Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Exerc Immunol Rev ; 27: 84-124, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33965901

RESUMO

BACKGROUND: Toll-like receptors (TLRs) are a family of transmembrane pattern recognition receptors that are mainly expressed on immune cells. Recognition of various exogenous and endogenous molecular patterns activates the TLR signalling cascade, which orchestrates an inflammatory immune response. Dysfunctional immune responses, including aberrant TLR signalling, are increasingly implicated in the associations between sedentarism, chronic low-grade systemic inflammation and various non-communicable diseases. Conversely, exercise exerts anti-inflammatory effects, which could be conferred through its immunomodulatory properties, potentially affecting TLRs. This study aims to systematically review the effects of exercise on human TLR expression. METHOD: A systematic literature search of Pubmed, Embase, The Cochrane Library and SPORTDiscus for articles addressing the impact of exercise (as isolated intervention) on TLRs in humans was conducted, ending in February 2020. RESULTS: A total of 66 articles were included. The publications were categorised according to exercise modality and duration: acute resistance exercise (4 studies), acute aerobic exercise (26 studies), resistance training program (9 studies), aerobic training program (16 studies), combined (i.e. resistance and aerobic) training program (8 studies) and chronic exercise not otherwise classifiable (9 studies). Five articles investigated more than one of the aforementioned exercise categories. Several trends could be discerned with regard to the TLR response in the different exercise categories. Acute resistance exercise seemed to elicit TLR upregulation, whereas acute aerobic exercise had less activating potential with the majority of responses being neutral or, especially in healthy participants, downregulatory. Chronic resistance and combined exercise programs predominantly resulted in unaltered or decreased TLR levels. In the chronic aerobic exercise category, mixed effects were observed, but the majority of measurements demonstrated unchanged TLR expression. CONCLUSION: Currently published research supports an interplay between exercise and TLR signalling, which seems to depend on the characteristics of the exercise. However, there was large heterogeneity in the study designs and methodologies. Therefore, additional research is required to further corroborate these findings, to define its pathophysiological implications and to elucidate the mechanism(s) linking exercise to TLR signalling.


Assuntos
Exercício Físico , Treinamento Resistido , Receptores Toll-Like , Humanos , Receptores de Reconhecimento de Padrão , Transdução de Sinais
2.
Cell Immunol ; 312: 51-60, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27993350

RESUMO

Sialoadhesin (Sn) is a surface receptor expressed on macrophages in steady state conditions, but during inflammation, Sn can be upregulated both on macrophages and on circulating monocytes. It was shown for different species that Sn becomes internalized after binding with monoclonal antibodies. These features suggest that Sn is a potential target for immunotherapies. In this study, human and mouse macrophages were treated with anti-Sn monoclonal antibodies or F(ab')2 fragments and the effect of their binding to Sn on phagocytosis was analyzed. Binding of antibodies to Sn resulted in delayed and reduced phagocytosis of fluorescent beads. No effect was observed on Fc-mediated phagocytosis or phagocytosis of bacteria by human macrophages. In contrast, an enhanced phagocytosis of bacteria by mouse macrophages was detected. These results showed that stimulation of Sn could have different effects on macrophage phagocytosis, depending both on the type of phagocytosis and cellular background.


Assuntos
Anticorpos Monoclonais/farmacologia , Inflamação/imunologia , Macrófagos/efeitos dos fármacos , Lectina 1 Semelhante a Ig de Ligação ao Ácido Siálico/metabolismo , Animais , Células Cultivadas , Humanos , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Fagocitose/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Lectina 1 Semelhante a Ig de Ligação ao Ácido Siálico/imunologia , Especificidade da Espécie
3.
Virol J ; 14(1): 157, 2017 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-28818113

RESUMO

BACKGROUND: Host proteases have been shown to play important roles in many viral activities such as entry, uncoating, viral protein production and disease induction. Therefore, these cellular proteases are putative targets for the development of antivirals that inhibit their activity. Host proteases have been described to play essential roles in Ebola, HCV, HIV and influenza, such that specific protease inhibitors are able to reduce infection. RSV utilizes a host protease in its replication cycle but its potential as antiviral target is unknown. Therefore, we evaluated the effect of protease inhibitors on RSV infection. METHODS: To measure the sensitivity of RSV infection to protease inhibitors, cells were infected with RSV and incubated for 18 h in the presence or absence of the inhibitors. Cells were fixed, stained and studied using fluorescence microscopy. RESULTS: Several protease inhibitors, representing different classes of proteases (AEBSF, Pepstatin A, E-64, TPCK, PMSF and aprotinin), were tested for inhibitory effects on an RSV A2 infection of HEp-2 cells. Different treatment durations, ranging from 1 h prior to inoculation and continuing for 18 h during the assay, were evaluated. Of all the inhibitors tested, AEBSF and TPCK significantly decreased RSV infection. To ascertain that the observed effect of AEBSF was not a specific feature related to HEp-2 cells, A549 and BEAS-2B cells were also used. Similar to HEp-2, an almost complete block in the number of RSV infected cells after 18 h of incubation was observed and the effect was dose-dependent. To gain insight into the mechanism of this inhibition, AEBSF treatment was applied during different phases of an infection cycle (pre-, peri- and post-inoculation treatment). The results from these experiments indicate that AEBSF is mainly active during the early entry phase of RSV. The inhibitory effect was also observed with other RSV isolates A1998/3-2 and A2000/3-4, suggesting that this is a general feature of RSV. CONCLUSION: RSV infection can be inhibited by broad serine protease inhibitors, AEBSF and TPCK. We confirmed that AEBSF inhibition is independent of the cell line used or RSV strain. The time point at which treatment with the inhibitor was most potent, was found to coincide with the expected moment of entry of the virion with the host cell.


