Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Vaccines (Basel) ; 12(3)2024 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-38543958

RESUMO

Porcine circovirus type 2 (PCV2) is the main causative agent of porcine circovirus-associated disease (PCVAD) that profoundly impacts the swine industry worldwide. While most of the commercial PCV vaccines are developed based on PCV genotype 2a (PCV2a), PCV genotype 2b (PCV2b) has become predominant since 2003. In this study, we developed and evaluated DNA-based bivalent vaccines covering both PCV2a and PCV2b. We generated a new immunogen, PCV2b-2a, by combining consensus sequences of the PCV2a and PCV2b capsid proteins (Cap2a and Cap2b) in a form of fusion protein. We also examined whether modifications of the PCV2b-2a fusion protein with a signal sequence (SS) and granulocyte macrophage-colony stimulating factor (GM-CSF) fusing with interleukine-4 (IL-4) (GI) could further improve the vaccine immunogenicity. An immunogenicity study of BALB/cAJcl mice revealed that the DNA vector pVAX1 co-expressing PCV2b-2a and GI (pVAX1.PCV2b-2a-GI) was most potent at inducing both antibody and cellular immune responses against Cap2a and Cap2b. Interestingly, the vaccines skewed the immune response towards Th1 phenotype (IgG2a > IgG1). By performing ELISA and ELISpot with predicted epitope peptides, the three most immunogenic B cell epitopes and five putative T cell epitopes were identified on Cap2a and Cap2b. Importantly, our DNA vaccines elicited broad immune responses recognizing both genotype-specific and PCV2-conserved epitopes. Sera from mice immunized with the DNAs expressing PCV2b-2a and PCV2b-2a-GI significantly inhibited PCV2a cell entry at serum dilution 1:8. All these results suggest a great potential of our PCV2b-2a-based vaccines, which can be further developed for use in other vaccine platforms to achieve both vaccine efficacy and economical production cost.

2.
PLoS One ; 18(11): e0292833, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37922253

RESUMO

The innate immune response is a first-line defense mechanism triggered by rabies virus (RABV). Interferon (IFN) signaling and ISG products have been shown to confer resistance to RABV at various stages of the virus's life cycle. Human tetherin, also known as bone marrow stromal cell antigen 2 (hBST2), is a multifunctional transmembrane glycoprotein induced by IFN that has been shown to effectively counteract many viruses through diverse mechanisms. Here, we demonstrate that hBST2 inhibits RABV budding by tethering new virions to the cell surface. It was observed that release of virus-like particles (VLPs) formed by RABV G (RABV-G VLPs), but not RABV M (RABV-G VLPs), were suppressed by hBST2, indicating that RABV-G has a specific effect on the hBST2-mediated restriction of RABV. The ability of hBST2 to prevent the release of RABV-G VLPs and impede RABV growth kinetics is retained even when hBST2 has mutations at dimerization and/or glycosylation sites, making hBST2 an antagonist to RABV, with multiple mechanisms possibly contributing to the hBST2-mediated suppression of RABV. Our findings expand the knowledge of host antiviral mechanisms that control RABV infection.


Assuntos
Vírus da Raiva , Raiva , Humanos , Vírus da Raiva/fisiologia , Raiva/prevenção & controle , Glicosilação , Asparagina/metabolismo , Cisteína/metabolismo , Dimerização , Liberação de Vírus , Antígeno 2 do Estroma da Médula Óssea/genética , Antígenos CD/metabolismo , Proteínas Ligadas por GPI/metabolismo
3.
Front Immunol ; 14: 1192604, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37287962

RESUMO

Porcine respiratory disease is multifactorial and most commonly involves pathogen co-infections. Major contributors include swine influenza A (swIAV) and porcine reproductive and respiratory syndrome (PRRSV) viruses. Experimental co-infection studies with these two viruses have shown that clinical outcomes can be exacerbated, but how innate and adaptive immune responses contribute to pathogenesis and pathogen control has not been thoroughly evaluated. We investigated immune responses following experimental simultaneous co-infection of pigs with swIAV H3N2 and PRRSV-2. Our results indicated that clinical disease was not significantly exacerbated, and swIAV H3N2 viral load was reduced in the lung of the co-infected animals. PRRSV-2/swIAV H3N2 co-infection did not impair the development of virus-specific adaptive immune responses. swIAV H3N2-specific IgG serum titers and PRRSV-2-specific CD8ß+ T-cell responses in blood were enhanced. Higher proportions of polyfunctional CD8ß+ T-cell subset in both blood and lung washes were found in PRRSV-2/swIAV H3N2 co-infected animals compared to the single-infected groups. Our findings provide evidence that systemic and local host immune responses are not negatively affected by simultaneous swIAV H3N2/PRRSV-2 co-infection, raising questions as to the mechanisms involved in disease modulation.


