Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.160
Filtrar
1.
Allergy ; 78(4): 1060-1072, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36315052

RESUMO

BACKGROUND: Ligelizumab is an anti-IgE monoclonal antibody binding IgE with higher affinity than omalizumab that is under clinical investigation for several IgE-mediated diseases. We previously showed that omalizumab removes IgE bound to FcεRI on plasmacytoid dendritic cells (pDCs) and restores their ability to produce IFN-α and regulatory T cells (Tregs). The aim of this work is to investigate the capacity of ligelizumab to regulate functional properties of pDCs in comparison with omalizumab. METHODS: pDCs were isolated from atopic donors and IgE was detached from FcεRI on pDCs with designed ankyrin repeat protein (DARPin) bi53-79. pDCs were resensitized with IgE alone or in the presence of ligelizumab or omalizumab prior to IgE-FcεRI crosslinking and Toll-like receptor 9 (TLR9) stimulation. Flow cytometry, ELISA, coculture experiments and intranuclear staining were performed to determine cytokine production and Treg generation. An antigen-specific model of resensitization and IgE-crosslinking was also performed. RESULTS: The levels of serum total free IgE show a non-linear positive correlation with the frequency of IgE+ pDCs displaying IgE bound to FcεRI within the 43 individual donors included in the study. Ligelizumab displays stronger capacity than omalizumab to block the binding of free IgE to FcεRI on human pDCs, resulting in a greater restoration of TLR9-L-induced IFN-α production. Ligelizumab also restores the ability of pDCs to generate FOXP3+ Tregs as previously reported for omalizumab. CONCLUSIONS: The uncovered novel molecular mechanisms of ligelizumab to regulate functional properties of pDCs from atopic donors might have important clinical implications for anti-IgE treatments in different IgE-mediated diseases.


Assuntos
Hipersensibilidade Imediata , Omalizumab , Humanos , Células Dendríticas , Fatores de Transcrição Forkhead/metabolismo , Imunoglobulina E , Omalizumab/farmacologia , Omalizumab/uso terapêutico , Receptores de IgE/metabolismo , Linfócitos T Reguladores/metabolismo , Receptor Toll-Like 9/metabolismo , Interferon-alfa/biossíntese
2.
EBioMedicine ; 80: 104047, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35561451

RESUMO

BACKGROUND: Type I interferon (IFN-I) production by plasmacytoid dendritic cells (pDCs) occurs during viral infection, in response to Toll-like receptor 7 (TLR7) stimulation and is more vigorous in females than in males. Whether this sex bias persists in ageing people is currently unknown. In this study, we investigated the effect of sex and aging on IFN-α production induced by PRR agonist ligands. METHODS: In a large cohort of individuals from 19 to 97 years old, we measured the production of IFN-α and inflammatory cytokines in whole-blood upon stimulation with either R-848, ODN M362 CpG-C, or cGAMP, which activate the TLR7/8, TLR9 or STING pathways, respectively. We further characterized the cellular sources of IFN-α. FINDINGS: We observed a female predominance in IFN-α production by pDCs in response to TLR7 or TLR9 ligands. The higher TLR7-driven IFN-α production in females was robustly maintained across ages, including the elderly. The sex-bias in TLR9-driven interferon production was lost after age 60, which correlated with the decline in circulating pDCs. By contrast, STING-driven IFN-α production was similar in both sexes, preserved with aging, and correlated with circulating monocyte numbers. Indeed, monocytes were the primary cellular source of IFN-α in response to cGAMP. INTERPRETATION: We show that the sex bias in the TLR7-induced IFN-I production is strongly maintained through ages, and identify monocytes as the main source of IFN-I production via STING pathway. FUNDING: This work was supported by grants from Région Occitanie/Pyrénées-Méditerranée (#12052910, Inspire Program #1901175), University Paul Sabatier, and the European Regional Development Fund (MP0022856).


Assuntos
Interferon-alfa , Monócitos , Receptor 7 Toll-Like , Adulto , Idoso , Idoso de 80 Anos ou mais , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Feminino , Humanos , Interferon-alfa/biossíntese , Interferon-alfa/sangue , Interferon-alfa/imunologia , Ligantes , Masculino , Proteínas de Membrana/sangue , Proteínas de Membrana/imunologia , Pessoa de Meia-Idade , Monócitos/imunologia , Monócitos/metabolismo , Receptor Toll-Like 9/metabolismo , Adulto Jovem
3.
Front Immunol ; 12: 718744, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34531865

RESUMO

COVID-19 associated multisystem inflammatory syndrome (MIS) is a rare condition mostly affecting children but also adults (MIS-A). Although severe systemic inflammation and multiorgan dysfunction are hallmarks of the syndrome, the underlying pathogenesis is unclear. We aimed to provide novel immunological and genetic descriptions of MIS-A patients. Cytokine responses (IL-6, IL-1ß, TNFα, CXCL10, type I, II and III interferons) following SARS-CoV-2 infection of peripheral blood mononuclear cells in vitro were analyzed as well as antibodies against IFNα and IFNω (by ELISA) in patients and healthy controls. We also performed whole exome sequencing (WES) of patient DNA. A total of five patients (ages 19, 23, 33, 38, 50 years) were included. The patients shared characteristic features, although organ involvement and the time course of disease varied slightly. SARS-CoV-2 in vitro infection of patient PBMCs revealed impaired type I and III interferon responses and reduced CXCL10 expression, whereas production of proinflammatory cytokines were less affected, compared to healthy controls. Presence of interferon autoantibodies was not detected. Whole exome sequencing analysis of patient DNA revealed 12 rare potentially disease-causing variants in genes related to autophagy, classical Kawasaki disease, restriction factors and immune responses. In conclusion, we observed an impaired production of type I and III interferons in response to SARS-CoV-2 infection and detected several rare potentially disease-causing gene variants potentially contributing to MIS-A.