Assuntos
Infecções por Vírus Respiratório Sincicial/virologia , Vírus Sincicial Respiratório Humano/efeitos dos fármacos , Vírus Sincicial Respiratório Humano/patogenicidade , Inibidores de Serina Proteinase/farmacologia , Sulfonas/antagonistas & inibidores , Internalização do Vírus/efeitos dos fármacos , Células A549 , Aprotinina/farmacologia , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Endopeptidases/efeitos dos fármacos , Humanos , Cinética , Leucina/análogos & derivados , Leucina/antagonistas & inibidores , Pepstatinas/antagonistas & inibidores , Fatores de Tempo , Tosilfenilalanil Clorometil Cetona/antagonistas & inibidores , Proteínas Virais/metabolismo
4.
Sci Rep ; 14(1): 12653, 2024 06 02.
Artigo em Inglês | MEDLINE | ID: mdl-38825590

RESUMO

Nonischaemic myocardial fibrosis is associated with cardiac dysfunction, malignant arrhythmias and sudden cardiac death. In the absence of a specific aetiology, its finding as late gadolinium enhancement (LGE) on cardiac magnetic resonance imaging is often attributed to preceding viral myocarditis. Athletes presenting with ventricular arrhythmias often have nonischaemic LGE. Previous studies have demonstrated an adverse effect of exercise on the course of acute viral myocarditis. In this study, we have investigated, for the first time, the impact of endurance training on longer-term outcomes such as myocardial fibrosis and arrhythmogenicity in a murine coxsackievirus B3 (CVB)-induced myocarditis model. Male C57BL/6J mice (n = 72) were randomly assigned to 8 weeks of forced treadmill running (EEX) or no exercise (SED). Myocarditis was induced 2 weeks later by a single intraperitoneal injection with CVB, versus vehicle in the controls (PBS). In a separate study, mice (n = 30) were subjected to pretraining for 13 weeks (preEEX), without continuation of exercise during myocarditis. Overall, continuation of exercise resulted in a milder clinical course of viral disease, with less weight loss and better preserved running capacity. CVB-EEX and preEEX-CVB mice tended to have a lower mortality rate. At sacrifice (i.e. 6 weeks after inoculation), the majority of virus was cleared from the heart. Histological assessment demonstrated prominent myocardial inflammatory infiltration and cardiomyocyte loss in both CVB groups. Inflammatory lesions in the CVB-EEX group contained higher numbers of pro-inflammatory cells (iNOS-reactive macrophages and CD8+ T lymphocytes) compared to these in CVB-SED. Treadmill running during myocarditis increased interstitial fibrosis [82.4% (CVB-EEX) vs. 56.3% (CVB-SED); P = 0.049]. Additionally, perivascular and/or interstitial fibrosis with extensive distribution was more likely to occur with exercise [64.7% and 64.7% (CVB-EEX) vs. 50% and 31.3% (CVB-SED); P = 0.048]. There was a numerical, but not significant, increase in the number of scars per cross-section (1.9 vs. 1.2; P = 0.195), with similar scar distribution and histological appearance in CVB-EEX and CVB-SED. In vivo electrophysiology studies did not induce sustained monomorphic ventricular tachycardia, only nonsustained (usually polymorphic) runs. Their cumulative beat count and duration paralleled the increased fibrosis between CVB-EEX and CVB-SED, but the difference was not significant (P = 0.084 for each). Interestingly, in mice that were subjected to pretraining only without continuation of exercise during myocarditis, no differences between pretrained and sedentary mice were observed at sacrifice (i.e. 6 weeks after inoculation and training cessation) with regard to myocardial inflammation, fibrosis, and ventricular arrhythmogenicity. In conclusion, endurance exercise during viral myocarditis modulates the inflammatory process with more pro-inflammatory cells and enhances perivascular and interstitial fibrosis development. The impact on ventricular arrhythmogenesis requires further exploration.