Assuntos
Coinfecção , Influenza Humana , Síndrome Respiratória e Reprodutiva Suína , Vírus da Síndrome Respiratória e Reprodutiva Suína , Animais , Suínos , Humanos , Vírus da Influenza A Subtipo H3N2 , Imunidade
4.
PeerJ ; 11: e14918, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36883057

RESUMO

Alveolar macrophages are tissue-resident immune cells that protect epithelial cells in the alveoli from invasion by pathogens, including severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Therefore, the interaction between macrophages and SARS-CoV-2 is inevitable. However, little is known about the role of macrophages in SARS-CoV-2 infection. Here, we generated macrophages from human induced pluripotent stem cells (hiPSCs) to investigate the susceptibility of hiPSC-derived macrophages (iMΦ) to the authentic SARS-CoV-2 Delta (B.1.617.2) and Omicron (B.1.1.529) variants as well as their gene expression profiles of proinflammatory cytokines during infection. With undetectable angiotensin-converting enzyme 2 (ACE2) mRNA and protein expression, iMΦ were susceptible to productive infection with the Delta variant, whereas infection of iMΦ with the Omicron variant was abortive. Interestingly, Delta induced cell-cell fusion or syncytia formation in iMΦ, which was not observed in Omicron-infected cells. However, iMΦ expressed moderate levels of proinflammatory cytokine genes in response to SARS-CoV-2 infection, in contrast to strong upregulation of these cytokine genes in response to polarization by lipopolysaccharide (LPS) and interferon-gamma (IFN-γ). Overall, our findings indicate that the SARS-CoV-2 Delta variant can replicate and cause syncytia formation in macrophages, suggesting that the Delta variant can enter cells with undetectable ACE2 levels and exhibit greater fusogenicity.


Assuntos
COVID-19 , Células Gigantes , Células-Tronco Pluripotentes Induzidas , Humanos , Enzima de Conversão de Angiotensina 2/genética , COVID-19/virologia , Citocinas/genética , Macrófagos , SARS-CoV-2/genética
5.
Sci Rep ; 12(1): 20474, 2022 11 28.
Artigo em Inglês | MEDLINE | ID: mdl-36443360

RESUMO

Sugarcane accounts for a large portion of the worlds sugar production. Modern commercial cultivars are complex hybrids of S. officinarum, S. spontaneum, and several other Saccharum species, resulting in an auto-allopolyploid with 8-12 copies of each chromosome. The current genome assembly gold standard is to generate a long read assembly followed by chromatin conformation capture sequencing to scaffold. We used the PacBio RSII and chromatin conformation capture sequencing to sequence and assemble the genome of a South East Asian commercial sugarcane cultivar, known as Khon Kaen 3. The Khon Kaen 3 genome assembled into 104,477 contigs totalling 7 Gb, which scaffolded into 56 pseudochromosomes containing 5.2 Gb of sequence. Genome annotation produced 242,406 genes from 30,927 orthogroups. Aligning the Khon Kaen 3 genome sequence to S. officinarum and S. spontaneum revealed a high level of apparent recombination, indicating a chimeric assembly. This assembly error is explained by high nucleotide identity between S. officinarum and S. spontaneum, where 91.8% of S. spontaneum aligns to S. officinarum at 94% identity. Thus, the subgenomes of commercial sugarcane are so similar that using short reads to correct long PacBio reads produced chimeric long reads. Future attempts to sequence sugarcane must take this information into account.