Assuntos
COVID-19/patologia , Citocinas/sangue , Interferon-alfa/biossíntese , Interferons/biossíntese , Síndrome de Resposta Inflamatória Sistêmica/patologia , Adulto , Autoanticorpos/sangue , Quimiocina CXCL10/biossíntese , Comorbidade , Exoma/genética , Feminino , Humanos , Interferon-alfa/imunologia , Interferons/imunologia , Leucócitos Mononucleares/imunologia , Masculino , Pessoa de Meia-Idade , SARS-CoV-2/imunologia , Sequenciamento do Exoma , Adulto Jovem , Interferon lambda
4.
J Gen Virol ; 102(7)2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34269676

RESUMO

Type I interferons (IFNs) are a first line of defence against viral infections. Upon infection, a first small wave of early type I IFN, mainly IFN-ß and particularly IFN-α4, are induced and bind to the type I IFN receptor (IFNAR) to amplify the IFN response. It was shown for several viruses that robust type I IFN responses require this positive feedback loop via the IFNAR. Recently, we showed that infection of IFNAR knockout mice with the orthomyxovirus Thogoto virus lacking the ML open reading frame (THOV(ML-)) results in the expression of unexpected high amounts of type I IFN. To investigate if IFNAR-independent IFN responses are unique for THOV(ML-), we performed infection experiments with several negative-strand RNA viruses using different routes and dosages for infection. A variety of these viruses induced type I IFN responses IFNAR-independently when using the intraperitoneal (i.p.) route for infection. In vitro studies demonstrated that myeloid dendritic cells (mDC) are capable of producing IFNAR-independent IFN-α responses that are dependent on the expression of the adaptor protein mitochondrial antiviral-signalling protein (MAVS) whereas pDC where entirely depending on the IFNAR feedback loop in vitro. Thus, depending on dose and route of infection, the IFNAR feedback loop is not strictly necessary for robust type I IFN expression and an IFNAR-independent type I IFN production might be the rule rather than the exception for infections with numerous negative-strand RNA viruses.


Assuntos
Interferon-alfa/biossíntese , Vírus de RNA de Sentido Negativo/imunologia , Infecções por Vírus de RNA/imunologia , Receptor de Interferon alfa e beta/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Células Dendríticas/imunologia , Células Dendríticas/virologia , Camundongos , Camundongos Endogâmicos C57BL , Células Mieloides/imunologia , Células Mieloides/virologia , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/virologia , Infecções por Vírus de RNA/virologia , Receptor de Interferon alfa e beta/genética , Thogotovirus , Carga Viral
5.
Front Immunol ; 12: 672729, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33995415

RESUMO

Plasmacytoid dendritic cells (pDCs) are a rare type of highly versatile immune cells that besides their specialized function of massive type I interferon (IFN-I) production are able to exert cytotoxic effector functions. However, diversification upon toll like receptor (TLR)-induced activation leads to highly heterogeneous responses that have not been fully characterized yet. Using droplet-based microfluidics, we showed that upon TLR7/8 and TLR9-induced single-cell activation only 1-3% secretes IFNα, and only small fractions upregulate cytotoxicity markers. Interestingly, this 1-3% of early IFN-producing pDCs, also known as first responders, express high levels of programmed death-ligand 1 (PD-L1) and TNF-related apoptosis-inducing ligand (TRAIL), which makes these hybrid cells similar to earlier described IFN-I producing killer pDCs (IKpDCs). IFN-I priming increases the numbers of IFNα producing cells up to 40%, but does not significantly upregulate the cytotoxicity markers. Besides, these so-called second responders do not show a cytotoxic phenotype as potent as observed for the first responders. Overall, our results indicate that the first responders are the key drivers orchestrating population wide IFN-I responses and possess high cytotoxic potential.


Assuntos
Citotoxicidade Imunológica/imunologia , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Interferon-alfa/biossíntese , Técnicas Analíticas Microfluídicas/métodos , Humanos , Fenótipo
6.
Vet Microbiol ; 253: 108958, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33387911

RESUMO

Porcine hemagglutinating encephalomyelitis virus (PHEV) is the cause of acute outbreaks of vomiting and wasting disease and/or encephalomyelitis in neonatal pigs, with naïve herds particularly vulnerable to clinical episodes. PHEV infections in older pigs are generally considered to be subclinical, but are poorly characterized in the refereed literature. In this study, twelve 7-week-old pigs were oronasally inoculated with 0.5 mL (1:128 HA titer) PHEV (Mengeling strain) and then followed through 42 days post inoculation (dpi). Fecal and oral fluid specimens were collected daily to evaluate viral shedding. Serum samples were tested for viremia, isotype-specific antibody responses, cytokine, and chemokine responses. Peripheral blood mononuclear cells were isolated to evaluate phenotype changes in immune cell subpopulations. No clinical signs were observed in PHEV inoculated pigs, but virus was detected in oral fluid (1-28 dpi) and feces (1-10 dpi). No viremia was detected, but a significant IFN-α response was observed in serum at 3 dpi, followed by the detection of IgM (dpi 7), and IgA/IgG (dpi 10). Flow cytometry revealed a one-off increase in cytotoxic T cells at 21 dpi. This study demonstrated that exposure of grower pigs to PHEV results in subclinical infection characterized by active viral replication and shedding followed by an active humoral and cell-mediated immune response that attenuates the course of the infection and results in viral clearance.


Assuntos
Betacoronavirus 1/isolamento & purificação , Infecções por Coronavirus/veterinária , Doenças dos Suínos/virologia , Animais , Anticorpos Antivirais/sangue , Infecções por Coronavirus/imunologia , Infecções por Coronavirus/virologia , Interferon-alfa/biossíntese , Interferon-alfa/sangue , Suínos , Doenças dos Suínos/sangue , Doenças dos Suínos/imunologia , Viremia
7.
Bioorg Med Chem ; 33: 116037, 2021 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-33515919

RESUMO

The conventional use of E. coli system for protein expression is limited to non-glycosylated proteins. While yeast, insect and mammalian systems are available to produce heterologous glycoproteins, developing an engineered E. coli-based glycosylation platform will provide a faster, more economical, and more convenient alternative. In this work, we present a two-step approach for production of a homogeneously glycosylated eukaryotic protein using the E. coli expression system. Human interferon α-2b (IFNα) is used as a model protein to illustrate this glycosylation scheme. In the first step, the N-glycosyltransferase from Actinobacillus pleuropneumoniae (ApNGT) is co-expressed for in vivo transfer of a glucose residue to IFNα at an NX(S/T) N-glycosylation sequon. Several E. coli systems were examined to evaluate the efficiency of IFNα N-glucosylation. In the second step, the N-glucosylated protein is efficiently elaborated with biantennary sialylated complex-type N-glycan using an in vitro chemoenzymatic method. The N-glycosylated IFNα product was found to be biologically active and displayed significantly improved proteolytic stability. This work presents a feasible E. coli-based glycosylation machinery for producing therapeutic eukaryotic glycoproteins.


Assuntos
Escherichia coli/metabolismo , Interferon-alfa/biossíntese , Actinobacillus pleuropneumoniae/enzimologia , Glucose/química , Glucose/metabolismo , Glucosiltransferases/metabolismo , Glicosilação , Interferon-alfa/química , Interferon-alfa/isolamento & purificação
8.
Bioprocess Biosyst Eng ; 44(3): 473-482, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33051705

RESUMO

Interferon α (IFN-α) plays a crucial role in the host's immune response. In this study, the amino acid sequence of porcine interferon α (PoIFN-α) was analyzed. Seven substitutions, S38F, H40Q, F43L, N78D, Y86C, S151A, and R156T, were mutated and obtained by aligning the sequences of PoIFN-α subtypes. The PoIFN-α mutants were designed, expressed, and purified in E. coli. The antiviral activities of these PoIFN-αs were measured in Vero and swine testis cells against vesicular stomatitis virus (VSV). Their inhibitory abilities on pseudorabies virus (PRV) were also examined. Commercial PoIFN-α was used as a control. We found the ideal inducer concentration of isopropyl ß-D-thiogalactoside was 1 mM, and the best time-point for induction was 8 h. The PoIFN-α mutant named PoIFN-α-156s had the highest antiviral activity, which was about 200-fold more than that of PoIFN-α. PoIFN-α-156s could inhibit VSV and PRV replication in a dose-dependent manner in vitro. The half-life of PoIFN-α-156s was longer than that of PoIFN-α in mice, and the effective antiviral action was higher than PoIFN-α. Animal experiments showed that PoIFN-α-156s could decrease the viral load after infection with VSV. Overall, these results suggest that recombinant PoIFN-α-156s has the ability of antivirus, and is feasible for veterinary clinical applications and fundamental research.


Assuntos
Substituição de Aminoácidos , Escherichia coli , Interferon-alfa , Mutação de Sentido Incorreto , Animais , Chlorocebus aethiops , Escherichia coli/genética , Escherichia coli/metabolismo , Interferon-alfa/biossíntese , Interferon-alfa/química , Interferon-alfa/genética , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Suínos , Células Vero
9.
PLoS One ; 15(11): e0240964, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33166307

RESUMO

BACKGROUND: Allergic asthma is a heterogenous disorder predominantly driven by a type 2 inflammatory response to aeroallergens. Therapeutic modulation to rebalance these type 2 responses may offer clinical benefit for allergic respiratory inflammatory diseases, with the potential for disease modification. GSK2245035, a selective toll-like receptor-7 agonist, preferentially stimulates the induction of type 1 interferon alpha, reducing type 2 responses. OBJECTIVE: This study investigated whether intranasal GSK2245035 reduced allergen-induced bronchial reactivity in mild allergic asthma. METHODS: This double-blind, placebo-controlled, parallel-group Phase IIa trial randomized (1:1) participants with mild allergic asthma to intranasal GSK2245035 20 ng or placebo once weekly for 8 weeks; follow-up was conducted 1, 4, and 12 weeks after treatment. Allergen-induced late asthmatic response 1 week after treatment was measured as minimum and weighted mean forced expiratory volume in 1 second (FEV1) 4-10 hours following bronchial allergen challenge (primary endpoint). Pharmacodynamic and allergic biomarkers, and adverse events, were assessed. A Bayesian analysis framework was used; a posterior probability >0.7 denoted primary endpoint success. RESULTS: Thirty-six participants were randomized (GSK2245035, n = 22; placebo, n = 14). The percentage attenuation in late asthmatic response was -4.6% (posterior probability: 0.385) and -10.5% (posterior probability: 0.303) for minimum and weighted mean FEV1, respectively. Type 2 responses were confirmed by changes in lung function, eosinophils (blood and sputum), interleukin-5 (sputum) and fractional exhaled nitric oxide biomarkers pre- and post-bronchial allergen challenge. However, no treatment effect was observed. Adverse events were reported by 10/14 (71%) and 21/22 (95%) participants in the placebo and GSK2245035 groups, respectively; headache was the most common. CONCLUSIONS AND CLINICAL RELEVANCE: Although target engagement was observed, weekly intranasal GSK2245035 20 ng for 8 weeks did not substantially attenuate the late asthmatic response in participants with mild allergic asthma. Overall, treatment was well tolerated.


Assuntos
Adenina/análogos & derivados , Asma/tratamento farmacológico , Piperidinas/administração & dosagem , Receptor 7 Toll-Like/agonistas , Adenina/administração & dosagem , Adenina/efeitos adversos , Administração Intranasal , Adulto , Alérgenos/administração & dosagem , Antiasmáticos/administração & dosagem , Antiasmáticos/efeitos adversos , Asma/imunologia , Asma/fisiopatologia , Testes de Provocação Brônquica , Método Duplo-Cego , Feminino , Volume Expiratório Forçado/efeitos dos fármacos , Humanos , Interferon-alfa/biossíntese , Masculino , Pessoa de Meia-Idade , Piperidinas/efeitos adversos , Estudo de Prova de Conceito , Rinite Alérgica/tratamento farmacológico , Rinite Alérgica/imunologia , Rinite Alérgica/fisiopatologia , Adulto Jovem
10.
J Neuroinflammation ; 17(1): 340, 2020 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-33183319

RESUMO

BACKGROUND: Interferon α (IFNα) is a cytokine whose production is increased endogenously in response to viral infection and in autoimmune diseases such as systemic lupus erythematosus (SLE). An elevated IFNα signature has been associated with clinically observed neuro-behavioural deficits such as mild cognitive impairment, fatigue, depression and psychosis in these diseases. However, the mechanisms underlying these neuropsychiatric symptoms remain largely unknown, and it is as yet unclear how IFNα signalling might influence central nervous system (CNS) function. Aberrant microglia-mediated synaptic pruning and function has recently been implicated in several neurodegenerative and neuropsychiatric diseases, but whether and how IFNα modulates these functions are not well defined. METHODS: Using a model of peripheral IFNα administration, we investigated gene expression changes due to IFNAR signalling in microglia. Bulk RNA sequencing on sorted microglia from wild type and microglia-specific Ifnar1 conditional knockout mice was performed to evaluate IFNα and IFNAR signalling-dependent changes in gene expression. Furthermore, the effects of IFNα on microglia morphology and synapse engulfment were assessed, via immunohistochemistry and flow cytometry. RESULTS: We found that IFNα exposure through the periphery induces a unique gene signature in microglia that includes the expected upregulation of multiple interferon-stimulated genes (ISGs), as well as the complement component C4b. We additionally characterized several IFNα-dependent changes in microglial phenotype, including expression of CD45 and CD68, cellular morphology and presynaptic engulfment, that reveal subtle brain region-specific differences. Finally, by specifically knocking down expression of IFNAR1 on microglia, we show that these changes are largely attributable to direct IFNAR signalling on microglia and not from indirect signalling effects through other CNS parenchymal cell types which are capable of IFNα-IFNAR signal transduction. CONCLUSIONS: Peripheral IFNα induces unique genetic and phenotypic changes in microglia that are largely dependent on direct signalling through microglial IFNAR. The IFNα-induced upregulation of C4b could play important roles in the context of aberrant synaptic pruning in neuropsychiatric disease.


Assuntos
Interferon-alfa/biossíntese , Microglia/metabolismo , Receptor de Interferon alfa e beta/biossíntese , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Feminino , Interferon-alfa/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microglia/patologia , Receptor de Interferon alfa e beta/deficiência , Receptor de Interferon alfa e beta/genética , Baço/metabolismo , Baço/patologia
11.
Cell Immunol ; 357: 104201, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32979763

RESUMO

The DNA binding protein AT-rich interacting domain 3a (ARID3a)2 is expressed in healthy human hematopoietic cord blood progenitors where its modulation influences myeloid versus B lineage development. ARID3a is also variably expressed in subsets of adult peripheral blood hematopoietic progenitors where the consequences of ARID3a expression are unknown. In B lymphocytes, Toll-like receptor (TLR)3 signaling induces ARID3a expression in association with Type I interferon inflammatory cytokines. We hypothesized that TLR ligand stimulation of peripheral blood hematopoietic progenitors would induce ARID3a expression resulting in interferon production, and potentially influencing lineage decisions. Our data revealed that the TLR9 agonist CpG induces ARID3a expression with interferon alpha synthesis in human hematopoietic progenitors. However, ARID3a expression was not associated with increased B lineage development. These results demonstrate the need for further experiments to better define how pathogen-associated responses influence hematopoiesis.


Assuntos
Proteínas de Ligação a DNA/biossíntese , Células-Tronco Hematopoéticas/metabolismo , Interferon-alfa/biossíntese , Receptores Toll-Like/sangue , Fatores de Transcrição/biossíntese , Adulto , Linfócitos B/citologia , Linfócitos B/metabolismo , Ilhas de CpG , Citocinas/metabolismo , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/sangue , Feminino , Citometria de Fluxo/métodos , Expressão Gênica , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Humanos , Interferon-alfa/genética , Interferon-alfa/imunologia , Ligantes , Pessoa de Meia-Idade , Transdução de Sinais , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/sangue
12.
Immunohorizons ; 4(6): 332-338, 2020 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-32554437

RESUMO

Several human autoimmune diseases are characterized by increased expression of type 1 IFN-stimulated genes in both the peripheral blood and tissue. The contributions of different type I IFNs to this gene signature are uncertain as the type I IFN family consists of 13 alphas and one each of ß, ε, κ, and ω subtypes. We sought to investigate the contribution of various IFNs to IFN signaling in primary human cell types. We stimulated primary skin, muscle, kidney, and PBMCs from normal healthy human donors with various TLR ligands and measured the expression of type I IFN subtypes and activation of downstream signaling by quantitative PCR. We show that IFNB1 is the dominant type I IFN expressed upon TLR3 and TLR4 stimulation, and its expression profile is associated with subsequent MX1 transcription. Furthermore, using an IFN-ß-specific neutralizing Ab, we show that MX1 expression is inhibited in a dose-dependent manner, suggesting that IFN-ß is the primary driver of IFN-stimulated genes following TLR3 and TLR4 engagement. Stimulation with TLR7/8 and TLR9 ligands induced IFNB1 and IFNA subtypes and MX1 expression only in PBMCs and not in tissue resident cell types. Concordantly, IFN-ß neutralization had no effect on MX1 expression in PBMCs potentially because of the combination of IFNB1 and IFNA expression. Combined, these data highlight the potential role for IFN-ß in driving local inflammatory responses in clinically relevant human tissue types and opportunities to treat local inflammation by targeting IFN-ß.


Assuntos
Interferon-alfa/biossíntese , Interferon beta/biossíntese , Proteínas de Resistência a Myxovirus/metabolismo , Células Cultivadas , Voluntários Saudáveis , Humanos , Interferon-alfa/genética , Interferon beta/genética , Ligantes , Proteínas de Resistência a Myxovirus/genética , Transdução de Sinais/imunologia , Receptor 3 Toll-Like , Receptor 4 Toll-Like , Receptor 7 Toll-Like , Receptor 8 Toll-Like , Receptor Toll-Like 9
13.
Sheng Wu Gong Cheng Xue Bao ; 36(4): 750-762, 2020 Apr 25.
Artigo em Chinês | MEDLINE | ID: mdl-32347069

RESUMO

PEGylation is considered one of the most successful techniques to improve the characteristics of protein drugs including to increase the circulating half-life of proteins in blood and to decrease their immunogenicity and antigenicity. One known PEG modification method is to attach PEG to the free amino group, typically at lysine residues or at the N-terminal amino acid with no selectivity, resulting in a heterogeneous product mixture. This lack of selectivity can present problems when a therapeutic PEGylated protein is being developed, because predictability of activity and manufacturing reproducibility are needed for regulatory approval. Enzymatic PEGylation of proteins is one route to overcome this limitation. Transglutaminases (TGase) are enzyme candidates for site-specific PEGylation. We use human interferon alpha 2a (IFN α2a) as a test case, and predict that the potential modification residues are Gln101 by computational approach as it contains 12 potential PEGylation sites. IFN α2a was PEGylated by Y shaped PEG40k-NH2 mediated by microbial transglutaminase. Our results show that the microbial transglutaminase mediated PEGylation of IFN α2a was site-specific only at the site of Gln101 in IFN α2a, yielding the single mono-conjugate PEG-Gln101-IFN α2a with a mass of 59 374.66 Da. Circular dichroism studies showed that PEG-Gln101-IFN α2a preserved the same secondary structures as native IFN α2a. As expected, the bioactivity and pharmacokinetic profile in rats of PEG-Gln101-IFN α2a revealed a significant improvement to unmodified IFN α2a, and better than PEGASYS.


Assuntos
Antivirais , Interferon-alfa , Polietilenoglicóis , Transglutaminases , Animais , Humanos , Interferon alfa-2/metabolismo , Interferon-alfa/biossíntese , Interferon-alfa/farmacocinética , Polietilenoglicóis/farmacocinética , Estrutura Secundária de Proteína , Ratos , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/farmacocinética , Proteínas Recombinantes/farmacologia , Reprodutibilidade dos Testes , Transglutaminases/metabolismo
14.
Front Immunol ; 11: 518, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32296431

RESUMO

Little is known about how tuberculosis (TB) impairs dendritic cell (DC) function and anti-TB immune responses. We previously showed that the B and T lymphocyte attenuator (BTLA), an immune inhibitory receptor, is involved in TB pathogenesis. Here, we examined whether BTLA expression in TB affects phenotypic and functional aspects of DCs. Active TB patients exhibited higher expression of BTLA in myeloid dendritic cells (mDCs) and plasmacytoid DCs (pDCs) subsets compared with healthy controls (HCs). BTLA expression was similarly high in untreated TB, TB relapse, and sputum-bacillus positive TB, but anti-TB therapy reduced TB-driven increases in frequencies of BTLA+ DCs. BTLA+ DCs in active TB showed decreased expression of the DC maturation marker CD83, with an increased expression of CCR7 in mDCs. BTLA+ DCs in active TB displayed a decreased ability to express HLA-DR and to uptake foreign antigen, with a reduced expression of the co-stimulatory molecule CD80, but not CD86. Functionally, BTLA+ DCs in active TB showed a decreased production of IL-12 and IFN-α as well as a reduced ability to stimulate allogeneic T-cell proliferative responses. BTLA+ mDCs produced larger amounts of IL-4 and TGF-ß than BTLA- mDCs in both HCs and APT patients. BTLA+ DCs from active TB patients showed a reduced ability to stimulate Mtb antigen-driven Th17 and Th22 polarizations as compared to those from HCs. Conversely, these BTLA+ DCs more readily promoted the differentiation of T regulatory cells (Treg) and Th2 than those from HCs. These findings suggest that TB-driven BTLA expression in DCs impairs the expression of functional DC surrogate markers and suppress the ability of DCs to induce anti-TB Th17 and Th22 response while promoting Th2 and Foxp3+ Tregs.


Assuntos
Células Dendríticas/imunologia , Receptores Imunológicos/imunologia , Linfócitos T Reguladores/imunologia , Células Th2/imunologia , Tuberculose Pulmonar/imunologia , Adolescente , Adulto , Idoso , Diferenciação Celular/imunologia , Feminino , Humanos , Interferon-alfa/biossíntese , Interleucina-12/biossíntese , Interleucina-4/biossíntese , Ativação Linfocitária/imunologia , Masculino , Pessoa de Meia-Idade , Fator de Crescimento Transformador beta/biossíntese , Adulto Jovem
15.
Sci Rep ; 10(1): 6740, 2020 04 21.
Artigo em Inglês | MEDLINE | ID: mdl-32317718

RESUMO

To protect the audiosensory organ from tissue damage from the immune system, the inner ear is separated from the circulating immune system by the blood-labyrinth barrier, which was previously considered an immune-privileged site. Recent studies have shown that macrophages are distributed in the cochlea, especially in the spiral ligament, spiral ganglion, and stria vascularis; however, the direct pathogen defence mechanism used by audiosensory receptor hair cells (HCs) has remained obscure. Here, we show that HCs are protected from pathogens by surrounding accessory supporting cells (SCs) and greater epithelial ridge (GER or Kölliker's organ) cells (GERCs). In isolated murine cochlear sensory epithelium, we established Theiler's murine encephalomyelitis virus, which infected the SCs and GERCs, but very few HCs. The virus-infected SCs produced interferon (IFN)-α/ß, and the viruses efficiently infected the HCs in the IFN-α/ß receptor-null sensory epithelium. Interestingly, the virus-infected SCs and GERCs expressed macrophage marker proteins and were eliminated from the cell layer by cell detachment. Moreover, lipopolysaccharide induced phagocytosis of the SCs without cell detachment, and the SCs phagocytosed the bacteria. These results reveal that SCs function as macrophage-like cells, protect adjacent HCs from pathogens, and provide a novel anti-infection inner ear immune system.


Assuntos
Células Ciliadas Auditivas Internas/fisiologia , Células Ciliadas Auditivas Externas/fisiologia , Células Labirínticas de Suporte/imunologia , Macrófagos/imunologia , Gânglio Espiral da Cóclea/fisiologia , Estria Vascular/fisiologia , Animais , Animais Recém-Nascidos , Escherichia coli/imunologia , Células Ciliadas Auditivas Internas/citologia , Células Ciliadas Auditivas Externas/citologia , Imunidade Inata , Interferon-alfa/biossíntese , Interferon-alfa/imunologia , Interferon beta/biossíntese , Interferon beta/imunologia , Células Labirínticas de Suporte/citologia , Células Labirínticas de Suporte/efeitos dos fármacos , Células Labirínticas de Suporte/virologia , Lipopolissacarídeos/farmacologia , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Macrófagos/virologia , Camundongos , Camundongos Endogâmicos ICR , Técnicas de Cultura de Órgãos , Fagocitose/efeitos dos fármacos , Saccharomyces cerevisiae/imunologia , Gânglio Espiral da Cóclea/citologia , Estria Vascular/citologia , Theilovirus/crescimento & desenvolvimento , Theilovirus/patogenicidade
16.
Rheumatology (Oxford) ; 59(10): 2992-3002, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-32160289

RESUMO

OBJECTIVE: Increased IFNα is important in the pathogenesis of SLE. Plasmacytoid dendritic cells are considered the main producer of IFNα upon Toll-like receptor pathway activation. However, which cells produce IFNα following stimulation with cyclic GMP-AMP synthase (cGAS) and stimulator of IFN genes (STING) in SLE remains unknown. We investigated the IFNα producing capacity of myeloid cells under cGAS-STING pathway stimulation. METHODS: IFNα levels in peripheral blood mononuclear cells from SLE patients and healthy controls stimulated with 2'3'c-GAMP, a stimulator of cGAS-STING, were measured by intracellular cytokine staining and flow cytometry. STING expression and its co-localization with TBK1 were examined by flow cytometry or confocal microscopy. The effects of in vitro exposure to IFNα on IFNα production and STING expression, and in vitro rapamycin treatment on IFNα production and STING, pTBK1 and IRF3 expression were examined. RESULTS: IFNα was produced by monocytes, conventional dendritic cells and plasmacytoid dendritic cells upon cGAS-STING pathway activation. The frequency of IFNα-producing monocytes positively correlated with SLE disease activity. STING expression and its co-localization with TBK1 were increased in lupus monocytes. Prior exposure to IFNα enhanced the IFNα-producing capacity of monocytes. Inhibition of the mechanistic target of the rapamycin (mTOR) pathway suppressed IFNα production from monocytes and downregulated enhanced STING expression and its downstream molecules. CONCLUSION: Enhanced IFNα from lupus monocytes induced by augmented STING pathway activation is associated with SLE pathogenesis. Suppression of the mTOR pathway downregulated the enhanced STING expression and the subsequent IFNα production by monocytes.


Assuntos
Interferon-alfa/biossíntese , Lúpus Eritematoso Sistêmico/metabolismo , Proteínas de Membrana/biossíntese , Monócitos/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Serina-Treonina Quinases TOR/antagonistas & inibidores , Adulto , Estudos de Casos e Controles , Células Dendríticas/metabolismo , Regulação para Baixo , Feminino , Citometria de Fluxo , Humanos , Imunossupressores/farmacologia , Interferon-alfa/farmacologia , Masculino , Microscopia Confocal , Pessoa de Meia-Idade , Monócitos/efeitos dos fármacos , Nucleotídeos Cíclicos/farmacologia , Sirolimo/farmacologia , Adulto Jovem
17.
Int J Mol Sci ; 21(4)2020 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-32098104

RESUMO

Nervous necrosis virus (NNV) results in high mortality rates of infected marine fish worldwide. Interferons (IFNs) are cytokines in vertebrates that suppress viral replication and regulate immune responses. Heterologous overexpression of fish IFN in bacteria could be problematic because of protein solubility and loss of function due to protein misfolding. In this study, a protein model of the IFN-α of Epinephelus septemfasciatus was built based on comparative modeling. In addition, PelB and SacB signal peptides were fused to the N-terminus of E. septemfasciatus IFN-α for overexpression of soluble, secreted IFN in Escherichia coli (E-IFN) and Bacillus subtilis (B-IFN). Cytotoxicity tests indicated that neither recombinant grouper IFN-α were cytotoxic to a grouper head kidney cell line (GK). The GK cells stimulated with E-IFN and B-IFN exhibited elevated expression of antiviral Mx genes when compared with the control group. The NNV challenge experiments demonstrated that GK cells pretreated or co-treated with E-IFN and B-IFN individually had three times the cell survival rates of untreated cells, indicating the cytoprotective ability of our recombinant IFNs. These data provide a protocol for the production of soluble, secreted, and functional grouper IFN of high purity, which may be applied to aquaculture fisheries for antiviral infection.


Assuntos
Bacillus subtilis , Escherichia coli , Proteínas de Peixes , Interferon-alfa , Perciformes/genética , Animais , Bacillus subtilis/genética , Bacillus subtilis/metabolismo , Linhagem Celular , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas de Peixes/biossíntese , Proteínas de Peixes/genética , Proteínas de Peixes/farmacologia , Interferon-alfa/biossíntese , Interferon-alfa/genética , Interferon-alfa/farmacologia , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética
18.
Front Immunol ; 11: 620283, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33708194

RESUMO

Apolipoprotein A-I mimetic peptides are amphipathic alpha-helix peptides that display similar functions to apolipoprotein A-I. Preclinical and clinical studies have demonstrated the safety and efficacy of apolipoprotein A-I mimetic peptides in multiple indications associated with inflammatory processes. In this study, we evaluated the effect of the long-term expression of L37pA in the liver by an adeno-associated virus (AAV-L37pA) on the expression of an adeno-associated virus encoding interferon-alpha (AAV-IFNα). Long-term IFNα expression in the liver leads to lethal hematological toxicity one month after AAV administration. Concomitant administration of AAV-L37pA prevented the lethal toxicity since the IFNα expression was reduced one month after AAV administration. To identify the mechanism of action of L37pA, a genomic and proteomic analysis was performed 15 days after AAV administration when a similar level of IFNα and interferon-stimulated genes were observed in mice treated with AAV-IFNα alone and in mice treated with AAV-IFNα and AAV-L37pA. The coexpression of the apolipoprotein A-I mimetic peptide L37pA with IFNα modulated the gene expression program of IFNα, inducing a significant reduction in inflammatory pathways affecting pathogen-associated molecular patterns receptor, dendritic cells, NK cells and Th1 immune response. The proteomic analysis confirmed the impact of the L37pA activity on several inflammatory pathways and indicated an activation of LXR/RXR and PPPARα/γ nuclear receptors. Thus, long-term expression of L37pA induces an anti-inflammatory effect in the liver that allows silencing of IFNα expression mediated by an adeno-associated virus.


Assuntos
Antivirais/farmacologia , Apolipoproteína A-I/agonistas , Inflamação/prevenção & controle , Interferon-alfa/toxicidade , Fígado/metabolismo , Pancitopenia/prevenção & controle , Animais , Dependovirus/genética , Feminino , Regulação Viral da Expressão Gênica , Inativação Gênica , Vetores Genéticos/genética , Inflamação/etiologia , Interferon-alfa/biossíntese , Interferon-alfa/sangue , Interferon-alfa/genética , Lipoproteínas/sangue , Fígado/patologia , Receptores X do Fígado/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , PPAR alfa/metabolismo , PPAR gama/metabolismo , Pancitopenia/etiologia , Proteoma , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Proteínas Recombinantes/toxicidade , Receptores X de Retinoides/metabolismo , Organismos Livres de Patógenos Específicos , Transgenes
19.
Nucleosides Nucleotides Nucleic Acids ; 39(1-3): 292-309, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31509065

RESUMO

Double-stranded RNAs consisting of 21-nucleotide passenger and guide strands, known as small interfering RNAs (siRNAs), can be used for the identification of gene functions and the regulation of genes involved in disease for therapeutics. The difficulty with unmodified siRNAs lies in the chemical synthesis of RNA, its degradation by RNase, the immune response derived from natural RNA, and the off-target effects mediated by the passenger strand. In this study, asymmetrical 18 base-paired double-strand oligonucleotides comprised of alternately combined DNAs and 2'-O-methyl RNAs, denoted as MED-siRNA, were evaluated. These modified oligonucleotides showed high RNase resistance, a reduced immune response, a highly efficient cleavage of target mRNA with binding to Argonaute 2 (Ago2) via RNA interference, and the subsequent reduction of target protein expression. These findings suggest the possibility of alternatives to unmodified siRNAs with potential use in therapeutics.


Assuntos
DNA/química , Oligonucleotídeos/química , RNA de Cadeia Dupla/química , Linhagem Celular Tumoral , Técnicas de Química Sintética , DNA/síntese química , Inativação Gênica , Humanos , Interferon-alfa/biossíntese , Leucócitos Mononucleares/metabolismo , Conformação de Ácido Nucleico , Oligonucleotídeos/síntese química , Clivagem do RNA , Interferência de RNA , RNA de Cadeia Dupla/síntese química , RNA Mensageiro/genética , Relação Estrutura-Atividade , Transfecção
20.
Cell Mol Immunol ; 17(5): 519-532, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31853001

RESUMO

Allergic asthma, a chronic inflammatory airway disease associated with type 2 cytokines, often originates in early life. Immune responses at an early age exhibit a Th2 cell bias, but the precise mechanisms remain elusive. Plasmacytoid dendritic cells (pDCs), which play a regulatory role in allergic asthma, were shown to be deficient in neonatal mice. We report here that this pDC deficiency renders neonatal mice more susceptible to severe allergic airway inflammation than adult mice in an OVA-induced experimental asthma model. Adoptive transfer of pDCs or administration of IFN-α to neonatal mice prevented the development of allergic inflammation in wild type but not in IFNAR1-/- mice. Similarly, adult mice developed more severe allergic inflammation when pDCs were depleted. The protective effects of pDCs were mediated by the pDC-/IFN-α-mediated negative regulation of the secretion of epithelial cell-derived CCL20, GM-CSF, and IL-33, which in turn impaired the recruitment of cDC2 and ILC2 cells to the airway. In asthmatic patients, the percentage of pDCs and the level of IFN-α were lower in children than in adults. These results indicate that impairment of pDC-epithelial cell crosstalk in neonates is a susceptibility factor for the development of allergen-induced allergic airway inflammation.


Assuntos
Células Dendríticas/metabolismo , Hipersensibilidade/patologia , Inflamação/patologia , Interferon-alfa/biossíntese , Pulmão/patologia , Transferência Adotiva , Animais , Animais Recém-Nascidos , Asma/imunologia , Asma/patologia , Brônquios/patologia , Líquido da Lavagem Broncoalveolar , Contagem de Células , Citocinas/metabolismo , Células Epiteliais/metabolismo , Feminino , Humanos , Hipersensibilidade/imunologia , Inflamação/imunologia , Mediadores da Inflamação/metabolismo , Pulmão/imunologia , Linfócitos/imunologia , Proteínas de Membrana/metabolismo , Camundongos Endogâmicos C57BL , Receptor de Interferon alfa e beta/metabolismo , Escarro/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA