in delayed virus clearance, compromised T cell migration and immunity and accumulation of versican, an ADAMTS5 proteoglycan substrate. Our research emphasises the importance of ADAMTS5 expression in the control of influenza virus infection and highlights the potential for development of ADAMTS5-based therapeutic strategies to reduce morbidity and mortality.


Asunto(s)
Proteína ADAMTS5/fisiología , Inmunidad Celular/fisiología , Orthomyxoviridae/inmunología , Linfocitos T/inmunología , Proteína ADAMTS5/genética , Animales , Inmunofenotipificación , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Versicanos/metabolismo , Pérdida de Peso
18.
Vaccine ; 34(9): 1172-9, 2016 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-26826545

RESUMEN

Influenza viruses are promising mucosal vaccine vectors for HIV but their use has been limited by difficulties in engineering the expression of large amounts of foreign protein. We developed recombinant influenza viruses incorporating the HIV-1 p24 gag capsid into the NS-segment of PR8 (H1N1) and X31 (H3N2) influenza viruses with the use of multiple 2A ribosomal skip sequences. Despite the insertion of a sizable HIV-1 gene into the influenza genome, recombinant viruses were readily rescued to high titers. Intracellular expression of p24 capsid was confirmed by in vitro infection assays. The recombinant influenza viruses were subsequently tested as mucosal vaccines in BALB/c mice. Recombinant viruses were attenuated and safe in immunized mice. Systemic and mucosal HIV-specific CD8 T-cell responses were elicited in mice that were immunized via intranasal route with a prime-boost regimen. Isolated HIV-specific CD8 T-cells displayed polyfunctional cytokine and degranulation profiles. Mice boosted via intravaginal route induced recall responses from the distal lung mucosa and developed heightened HIV-specific CD8 T-cell responses in the vaginal mucosa. These findings demonstrate the potential utility of recombinant influenza viruses as vaccines for mucosal immunity against HIV-1 infection.


Asunto(s)
Vacunas contra el SIDA/inmunología , Linfocitos T CD8-positivos/inmunología , Proteína p24 del Núcleo del VIH/inmunología , Inmunidad Mucosa , Subtipo H1N1 del Virus de la Influenza A , Subtipo H3N2 del Virus de la Influenza A , Animales , Línea Celular , Femenino , Ingeniería Genética , VIH-1 , Humanos , Inmunización Secundaria , Pulmón/inmunología , Ratones Endogámicos BALB C , Membrana Mucosa/inmunología , Vacunas Atenuadas/inmunología , Vacunas Sintéticas/inmunología , Vagina/inmunología
19.
Rev Med Virol ; 25(6): 406-30, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26467906

RESUMEN

The emergence of H5N1 highly pathogenic avian influenza has caused a heavy socio-economic burden through culling of poultry to minimise human and livestock infection. Although human infections with H5N1 have to date been limited, concerns for the pandemic potential of this zoonotic virus have been greatly intensified following experimental evidence of aerosol transmission of H5N1 viruses in a mammalian infection model. In this review, we discuss the dominance of the haemagglutinin cleavage site motif as a pathogenicity determinant, the host-pathogen molecular interactions driving cleavage activation, reverse genetics manipulations and identification of residues key to haemagglutinin cleavage site functionality and the mechanisms of cell and tissue damage during H5N1 infection. We specifically focus on the disease in chickens, as it is in this species that high pathogenicity frequently evolves and from which transmission to the human population occurs. With >75% of emerging infectious diseases being of zoonotic origin, it is necessary to understand pathogenesis in the primary host to explain spillover events into the human population.


Asunto(s)
Glicoproteínas Hemaglutininas del Virus de la Influenza/metabolismo , Interacciones Huésped-Patógeno , Subtipo H5N1 del Virus de la Influenza A/patogenicidad , Gripe Aviar/patología , Gripe Aviar/virología , Proteolisis , Factores de Virulencia/metabolismo , Animales , Pollos , Humanos , Gripe Aviar/transmisión , Zoonosis/patología , Zoonosis/transmisión , Zoonosis/virología
20.
J Virol ; 89(5): 2672-83, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25520513

RESUMEN

UNLABELLED: Highly pathogenic avian influenza virus infection is associated with severe mortality in both humans and poultry. The mechanisms of disease pathogenesis and immunity are poorly understood although recent evidence suggests that cytokine/chemokine dysregulation contributes to disease severity following H5N1 infection. Influenza A virus infection causes a rapid influx of inflammatory cells, resulting in increased reactive oxygen species production, cytokine expression, and acute lung injury. Proinflammatory stimuli are known to induce intracellular reactive oxygen species by activating NADPH oxidase activity. We therefore hypothesized that inhibition of this activity would restore host cytokine homeostasis following avian influenza virus infection. A panel of airway epithelial and immune cells from mammalian and avian species were infected with A/Puerto Rico/8/1934 H1N1 virus, low-pathogenicity avian influenza H5N3 virus (A/duck/Victoria/0305-2/2012), highly pathogenic avian influenza H5N1 virus (A/chicken/Vietnam/0008/2004), or low-pathogenicity avian influenza H7N9 virus (A/Anhui/1/2013). Quantitative real-time reverse transcriptase PCR showed that H5N1 and H7N9 viruses significantly stimulated cytokine (interleukin-6, beta interferon, CXCL10, and CCL5) production. Among the influenza-induced cytokines, CCL5 was identified as a potential marker for overactive immunity. Apocynin, a Nox2 inhibitor, inhibited influenza-induced cytokines and reactive oxygen species production, although viral replication was not significantly altered in vitro. Interestingly, apocynin treatment significantly increased influenza virus-induced mRNA and protein expression of SOCS1 and SOCS3, enhancing negative regulation of cytokine signaling. These findings suggest that apocynin or its derivatives (targeting host responses) could be used in combination with antiviral strategies (targeting viruses) as therapeutic agents to ameliorate disease severity in susceptible species. IMPORTANCE: Highly pathogenic avian influenza virus infection causes severe morbidity and mortality in both humans and poultry. Wide-spread antiviral resistance necessitates the need for the development of additional novel therapeutic measures to modulate overactive host immune responses after infection. Disease severity following avian influenza virus infection can be attributed in part to hyperinduction of inflammatory mediators such as cytokines, chemokines, and reactive oxygen species. Our study shows that highly pathogenic avian influenza H5N1 virus and low-pathogenicity avian influenza H7N9 virus (both associated with human fatalities) promote inactivation of FoxO3 and downregulation of the TAM receptor tyrosine kinase, Tyro3, leading to augmentation of the inflammatory cytokine response. Inhibition of influenza-induced reactive oxygen species with apocynin activated FoxO3 and stimulated SOCS1 and SOCS3 proteins, restoring cytokine homeostasis. We conclude that modulation of host immune responses with antioxidant and/or anti-inflammatory agents in combination with antiviral therapy may have important therapeutic benefits.


Asunto(s)
Virus de la Influenza A/inmunología , Especies Reactivas de Oxígeno/toxicidad , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Acetofenonas/metabolismo , Animales , Antioxidantes/metabolismo , Línea Celular , Pollos , Citocinas/biosíntesis , Patos , Perfilación de la Expresión Génica , Humanos , Especies Reactivas de Oxígeno/antagonistas & inhibidores , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteína 1 Supresora de la Señalización de Citocinas , Proteína 3 Supresora de la Señalización de Citocinas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...