Assuntos
Saccharum , Saccharum/genética , Tailândia , Cromatina , Grão Comestível , Análise de Sequência de DNA
6.
Front Immunol ; 12: 758368, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34858411

RESUMO

The porcine respiratory disease complex (PRDC) is responsible for significant economic losses in the pig industry worldwide. Porcine reproductive and respiratory syndrome virus (PRRSV) and swine influenza virus are major viral contributors to PRDC. Vaccines are cost-effective measures for controlling PRRS, however, their efficacy in the context of co-infections has been poorly investigated. In this study, we aimed to determine the effect of PRRSV-2 and swine influenza H3N2 virus co-infection on the efficacy of PRRSV modified live virus (MLV) vaccination, which is widely used in the field. Following simultaneous challenge with contemporary PRRSV-2 and H3N2 field isolates, we found that the protective effect of PRRS MLV vaccination on clinical disease and pathology was abrogated, although viral load was unaffected and antibody responses were enhanced. In contrast, co-infection in non-immunized animals reduced PRRSV-2 viremia and H3N2 virus load in the upper respiratory tract and potentiated T cell responses against both PRRSV-2 and H3N2 in the lung. Further analysis suggested that an upregulation of inhibitory cytokines gene expression in the lungs of vaccinated pigs may have influenced responses to H3N2 and PRRSV-2. These findings provide important insights into the effect of viral co-infections on PRRS vaccine efficacy that may help identify more effective vaccination strategies against PRDC in the field.


Assuntos
Coinfecção/veterinária , Vírus da Influenza A Subtipo H3N2/imunologia , Infecções por Orthomyxoviridae/imunologia , Síndrome Respiratória e Reprodutiva Suína/prevenção & controle , Vírus da Síndrome Respiratória e Reprodutiva Suína/imunologia , Vacinas Virais/imunologia , Animais , Anticorpos Antivirais/biossíntese , Coinfecção/imunologia , Coinfecção/virologia , Citocinas/biossíntese , Citocinas/genética , Conjuntos de Dados como Assunto , Cães , Feminino , Células Madin Darby de Rim Canino , Infecções por Orthomyxoviridae/complicações , Infecções por Orthomyxoviridae/virologia , Síndrome Respiratória e Reprodutiva Suína/virologia , Suínos , Vacinação/veterinária , Eficácia de Vacinas , Vacinas Atenuadas/imunologia , Carga Viral , Viremia/prevenção & controle , Viremia/virologia
7.
Int J Mol Sci ; 22(21)2021 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-34769416

RESUMO

Rabies is a deadly viral disease caused by the rabies virus (RABV), transmitted through a bite of an infected host, resulting in irreversible neurological symptoms and a 100% fatality rate in humans. Despite many aspects describing rabies neuropathogenesis, numerous hypotheses remain unanswered and concealed. Observations obtained from infected primary neurons or mouse brain samples are more relevant to human clinical rabies than permissive cell lines; however, limitations regarding the ethical issue and sample accessibility become a hurdle for discovering new insights into virus-host interplays. To better understand RABV pathogenesis in humans, we generated human-induced pluripotent stem cell (hiPSC)-derived neurons to offer the opportunity for an inimitable study of RABV infection at a molecular level in a pathologically relevant cell type. This study describes the characteristics and detailed proteomic changes of hiPSC-derived neurons in response to RABV infection using LC-MS/MS quantitative analysis. Gene ontology (GO) enrichment of differentially expressed proteins (DEPs) reveals temporal changes of proteins related to metabolic process, immune response, neurotransmitter transport/synaptic vesicle cycle, cytoskeleton organization, and cell stress response, demonstrating fundamental underlying mechanisms of neuropathogenesis in a time-course dependence. Lastly, we highlighted plausible functions of heat shock cognate protein 70 (HSC70 or HSPA8) that might play a pivotal role in regulating RABV replication and pathogenesis. Our findings acquired from this hiPSC-derived neuron platform help to define novel cellular mechanisms during RABV infection, which could be applicable to further studies to widen views of RABV-host interaction.


Assuntos
Células-Tronco Pluripotentes Induzidas/metabolismo , Neurônios/metabolismo , Proteoma/metabolismo , Vírus da Raiva/metabolismo , Raiva/virologia , Células Cultivadas , Interações Hospedeiro-Patógeno , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/virologia , Neurônios/citologia , Neurônios/virologia , Raiva/metabolismo , Vírus da Raiva/isolamento & purificação , Vírus da Raiva/patogenicidade
8.
Arch Virol ; 162(9): 2553-2563, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28474223

RESUMO

The ORF3 accessory protein has been shown to impede reverse genetics of cell-culture-adapted porcine epidemic diarrhea virus (PEDV). Its absence or truncated variants are also associated with viral attenuation in vivo. Here, three ORF3 variants (ORF3NP12, ORF3NP14 and ORF3RB14) and their truncated counterparts were investigated for their regulatory role in recovery of cell-adapted PEDV in vitro. We demonstrate that ORF3NP12, but not the truncated form, can inhibit recovery of reverse-genetics-derived PEDV when expressed in trans. When testing with other RNA viruses, ORF3 was found to inhibit rescue of porcine respiratory and reproductive syndrome virus (PRRSV), but not of influenza virus. Interestingly, results from mutagenesis of ORF3NP12 suggest that F81 and M167 are responsible for impairing PEDV rescue in vitro. By changing specific residues of ORF3, the recombinant PEDV bearing the modified ORF3NP12 can be productively propagated in VeroE6-APN cells. These results may provide mechanistic insights into ORF3-mediated inhibition of PEDV replication in new host cells.


Assuntos
Vírus da Diarreia Epidêmica Suína/fisiologia , Proteínas Virais/metabolismo , Replicação Viral/fisiologia , Sequência de Aminoácidos , Animais , Chlorocebus aethiops , Infecções por Coronavirus/veterinária , Infecções por Coronavirus/virologia , Regulação Viral da Expressão Gênica/fisiologia , Células HEK293 , Humanos , Mutação Puntual , Suínos , Doenças dos Suínos/virologia , Células Vero , Proteínas Virais/genética
9.
J Gen Virol ; 96(8): 2206-2218, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25979733

RESUMO

Porcine epidemic diarrhoea virus (PEDV) causes acute diarrhoea and dehydration in swine of all ages, with significant mortality in neonatal pigs. The recent rise of PEDV outbreaks in Asia and North America warrants an urgent search for effective vaccines. However, PEDV vaccine research has been hampered by difficulties in isolating and propagating the virus in mammalian cells, thereby complicating the recovery of infectious PEDV using a full-length infectious clone. Here, we engineered VeroE6 cells to stably express porcine aminopeptidase N (pAPN) and used them as a platform to obtain a high-growth variant of PEDV, termed PEDVAVCT12. Subsequently, the full-length cDNA clone was constructed by assembling contiguous cDNA fragments encompassing the complete genome of PEDVAVCT12 in a bacterial artificial chromosome. Infectious PEDV could be recovered, and the rescued virus displayed phenotypic properties identical to the parental virus. Interestingly, we found that PEDVAVCT12 contained a C-terminal deletion of the spike gene, resulting in disruption of the ORF3 start codon. When a functional ORF3 gene was restored, the recombinant virus could not be rescued, suggesting that ORF3 could suppress PEDV replication in vitro. In addition, a high-growth and genetically stable recombinant PEDV expressing a foreign protein could be rescued by replacing the ORF3 gene with the mCherry gene. Together, the results of this study provide a means to generate genetically defined PEDV as a promising vaccine candidate.


Assuntos
Infecções por Coronavirus/veterinária , DNA Complementar/genética , DNA Viral/genética , Diarreia/veterinária , Vírus da Diarreia Epidêmica Suína/genética , Doenças dos Suínos/virologia , Animais , Sequência de Bases , Chlorocebus aethiops , Infecções por Coronavirus/virologia , DNA Complementar/metabolismo , DNA Viral/metabolismo , Diarreia/virologia , Genoma Viral , Dados de Sequência Molecular , Vírus da Diarreia Epidêmica Suína/isolamento & purificação , Vírus da Diarreia Epidêmica Suína/fisiologia , Suínos , Células Vero
10.
Virus Res ; 195: 1-8, 2015 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-25300804

RESUMO

Porcine reproductive and respiratory syndrome virus (PRRSV) is the causative agent for a swine disease affecting the pig industry worldwide. Infection with PRRSV leads to reproductive complications, respiratory illness, and weak immunity to secondary infections. To better control PRRSV infection, novel approaches for generating control measures are critically needed. Here, in vitro Gibson assembly (GA) of viral genomic cDNA fragments was tested for its use as a quick and simple method to recover infectious PRRSV in cell culture. GA involves the activities of T5-exonuclease, Phusion polymerase, and Taq ligase to join overlapping cDNA fragments in an isothermal condition. Four overlapping cDNA fragments covering the entire PRRSV genome and one vector fragment were used to create a plasmid capable of expressing the PRRSV genome. The assembled product was used to transfect a co-culture of 293T and MARC-145 cells. Supernatants from the transfected cells were then passaged onto MARC-145 cells to rescue infectious virus particles. Verification and characterization of the recovered virus confirmed that the GA protocol generated infectious PRRSV that had similar characteristics to the parental virus. This approach was then tested for the generation of a chimeric virus. By replacing one of the four genomic fragments with that of another virus strain, a chimeric virus was successfully recovered via GA. In conclusion, this study describes for the first time the use of GA as a simple, yet powerful tool for generating infectious PRRSV needed for studying PRRSV biology and developing novel vaccines.


Assuntos
DNA Complementar/genética , Genoma Viral , Vírus da Síndrome Respiratória e Reprodutiva Suína/genética , RNA Viral/genética , Genética Reversa/métodos , Animais , Linhagem Celular , Expressão Gênica , Vetores Genéticos , Plasmídeos , Suínos , Transfecção
11.
Mol Biotechnol ; 56(4): 351-9, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24146431

RESUMO

Influenza virus nonstructural protein-1 (NS1) is abundantly expressed in influenza virus infected cells. NS1 is well recognized for counteracting host antiviral activities and regulating host and viral protein expression. When used as a plasmid component in DNA transfection, NS1 was shown to significantly increase expression levels of a cotransfected gene of different plasmid. Our previous studies demonstrated that addition of an NS1 plasmid increased the expression levels of influenza virus secreted neuraminidase (sNA) gene in 293T cells. In this study, we improved the utilization of NS1 as an enhancer for transient protein expression by generating pFluNS1 plasmid to contain two expression cassettes; one encoding an NS1 gene and another encoding a gene of interest. pFluNS1 is expected to codeliver the NS1 gene into the same cells receiving the gene of interest. The plasmid is therefore designed to induce higher protein expression levels than a cotransfection of an NS1 plasmid and a plasmid containing a gene of interest. To test the efficiency of pFluNS1, influenza virus sNA and non-viral DsRed genes were cloned into pFluNS1. The expression of these genes from pFluNS1 was then compared to the expression from a cotransfection of an NS1 plasmid and an expression plasmid coding for sNA or DsRed. We found that gene expression from pFluNS1 reached equal or higher levels to those derived from the cotransfection. Because the expression from pFluNS1 needs only one plasmid, a lesser amount of transfection reagent was required. Thus, the use of pFluNS1 provides a transfection approach that reduces the cost of protein expression without compromising high levels of protein expression. Together, these data suggest that pFluNS1 can serve as a novel alternative for an efficient transient protein expression in mammalian cells.


Assuntos
Vírus da Influenza A/genética , Influenza Humana/genética , Proteínas não Estruturais Virais/genética , Linhagem Celular , Clonagem Molecular , Regulação Viral da Expressão Gênica , Humanos , Vírus da Influenza A/patogenicidade , Influenza Humana/patologia , Proteínas não Estruturais Virais/biossíntese , Proteínas não Estruturais Virais/isolamento & purificação
12.
PLoS One ; 8(8): e74022, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24015313

RESUMO

Virulent strains of Newcastle disease virus (NDV) can cause devastating disease in chickens worldwide. Although the current vaccines are substantially effective, they do not completely prevent infection, virus shedding and disease. To produce genotype-matched vaccines, a full-genome reverse genetics system has been used to generate a recombinant virus in which the F protein cleavage site has been changed to that of avirulent vaccine virus. In the other strategy, the vaccines have been generated by replacing the F and HN genes of a commercial vaccine strain with those from a genotype-matched virus. However, the protective efficacy of a chimeric virus vaccine has not been directly compared with that of a full-genome virus vaccine developed by reverse genetics. Therefore, in this study, we evaluated the protective efficacy of genotype VII matched chimeric vaccines by generating three recombinant viruses based on avirulent LaSota (genotype II) strain in which the open reading frames (ORFs) encoding the F and HN proteins were replaced, individually or together, with those of the circulating and highly virulent Indonesian NDV strain Ban/010. The cleavage site of the Ban/010 F protein was mutated to the avirulent motif found in strain LaSota. In vitro growth characteristics and a pathogenicity test indicated that all three chimeric viruses retained the highly attenuated phenotype of the parental viruses. Immunization of chickens with chimeric and full-length genome VII vaccines followed by challenge with virulent Ban/010 or Texas GB (genotype II) virus demonstrated protection against clinical disease and death. However, only those chickens immunized with chimeric rLaSota expressing the F or F plus HN proteins of the Indonesian strain were efficiently protected against shedding of Ban/010 virus. Our findings showed that genotype-matched vaccines can provide protection to chickens by efficiently preventing spread of virus, primarily due to the F protein.


Assuntos
Mutação , Doença de Newcastle/prevenção & controle , Vírus da Doença de Newcastle , Proteínas Virais de Fusão , Vacinas Virais , Animais , Linhagem Celular Tumoral , Embrião de Galinha , Galinhas , Humanos , Imunização , Doença de Newcastle/genética , Doença de Newcastle/imunologia , Doença de Newcastle/transmissão , Vírus da Doença de Newcastle/genética , Vírus da Doença de Newcastle/imunologia , Proteínas Virais de Fusão/genética , Proteínas Virais de Fusão/imunologia , Vacinas Virais/genética , Vacinas Virais/imunologia , Vacinas Virais/farmacologia
13.
J Virol Methods ; 178(1-2): 44-51, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21893099

RESUMO

Influenza neuraminidase (NA) is a major target for anti-influenza drugs. With an increasing number of viruses resistant to the anti-NA drug oseltamivir, functionally active recombinant NA is needed for screening novel anti-NA compounds. In this study, the secretable NA (sNA) head domain of influenza A/Vietnam/DT-036/05 (H5N1) virus was expressed successfully in human embryonic kidney (HEK-293T) cells and shown to be enzymatically active. The inclusion of a plasmid encoding nonstructural protein 1 (NS1) of influenza A/Puerto Rico/8/34 virus with the sNA plasmid in the cotransfection demonstrated an increase in H5N1 sNA expression by 7.4 fold. Subsequently, the sNA/NS1 cotransfection protocol in serum-free 293-F suspension cell culture was optimized to develop a rapid transient gene expression (TGE) system for expression of large amounts of H5N1 sNA. Under optimized conditions, NS1 enhanced H5N1 sNA expression by 4.2 fold. The resulting H5N1 sNA displayed comparable molecular weight, glycosylation, K(m) for MUNANA, and K(i) for oseltamivir carboxylate to those of H5N1 NA on the virus surface. Taken together, the NS1-enhancing sNA expression strategy presented in this study could be used for rapid high-level expression of enzymatically active H5N1 sNA in suspension mammalian cells. This strategy may be applied for expression of sNA of other strains of influenza virus as well as the other recombinant proteins.


Assuntos
Fibroblastos/metabolismo , Neuraminidase/metabolismo , Proteínas não Estruturais Virais/metabolismo , Proteínas Virais/metabolismo , Biotecnologia/métodos , Técnicas de Cultura de Células/métodos , Linhagem Celular , Clonagem Molecular , Expressão Gênica , Humanos , Virus da Influenza A Subtipo H5N1/genética , Neuraminidase/biossíntese , Neuraminidase/genética , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas não Estruturais Virais/genética , Proteínas Virais/biossíntese , Proteínas Virais/genética
14.
Immunol Res ; 41(1): 15-25, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18040886

RESUMO

Leishmaniasis is a vector-borne disease found in many countries worldwide. The causative agent of the disease, Leishmania spp., lives as an obligate intracellular parasite within mammalian hosts. Since tissue macrophages are major target cells for parasite replication, the outcome of infection depends largely on the activation status of these cells. L-arginine is a crucial amino acid required for both nitric oxide (NO)-mediated parasite killing and polyamine-mediated parasite replication. This review highlights the significance of L-arginine as a factor determining the outcomes of Leishmania infection in vitro and its influences on host immune responses in vivo. Various therapeutic approaches targeting L-arginine metabolic pathways during infections with Leishmania are also discussed.


Assuntos
Arginina/metabolismo , Imunidade Inata , Leishmania , Leishmaniose/imunologia , Leishmaniose/metabolismo , Animais , Arginase/imunologia , Arginase/metabolismo , Arginina/imunologia , Transportador 2 de Aminoácidos Catiônicos/imunologia , Transportador 2 de Aminoácidos Catiônicos/metabolismo , Humanos , Leishmaniose/parasitologia , Leishmaniose/terapia , Ativação de Macrófagos , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/parasitologia , Camundongos , Óxido Nítrico/imunologia , Óxido Nítrico/metabolismo , Poliaminas/imunologia , Poliaminas/metabolismo , Células Th1/imunologia , Células Th1/metabolismo , Células Th2/imunologia , Células Th2/metabolismo
15.
Int J Parasitol ; 38(3-4): 417-29, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17959178

RESUMO

Leishmania amazonensis infection, occurring predominantly in Central and South America, can manifest itself in several forms, including those of cutaneous and diffuse cutaneous leishmaniasis. The outcome of L. amazonensis infection depends largely on host immune responses to the parasites. While CD4+ T cell activation is a prerequisite for pathogenesis in L. amazonensis-infected mice, the roles of B cells and their antibody production are unclear. In this study, we provide evidence suggesting that B cells and antibodies are involved in disease pathogenesis. We documented a correlation between B cell activation and lesion progress in immunocompetent mice. In the absence of functional B cells and antibodies, JhD mice showed a delayed onset of disease and developed small lesions. Histological examination of these mice revealed a significant reduction in CD4+ and CD8+ T cells, but not in MAC1+ macrophages, at the infection site. In contrast to the wild-type mice that showed typical tissue necrosis, L. amazonensis-infected JhD mice showed no or minimal signs of necrotic foci. A marked reduction in CD4+ T cell proliferation and cytokine (IFN-gamma and IL-10) production in infected JhD mice suggested an involvement of B cells and antibodies in the priming of parasite-specific T cells. This notion was further supported by the observations that adoptive transfer of B cells or antibodies could restore CD4+ T cell activation and migration in infected JhD mice. Moreover, antibody coating of parasites could stimulate dendritic cells to produce high levels of cytokines and increase their ability to prime nai ve CD4+ T cells. Since CD4+ T cells are crucial to disease pathogenesis, this study suggests that B cells and their antibody production enhanced L. amazonensis infection, partially by promoting T cell priming and cellular migration to the infection site.


Assuntos
Anticorpos Antiprotozoários/sangue , Linfócitos B/imunologia , Leishmania/fisiologia , Leishmaniose Cutânea/imunologia , Animais , Brasil , Linfócitos T CD4-Positivos/imunologia , Técnicas de Cocultura , Citocinas/sangue , Células Dendríticas/imunologia , Feminino , Citometria de Fluxo , Interações Hospedeiro-Parasita , Imuno-Histoquímica , Leishmaniose Tegumentar Difusa/imunologia , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Mutantes
16.
Infect Immun ; 75(6): 2802-10, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17387163

RESUMO

Leishmania spp. are obligate intracellular parasites, requiring a suitable microenvironment for their growth within host cells. We previously reported that the growth of Leishmania amazonensis amastigotes in murine macrophages (Mphis) was enhanced in the presence of gamma interferon (IFN-gamma), a Th1 cytokine normally associated with classical Mphi activation and killing of intracellular pathogens. In this study, we provided several lines of evidence suggesting that IFN-gamma-mediated parasite growth enhancement was associated with L-arginine transport via mouse cationic amino acid transporter 2B (mCAT-2B). (i) mRNA expression of Slc7A2, the gene encoding for mCAT-2B, as well as L-arginine transport was increased in IFN-gamma-treated Mphis. (ii) Supplementation of L-arginine in Mphi cultures increased parasite growth. (iii) Parasite growth enhancement in wild-type Mphis was inhibited in the presence of nonmetabolized L-arginine analogues. (iv) IFN-gamma-mediated parasite growth was absent in Mphis derived from mCAT-2B-deficient mice. Although we detected a clear upregulation of mCAT-2B and L-arginine transport, no measurable iNOS or arginase activities were observed in IFN-gamma-treated, infected Mphis. Together, these data suggest an involvement of a novel L-arginine usage independent of iNOS and arginase activities during IFN-gamma-mediated parasite growth enhancement. A possible role of mCAT-2B in supplying L-arginine directly to the parasites for their proliferation is discussed.


Assuntos
Arginina/fisiologia , Transportador 2 de Aminoácidos Catiônicos/fisiologia , Leishmania mexicana/fisiologia , Macrófagos/parasitologia , Animais , Arginina/farmacologia , Transportador 2 de Aminoácidos Catiônicos/farmacologia , Citotoxicidade Imunológica , Leishmania mexicana/crescimento & desenvolvimento , Leishmania mexicana/imunologia , Leishmaniose , Ativação de Macrófagos , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos BALB C
17.
Cytokine ; 35(1-2): 100-6, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16919469

RESUMO

IL-6 and TNF-alpha are proinflammatory cytokines involved in various inflammatory or non-inflammatory disorders characterized by muscle wasting. While infiltrating leukocytes are known to be the major source of these cytokines, it is unclear whether muscle cells also contribute to local inflammation. In this study, we first showed that cultured muscle cells and naive mouse muscle tissue were capable of producing IL-6 and TNF-alpha. We demonstrated an increased expression of IL-6 and TNF-alpha on muscle sections of C57BL/6J mice immunized with acetylcholine receptor (AChR) in the complete Freund's adjuvant (CFA) or with CFA only. In the presence of IL-6 or TNF-alpha, cultured AChR-expressing mouse (G-8) and human (TE671) skeletal muscle cells showed significantly decreased alpha-bungarotoxin-binding sites as measured by cellular ELISA. Moreover, IL-6- or TNF-alpha-treated muscle cells displayed a considerable increase in apoptotic cell ratios. Collectively, this study suggests a direct role for these two cytokines in muscle cell destruction and a possibility of muscle cell damage via an autocrine fashion.


Assuntos
Antígenos/imunologia , Interleucina-6/biossíntese , Células Musculares/imunologia , Receptores Colinérgicos/imunologia , Linfócitos T/imunologia , Fator de Necrose Tumoral alfa/biossíntese , Animais , Antígenos/administração & dosagem , Comunicação Autócrina/imunologia , Linhagem Celular , Linhagem Celular Tumoral , Humanos , Interleucina-6/genética , Camundongos , Camundongos Endogâmicos C57BL , Células Musculares/metabolismo , Células Musculares/patologia , Fator de Necrose Tumoral alfa/genética
18.
Infect Immun ; 72(2): 988-95, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14742545

RESUMO

During Leishmania major infection in mice, gamma interferon (IFN-gamma) plays an essential role in controlling parasite growth and disease progression. In studies designed to ascertain the role of IFN-gamma in Leishmania amazonensis infection, we were surprised to find that IFN-gamma could promote L. amazonensis amastigote replication in macrophages (Mphis), although it activated Mphis to kill promastigotes. The replication-promoting effect of IFN-gamma on amastigotes was independent of the source and genetic background of Mphis, was apparently not affected by surface opsonization of amastigotes, was not mediated by interleukin-10 or transforming growth factor beta, and was observed at different temperatures. Consistent with the different fates of promastigotes and amastigotes in IFN-gamma-stimulated Mphis, L. amazonensis-specific Th1 transfer helped recipient mice control L. amazonensis infection established by promastigotes but not L. amazonensis infection established by amastigotes. On the other hand, IFN-gamma could stimulate Mphis to limit amastigote replication when it was coupled with lipopolysaccharides but not when it was coupled with tumor necrosis factor alpha. Thus, IFN-gamma may play a bidirectional role at the level of parasite-Mphi interactions; when it is optimally coupled with other factors, it has a protective effect against infection, and in the absence of such synergy it promotes amastigote growth. These results reveal a quite unexpected aspect of the L. amazonensis parasite and have important implications for understanding the pathogenesis of the disease and for developing vaccines and immunotherapies.


Assuntos
Interferon gama/farmacologia , Leishmania mexicana/crescimento & desenvolvimento , Leishmaniose Cutânea/etiologia , Macrófagos/parasitologia , Animais , Interleucina-10/fisiologia , Lipopolissacarídeos/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Temperatura , Células Th1/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...