Assuntos
Arritmias Cardíacas , Infecções por Coxsackievirus , Modelos Animais de Doenças , Enterovirus Humano B , Fibrose , Camundongos Endogâmicos C57BL , Miocardite , Condicionamento Físico Animal , Animais , Miocardite/virologia , Miocardite/patologia , Masculino , Camundongos , Arritmias Cardíacas/etiologia , Infecções por Coxsackievirus/patologia , Infecções por Coxsackievirus/complicações , Miocárdio/patologia , Treino Aeróbico
5.
BMC Biotechnol ; 13: 33, 2013 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-23575465

RESUMO

BACKGROUND: Sialoadhesin (Sn)-expressing monocytes/macrophages have been associated with several diseases like inflammatory and autoimmune disorders as well as viral infections, and they also appear to play a role in the initiation of an adaptive immune response. This makes Sn-expressing cells not only attractive targets for cell-directed therapies, but also an appealing target for vaccination. Furthermore, since Sn was shown to be an endocytic receptor, the conjugation of effector molecules to an Sn-specific ligand should allow intracellular delivery of these conjugates. Previously, we developed functional Sn-specific immunoconjugates that were generated via chemical coupling. Although successful, the system requires significant optimization for each immunoconjugate to be made. To generate a more flexible and controlled system, we developed a recombinant antibody vector allowing the creation of genetic antibody fusion constructs. This paper reports on the characterization of the recombinant antibody and the evaluation of its use for Sn-directed targeting. RESULTS: The variable domains of the porcine Sn-specific monoclonal antibody 41D3 were sequenced and cloned in frame with a mouse IgG1 backbone. Transfection of HEK293T cells with the resulting plasmid led to the secretion of fully assembled IgG into the culture medium. This recombinant antibody rec41D3 was shown to specifically bind to porcine Sn with a comparable affinity as the native monoclonal antibody. In addition, rec41D3 also induced Sn endocytosis in primary macrophages and resided for prolonged times in early/late endosomes. To allow the generation of antibody fusion constructs, a multiple cloning site was introduced at the C-terminus of the heavy chain. Two fusion constructs were generated, one containing a V5 peptide tag and one containing an eGFP molecule. Both constructs were shown to be efficiently produced in HEK293T cells and easily purified using standard protein G chromatography. In addition, both V5 and eGFP were shown to be co-internalized together with rec41D3 into Sn-expressing primary macrophages. CONCLUSIONS: A recombinant antibody allowing targeted delivery of peptides and proteins to Sn-expressing macrophages was developed. Production and purification of antibody fusion constructs was possible without major optimization and with batch to batch consistency, confirming the development of a versatile antibody vector to evaluate Sn-directed targeting strategies in a porcine animal model.


Assuntos
Anticorpos Monoclonais/metabolismo , Macrófagos/metabolismo , Lectina 1 Semelhante a Ig de Ligação ao Ácido Siálico/imunologia , Animais , Anticorpos Monoclonais/genética , Anticorpos Monoclonais/imunologia , Endocitose , Endossomos/metabolismo , Proteínas de Fluorescência Verde/imunologia , Células HEK293 , Humanos , Imunoglobulina G/genética , Imunoglobulina G/metabolismo , Região Variável de Imunoglobulina/genética , Região Variável de Imunoglobulina/metabolismo , Lisossomos/metabolismo , Macrófagos/imunologia , Camundongos , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Suínos , Transfecção
6.
Sci Transl Med ; 15(710): eadg6050, 2023 08 23.
Artigo em Inglês | MEDLINE | ID: mdl-37611082

RESUMO

The RSVPreF3-AS01 vaccine, containing the respiratory syncytial virus (RSV) prefusion F protein and the AS01 adjuvant, was previously shown to boost neutralization responses against historical RSV strains and to be efficacious in preventing RSV-associated lower respiratory tract diseases in older adults. Although RSV F is highly conserved, variation does exist between strains. Here, we characterized variations in the major viral antigenic sites among contemporary RSV sequences when compared with RSVPreF3 and showed that, in older adults, RSVPreF3-AS01 broadly boosts neutralization responses against currently dominant and antigenically distant RSV strains. RSV-neutralizing responses are thought to play a central role in preventing RSV infection. Therefore, the breadth of RSVPreF3-AS01-elicited neutralization responses may contribute to vaccine efficacy against contemporary RSV strains and those that may emerge in the future.


Assuntos
Infecções por Vírus Respiratório Sincicial , Vacinas , Humanos , Idoso , Vírus Sinciciais Respiratórios , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Antígenos Virais
7.
Microbiol Spectr ; 11(3): e0436822, 2023 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-37212711

RESUMO

The respiratory syncytial virus (RSV) represents the leading cause of viral lower respiratory tract infections (LRTI) in children worldwide and is associated with significant morbidity and mortality rates. The clinical picture of an RSV infection differs substantially between patients, and the role of viral co-infections is poorly investigated. During two consecutive winter seasons from October 2018 until February 2020, we prospectively included children up to 2 years old presenting with an acute LRTI, both ambulatory and hospitalized. We collected clinical data and tested nasopharyngeal secretions for a panel of 16 different respiratory viruses with multiplex RT-qPCR. Disease severity was assessed with traditional clinical parameters and scoring systems. A total of 120 patients were included, of which 91.7% were RSV positive; 42.5% of RSV-positive patients had a co-infection with at least one other respiratory virus. We found that patients suffering from a single RSV infection had higher pediatric intensive care unit (PICU) admission rates (OR = 5.9, 95% CI = 1.53 to 22.74), longer duration of hospitalization (IRR = 1.25, 95% CI = 1.03 to 1.52), and a higher Bronchiolitis Risk of Admission Score (BRAS) (IRR = 1.31, 95% CI = 1.02 to 1.70) compared to patients with RSV co-infections. No significant difference was found in saturation on admission, O2 need, or ReSViNET-score. In our cohort, patients with a single RSV infection had increased disease severity compared to patients with RSV co-infections. This suggests that the presence of viral co-infections might influence the course of RSV bronchiolitis, but heterogeneity and small sample size in our study prevents us from drawing strong conclusions. IMPORTANCE RSV is worldwide the leading cause of serious airway infections. Up to 90% of children will be infected by the age of 2. RSV symptoms are mostly mild and typically mimic a common cold in older children and adolescents, but younger children can develop severe lower respiratory tract disease, and currently it is unclear why certain children develop severe disease while others do not. In this study, we found that children with a single RSV infection had a higher disease severity compared to patients with viral co-infections, suggesting that the presence of a viral co-infection could influence the course of an RSV bronchiolitis. As preventive and therapeutic options for RSV-associated disease are currently limited, this finding could potentially guide physicians to decide which patients might benefit from current or future treatment options early in the course of disease, and therefore, warrants further investigation.


Assuntos
Bronquiolite , Coinfecção , Infecções por Vírus Respiratório Sincicial , Vírus Sincicial Respiratório Humano , Infecções Respiratórias , Viroses , Vírus , Criança , Adolescente , Humanos , Lactente , Coinfecção/epidemiologia , Infecções por Vírus Respiratório Sincicial/epidemiologia , Infecções por Vírus Respiratório Sincicial/diagnóstico , Bronquiolite/epidemiologia , Infecções Respiratórias/epidemiologia , Fatores de Risco
8.
J Gen Virol ; 93(Pt 7): 1548-1555, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22492916

RESUMO

Porcine circovirus 2 (PCV2) is the causative agent of porcine circovirus-associated diseases in pigs. Previously, it was demonstrated that mAbs 16G12, 38C1, 63H3 and 94H8 directed against the PCV2 capsid protein recognize PCV2 strains Stoon-1010 (PCV2a), 48285 (PCV2b), 1121 (PCV2a), 1147 (PCV2b) and II9F (PCV2b), but only neutralize Stoon-1010 and 48285. This points to the existence of two distinct PCV2 neutralization phenotypes: phenotype α (mAb recognition with neutralization; Stoon-1010 and 48285) and phenotype ß (mAb recognition without neutralization; 1121, 1147 and II9F). In the present study, amino acids that are important in determining the neutralization phenotype were identified in the capsid. Mutation of T at position 190 to A in strain 48285 (phenotype α) resulted in a capsid resembling that of strain 1147 (phenotype ß) and caused a loss of neutralization (switch from α to ß). Mutations of P at position 151 to T and A at position 190 to T in strain II9F (phenotype ß) resulted in a capsid resembling that of strain 48285 (phenotype α) and gave a gain of neutralization (switch from ß to α). Mutations of T at position 131 to P and of E at position 191 to R in Stoon-1010 (phenotype α) changed the capsid into that of 1121 (phenotype ß) and reduced neutralization (switch from α to ß). This study demonstrated that single amino acid changes in the capsid result in a phenotypic switch from α to ß or ß to α.


Assuntos
Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/imunologia , Circovirus/genética , Circovirus/imunologia , Mutação de Sentido Incorreto , Substituição de Aminoácidos , Animais , Anticorpos Monoclonais/imunologia , Testes de Neutralização
9.
PLoS Pathog ; 6(1): e1000730, 2010 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-20084110

RESUMO

The porcine reproductive and respiratory syndrome virus (PRRSV) is a major threat to swine health worldwide and is considered the most significant viral disease in the swine industry today. In past years, studies on the entry of the virus into its host cell have led to the identification of a number of essential virus receptors and entry mediators. However, viral counterparts for these molecules have remained elusive and this has made rational development of new generation vaccines impossible. The main objective of this study was to identify the viral counterparts for sialoadhesin, a crucial PRRSV receptor on macrophages. For this purpose, a soluble form of sialoadhesin was constructed and validated. The soluble sialoadhesin could bind PRRSV in a sialic acid-dependent manner and could neutralize PRRSV infection of macrophages, thereby confirming the role of sialoadhesin as an essential PRRSV receptor on macrophages. Although sialic acids are present on the GP(3), GP(4) and GP(5) envelope glycoproteins, only the M/GP(5) glycoprotein complex of PRRSV was identified as a ligand for sialoadhesin. The interaction was found to be dependent on the sialic acid binding capacity of sialoadhesin and on the presence of sialic acids on GP(5). These findings not only contribute to a better understanding of PRRSV biology, but the knowledge and tools generated in this study also hold the key to the development of a new generation of PRRSV vaccines.


Assuntos
Glicoproteínas/metabolismo , Glicoproteínas de Membrana/metabolismo , Ácido N-Acetilneuramínico/metabolismo , Vírus da Síndrome Respiratória e Reprodutiva Suína/patogenicidade , Receptores Imunológicos/metabolismo , Proteínas do Envelope Viral/metabolismo , Animais , Western Blotting , Eletroforese em Gel de Poliacrilamida , Macrófagos/metabolismo , Macrófagos/virologia , Reação em Cadeia da Polimerase , Vírus da Síndrome Respiratória e Reprodutiva Suína/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Lectina 1 Semelhante a Ig de Ligação ao Ácido Siálico , Suínos , Ligação Viral
10.
Vet Res ; 43: 47, 2012 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-22630829

RESUMO

Porcine reproductive and respiratory syndrome virus (PRRSV) is an arterivirus that shows a restricted in vivo tropism for subsets of porcine macrophages, with alveolar macrophages being major target cells. The virus is associated with respiratory problems in pigs of all ages and is commonly isolated on farms with porcine respiratory disease complex (PRDC). Due to virus-induced macrophage death early in infection, PRRSV hampers the innate defence against pathogens in the lungs. In addition, the virus might also directly affect the antimicrobial functions of macrophages. This study examined whether interaction of European genotype PRRSV with primary alveolar macrophages (PAM) affects their phagocytic capacity. Inoculation of macrophages with both subtype I PRRSV (LV) and subtype III PRRSV (Lena) showed that the virus inhibits PAM phagocytosis. Similar results were obtained using inactivated PRRSV (LV), showing that initial interaction of the virion with the cell is sufficient to reduce phagocytosis, and that no productive infection is required. When macrophages were incubated with sialoadhesin- (Sn) or CD163-specific antibodies, two entry mediators of the virus, only Sn-specific antibodies downregulated the phagocytic capacity of PAM, indicating that interaction with Sn, but not CD163, mediates the inhibitory effect of PRRSV on phagocytosis. In conclusion, this study shows that European genotype PRRSV inhibits PAM phagocytosis in vitro, through the interaction with its internalization receptor Sn. If similar events occur in vivo, this interaction may be important in the development of PRDC, as often seen in the field.


Assuntos
Macrófagos Alveolares/imunologia , Fagocitose , Síndrome Respiratória e Reprodutiva Suína/imunologia , Vírus da Síndrome Respiratória e Reprodutiva Suína/imunologia , Lectina 1 Semelhante a Ig de Ligação ao Ácido Siálico/genética , Animais , Síndrome Respiratória e Reprodutiva Suína/virologia , Vírus da Síndrome Respiratória e Reprodutiva Suína/genética , Lectina 1 Semelhante a Ig de Ligação ao Ácido Siálico/metabolismo , Suínos
11.
J Virol ; 84(6): 3101-5, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20032174

RESUMO

Scavenger receptor CD163 is a key entry mediator for porcine reproductive and respiratory syndrome virus (PRRSV). To identify the CD163 protein domains involved in PRRSV infection, deletion mutants and chimeric mutants were created. Infection experiments revealed that scavenger receptor cysteine-rich (SRCR) domain 5 (SRCR 5) is essential for PRRSV infection, while the four N-terminal SRCR domains and the cytoplasmic tail are not required. The remaining CD163 protein domains need to be present but can be replaced by corresponding SRCR domains from CD163-L1, resulting in reduced (SRCR 6 and interdomain regions) or unchanged (SRCR 7 to SRCR 9) infection efficiency. In addition, CD163-specific antibodies recognizing SRCR 5 are able to reduce PRRSV infection.


Assuntos
Antígenos CD/química , Antígenos CD/metabolismo , Antígenos de Diferenciação Mielomonocítica/química , Antígenos de Diferenciação Mielomonocítica/metabolismo , Síndrome Respiratória e Reprodutiva Suína/metabolismo , Vírus da Síndrome Respiratória e Reprodutiva Suína/metabolismo , Receptores de Superfície Celular/química , Receptores de Superfície Celular/metabolismo , Animais , Antígenos CD/genética , Antígenos de Diferenciação Mielomonocítica/genética , Humanos , Síndrome Respiratória e Reprodutiva Suína/virologia , Vírus da Síndrome Respiratória e Reprodutiva Suína/genética , Vírus da Síndrome Respiratória e Reprodutiva Suína/patogenicidade , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Estrutura Terciária de Proteína , Receptores de Superfície Celular/genética , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Receptores Depuradores Classe B/genética , Receptores Depuradores Classe B/metabolismo , Deleção de Sequência , Suínos , Internalização do Vírus
12.
Cell Immunol ; 271(2): 462-73, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21944562

RESUMO

Sialoadhesin (Sn) is a macrophage-restricted endocytic receptor involved in cell-cell, cell-matrix and cell-pathogen interactions. Recently, porcine Sn (pSn) was shown to be involved in signaling and lately Sn is gaining interest as a potential target for immunotherapy. However, little is known about the effect of ligand binding to Sn on macrophage effector functions. In this study, we tested the effect of antibody binding to pSn on macrophage viability, phagocytosis of microspheres, uptake and processing of soluble antigens, reactive oxygen/nitrogen species production, MHC I and MHC II cell surface expression and cytokine production. This was done by treatment of porcine primary alveolar macrophages with the pSn-specific mAb 41D3, or an isotype-matched control mAb. No significant effect on most effector functions under study was observed, except for a significant reduction of phagocytosis. Thus, antibody binding to pSn can downregulate phagocytosis, which could have implications on homeostasis, infectious and immune diseases, and immunotherapy.


Assuntos
Macrófagos Alveolares/imunologia , Glicoproteínas de Membrana/imunologia , Fagocitose/imunologia , Receptores Imunológicos/imunologia , Animais , Anticorpos Monoclonais/administração & dosagem , Apresentação de Antígeno , Membrana Celular/imunologia , Membrana Celular/metabolismo , Citocinas/biossíntese , Antígenos de Histocompatibilidade Classe I/metabolismo , Antígenos de Histocompatibilidade Classe II/metabolismo , Técnicas In Vitro , Ligantes , Macrófagos Alveolares/metabolismo , Glicoproteínas de Membrana/metabolismo , Espécies Reativas de Nitrogênio/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Receptores Imunológicos/metabolismo , Lectina 1 Semelhante a Ig de Ligação ao Ácido Siálico , Suínos
13.
Virol J ; 8: 160, 2011 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-21477345

RESUMO

PRRS is a pig disease of major economic importance that causes respiratory and reproductive problems in pigs. Over the last years it has become clear that PRRSV heterogeneity is increasing. Consequently, this has a potential impact on diagnosis and strategies to counter this disease. The use of sequence-independent PCR techniques for the detection and characterization of PRRSV could be useful to bypass problems associated with the heterogeneity of this virus. A random PCR cloning approach was tested for the characterization of PRRSV strain 07V063 of unknown genetic background that circulated on a Belgian farm. By using this approach, 7305 bp of sequence data were obtained, distributed randomly across the genome. Using RT-PCR with strain-specific primers, the full length sequence (15014 nt) was obtained. Phylogenetic relationships using ORF5 and ORF1a (NSP2) sequences showed that 07V063 was classified in type 1 subtype 1 and that 07V063 was genetically different from prototype Lelystad Virus (LV). 07V063 showed 87-93% aa identity with LV ORFs coding for structural proteins. Most variation (compared to LV) was noticed in Nsp2 (81% identity) with a deletion of 28 aa. This deletion was different from other known deletions in this ORF. In conclusion, it is shown that this random PCR cloning approach can be used for the characterization of new PRRSV strains of unknown genetic background.


Assuntos
Clonagem Molecular/métodos , Genoma Viral , Reação em Cadeia da Polimerase/métodos , Síndrome Respiratória e Reprodutiva Suína/virologia , Vírus da Síndrome Respiratória e Reprodutiva Suína/genética , Vírus da Síndrome Respiratória e Reprodutiva Suína/isolamento & purificação , Sequência de Aminoácidos , Animais , Sequência de Bases , Dados de Sequência Molecular , Filogenia , Síndrome Respiratória e Reprodutiva Suína/diagnóstico , Vírus da Síndrome Respiratória e Reprodutiva Suína/química , Vírus da Síndrome Respiratória e Reprodutiva Suína/classificação , Alinhamento de Sequência , Suínos , Proteínas Virais/química , Proteínas Virais/genética
14.
Viruses ; 13(6)2021 05 27.
Artigo em Inglês | MEDLINE | ID: mdl-34072044

RESUMO

Myocarditis is an inflammatory disease of the heart with viral infections being the most common aetiology. Its complex biology remains poorly understood and its clinical management is one of the most challenging in the field of cardiology. Toll-like receptors (TLRs), a family of evolutionarily conserved pattern recognition receptors, are increasingly known to be implicated in the pathophysiology of viral myocarditis. Their central role in innate and adaptive immune responses, and in the inflammatory reaction that ensues, indeed makes them prime candidates to profoundly affect every stage of the disease process. This review describes the pathogenesis and pathophysiology of viral myocarditis, and scrutinises the role of TLRs in every phase. We conclude with directions for future research in this field.


Assuntos
Imunidade Inata , Inflamação/virologia , Miocardite/imunologia , Miocardite/virologia , Miocárdio/patologia , Receptores Toll-Like/imunologia , Viroses/complicações , Animais , Modelos Animais de Doenças , Humanos , Camundongos , Miocardite/complicações , Miocardite/etiologia , Receptores de Reconhecimento de Padrão , Viroses/imunologia
15.
J Gen Virol ; 91(Pt 7): 1659-67, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20410315

RESUMO

Porcine reproductive and respiratory syndrome virus (PRRSV) emerged in the late 1980s and rapidly became one of the most significant viral pathogens in the swine industry. In vivo, the virus shows a very narrow cell tropism and targets specific subsets of porcine macrophages. The entry of PRRSV into its host cell is the first crucial step in infection and has been the focus of many fundamental studies. This review provides a comprehensive overview of the current knowledge on PRRSV entry into the porcine macrophage, covering virus binding, internalization and genome release, and integrates these findings into a general model of the entry process.


Assuntos
Macrófagos/virologia , Vírus da Síndrome Respiratória e Reprodutiva Suína/fisiologia , Suínos , Internalização do Vírus , Animais , Macrófagos/fisiologia , Tropismo Viral/fisiologia , Replicação Viral/fisiologia
16.
BMC Biotechnol ; 10: 48, 2010 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-20587060

RESUMO

BACKGROUND: Porcine reproductive and respiratory syndrome virus (PRRSV) causes major economic losses in the pig industry worldwide. In vivo, the virus infects a subpopulation of tissue macrophages. In vitro, PRRSV only replicates in primary pig macrophages and African green monkey kidney derived cells, such as Marc-145. The latter is currently used for vaccine production. However, since virus entry in Marc-145 cells is different compared to entry in primary macrophages, specific epitopes associated with virus entry could potentially alter upon growth on Marc-145 cells. To avoid this, we constructed CHO and PK15 cell lines recombinantly expressing the PRRSV receptors involved in virus entry into macrophages, sialoadhesin (Sn) and CD163 (CHOSn-CD163 and PK15Sn-CD163) and evaluated their potential for production of PRRSV. RESULTS: Detailed analysis of PRRSV infection revealed that LV and VR-2332 virus particles could attach to and internalize into the CHOSn-CD163 and PK15Sn-CD163 cells. Initially, this occurred less efficiently for macrophage grown virus than for Marc-145 grown virus. Upon internalization, disassembly of the virus particles was observed. The two cell lines could be infected with PRRSV strains LV and VR-2332. However, it was observed that Marc-145 grown virus infected the cells more efficiently than macrophage grown virus. If the cells were treated with neuraminidase to remove cis-acting sialic acids that hinder the interaction of the virus with Sn, the amount of infected cells with macrophage grown virus increased. Comparison of both cell lines showed that the PK15Sn-CD163 cell line gave in general better results than the CHOSn-CD163 cell line. Only 2 out of 5 PRRSV strains replicated well in CHOSn-CD163 cells. Furthermore, the virus titer of all 5 PRRSV strains produced after passaging in PK15Sn-CD163 cells was similar to the virus titer of those strains produced in Marc-145 cells. Analysis of the sequence of the structural proteins of original virus and virus grown for 5 passages on PK15Sn-CD163 cells showed either no amino acid (aa) changes (VR-2332 and 07V063), one aa (LV), two aa (08V194) or three aa (08V204) changes. None of these changes are situated in known neutralizing epitopes. CONCLUSIONS: A PRRSV susceptible cell line was constructed that can grow virus to similar levels compared to currently available cell lines. Mutations induced by growth on this cell lines were either absent or minimal and located outside known neutralizing epitopes. Together, the results show that this cell line can be used to produce vaccine virus and for PRRSV virus isolation.


Assuntos
Antígenos CD/genética , Antígenos de Diferenciação Mielomonocítica/genética , Linhagem Celular , Glicoproteínas de Membrana/genética , Vírus da Síndrome Respiratória e Reprodutiva Suína/fisiologia , Receptores de Superfície Celular/genética , Receptores Imunológicos/genética , Cultura de Vírus , Animais , Células CHO , Técnicas de Cultura de Células , Cricetinae , Cricetulus , Macrófagos/virologia , Lectina 1 Semelhante a Ig de Ligação ao Ácido Siálico , Suínos , Transfecção
17.
Arch Virol ; 155(3): 371-8, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20052599

RESUMO

Glycoprotein 4 (GP4) of porcine reproductive and respiratory syndrome virus (PRRSV) contains a highly variable neutralizing epitope. The present study aimed to investigate whether this epitope is susceptible to immunoselection by antibodies in vitro. Cultivation of PRRSV in vitro in the continuous presence of neutralizing monoclonal antibodies (mAbs) directed against this epitope resulted in the selection of mAb-resistant PRRSV strains within five passages. Comparison of the GP4 amino acid (aa) sequence of the original PRRSV strain with the GP4 aa sequences of the mAb-resistant PRRSV strains revealed aa substitutions within this epitope. In conclusion, this study shows that the neutralizing epitope on GP4 is susceptible to immunoselection by antibodies in vitro.


Assuntos
Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Epitopos de Linfócito B/imunologia , Vírus da Síndrome Respiratória e Reprodutiva Suína/imunologia , Seleção Genética , Proteínas Virais/imunologia , Sequência de Aminoácidos , Substituição de Aminoácidos/genética , Animais , Linhagem Celular , Análise Mutacional de DNA , Dados de Sequência Molecular , Mutação de Sentido Incorreto , Análise de Sequência de DNA , Inoculações Seriadas , Suínos
18.
J Virol ; 82(3): 1128-35, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18032516

RESUMO

Recently, Misinzo et al. (G. Misinzo, P. Meerts, M. Bublot, J. Mast, H. M. Weingartl, and H. J. Nauwynck, J. Gen. Virol. 86:2057-2068, 2005) reported that inhibiting endosome-lysosome system acidification reduced porcine circovirus 2 (PCV2) infection of monocytic 3D4/31 cells. The present study examined the effect of inhibiting endosome-lysosome system acidification in epithelial cells, since epithelial cells support PCV2 infection in vivo and are used in culturing PCV2 in vitro. Ammonium chloride (NH(4)Cl), chloroquine diphosphate (CQ), and monensin were used to inhibit endosome-lysosome system acidification. NH(4)Cl, CQ, or monensin increased PCV2 (Stoon-1010) infection by 726% +/- 110%, 1,212% +/- 34%, and 1,100% +/- 179%, respectively, in porcine kidney (PK-15) cells; by 128% +/- 7%, 158% +/- 3%, and 142% +/- 11% in swine kidney cells; by 160% +/- 28%, 446% +/- 50%, and 162% +/- 56% in swine testicle (ST) cells; and by 313% +/- 25%, 611% +/- 86%, and 352% +/- 44% in primary kidney epithelial cells. Similarly, increased PCV2 infection was observed with six other PCV2 strains in PK-15 cells treated with endosome-lysosome system acidification inhibitors. The mechanism behind increased PCV2 infection was further investigated in PK-15 cells using CQ. PCV2 infection of PK-15 cells was increased only when CQ was added early during PCV2 infection. CQ did not affect PCV2 virus-like particle (VLP) attachment to PK-15 cells but increased the disassembly of internalized PCV2 VLPs. In untreated PK-15 cells, internalized PCV2 VLPs localized within the endosome-lysosome system. PCV2 infection of untreated 3D4/31 and PK-15 cells and CQ-treated PK-15 cells was blocked by a serine protease inhibitor [4-(2-aminoethyl) benzenesulfonyl fluoride hydrochloride] but not by aspartyl protease (pepstatin A), cysteine protease (E-64), and metalloprotease (phosphoramidon) inhibitors. These results suggest that serine protease-mediated PCV2 disassembly is enhanced in porcine epithelial cells but inhibited in monocytic cells after inhibition of endosome-lysosome system acidification.


Assuntos
Circovirus/fisiologia , Endossomos/virologia , Células Epiteliais/virologia , Lisossomos/virologia , Cloreto de Amônio/farmacologia , Animais , Linhagem Celular , Cloroquina/análogos & derivados , Cloroquina/farmacologia , Endossomos/efeitos dos fármacos , Endossomos/metabolismo , Ionóforos/farmacologia , Lisossomos/efeitos dos fármacos , Lisossomos/metabolismo , Monensin/farmacologia , Serina Endopeptidases/metabolismo , Suínos , Ligação Viral/efeitos dos fármacos
19.
Arch Virol ; 154(12): 1939-43, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19885719

RESUMO

The porcine reproductive and respiratory syndrome virus (PRRSV) enters its target cell via clathrin-mediated endocytosis. Using dominant-negative Rab5 and Rab7 mutants, we show that upon internalization, PRRSV enters early endosomes but does not continue through the endocytic pathway to late endosomes. This was confirmed via colocalization experiments visualizing PRRSV and markers for different compartments of the endocytic pathway. Furthermore, it was shown that PRRSV colocalizes with its internalization receptor, sialoadhesin, on the cell surface and beneath the plasma membrane, while CD163 and PRRSV only meet in early endosomes.


Assuntos
Antígenos CD/metabolismo , Antígenos de Diferenciação Mielomonocítica/metabolismo , Endossomos/virologia , Vírus da Síndrome Respiratória e Reprodutiva Suína/patogenicidade , Receptores de Superfície Celular/metabolismo , Animais , Linhagem Celular , Endocitose , Endossomos/metabolismo , Humanos , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/virologia , Glicoproteínas de Membrana/metabolismo , Vírus da Síndrome Respiratória e Reprodutiva Suína/metabolismo , Receptores Imunológicos/metabolismo , Lectina 1 Semelhante a Ig de Ligação ao Ácido Siálico , Suínos , Fatores de Tempo
20.
Viruses ; 11(11)2019 11 06.
Artigo em Inglês | MEDLINE | ID: mdl-31698728

RESUMO

Respiratory Syncytial Virus (RSV) is a very important viral pathogen in children, immunocompromised and cardiopulmonary diseased patients and the elderly. Most of the published research with RSV was performed on RSV Long and RSV A2, isolated in 1956 and 1961, yet recent RSV isolates differ from these prototype strains. Additionally, these viruses have been serially passaged in cell culture, which may result in adaptations that affect virus-host interactions. We have isolated RSV from mucosal secretions of 12 patients in the winters 2016-2017 and 2017-2018, of which eight RSV-A subtypes and four RSV-B subtypes. Passage 3 of the isolates was assessed for viral replication kinetics and infectious virus production in HEp-2, A549 and BEAS-2B cells, thermal stability at 37 °C, 32 °C and 4 °C, syncytia formation, neutralization by palivizumab and mucin mRNA expression in infected A549 cells. We observed that viruses isolated in one RSV season show differences on the tested assays. Furthermore, comparison with RSV A2 and RSV B1 reveals for some RSV isolates differences in viral replication kinetics, thermal stability and fusion capacity. Major differences are, however, not observed and differences between the recent isolates and reference strains is, overall, similar to the observed variation in between the recent isolates. One clinical isolate (BE/ANT-A11/17) replicated very efficiently in all cell lines, and remarkably, even better than RSV A2 in the HEp-2 cell line.


Assuntos
Infecções por Vírus Respiratório Sincicial , Vírus Sincicial Respiratório Humano/isolamento & purificação , Células A549 , Bélgica/epidemiologia , Bronquiolite/virologia , Linhagem Celular , Criança , Pré-Escolar , Humanos , Mucinas/metabolismo , Infecções por Vírus Respiratório Sincicial/epidemiologia , Infecções por Vírus Respiratório Sincicial/virologia , Estações do Ano , Replicação Viral
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa