Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Int Immunopharmacol ; 89(Pt B): 107099, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33091819

RESUMO

Combinatory enhancement of innate and adaptive immune responses is a promising strategy in immunotherapeutic drug development. Bifunctional macromolecules that simultaneously target two mechanisms may provide additional advantages over the combination of targeting two single pathways. Interferon alpha (IFNα) has been used clinically against viral infection such as the chronic infection of hepatitis B virus (CHB) as well as some types of cancers. OX40 is a costimulatory immune checkpoint molecule involved in the activation of T lymphocytes. To test whether simultaneously activating IFNα and OX40 signaling pathway could produce a synergistic therapeutic effect on CHB and tumors, we designed a bifunctional fusion protein composed of a mouse OX40 agonistic monoclonal antibody (OX86) and a mouse IFNα4, joined by a flexible (GGGGS)3 linker. This fusion protein, termed OX86-IFN, can activate both IFNα and OX40. We demonstrated that OX86-IFN could effectively activate T lymphocytes in the peripheral blood of mice. Furthermore, we showed that OX86-IFN had superior therapeutic effect to monotherapies in HBV hydrodynamic transfection and syngeneic tumor models. Collectively, our data suggests that simultaneously targeting interferon and OX40 signaling pathways by bifunctional molecule OX86-IFN elicits potent antiviral and antitumor activities, which could provide a new strategy in developing therapeutic agents against viral infection and tumors.


Assuntos
Antineoplásicos Imunológicos/farmacologia , Antivirais/farmacologia , Hepatite B Crônica/tratamento farmacológico , Interferon-alfa/agonistas , Neoplasias/tratamento farmacológico , Receptores OX40/agonistas , Proteínas Recombinantes de Fusão/farmacologia , Imunidade Adaptativa/efeitos dos fármacos , Animais , Antineoplásicos Imunológicos/administração & dosagem , Antivirais/administração & dosagem , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/metabolismo , Linhagem Celular Tumoral , Modelos Animais de Doenças , Vírus da Hepatite B/efeitos dos fármacos , Imunidade Inata/efeitos dos fármacos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Terapia de Alvo Molecular/métodos , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Transplante de Neoplasias , Receptor de Interferon alfa e beta/agonistas , Proteínas Recombinantes de Fusão/administração & dosagem
2.
Antiviral Res ; 161: 163-168, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30521835

RESUMO

Arboviruses represent a group of pathogens that can spread efficiently throughout human populations by hematophagous arthropod vectors. The mosquito-borne (re)emerging Chikungunya and Dengue viruses belong to the alphavirus and flavivirus genus, respectively, with no approved therapeutics or safe vaccines for humans. Transmitted by the same vector Aedes spp., these viruses cause significant morbidity and mortality in endemic areas. Due to the increasing likelihood of co-circulation and co-infection with viruses, we aimed to identify a pharmacologically targetable host factor that can inhibit multiple viruses and show that a potent antagonist of prolyl tRNA synthetase (halofuginone) suppresses both Chikungunya and Dengue viruses. Host tRNA synthetase inhibition may signify an additional approach to combat present and future epidemic pathogens.


Assuntos
Aedes/enzimologia , Aminoacil-tRNA Sintetases/antagonistas & inibidores , Antivirais/farmacologia , Vírus Chikungunya/efeitos dos fármacos , Vírus da Dengue/efeitos dos fármacos , Piperidinas/farmacologia , Quinazolinonas/farmacologia , Aedes/virologia , Animais , Células Cultivadas , Febre de Chikungunya/virologia , Dengue/virologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/virologia , Prepúcio do Pênis/citologia , Interações entre Hospedeiro e Microrganismos , Humanos , Proteínas de Insetos/antagonistas & inibidores , Masculino , Mosquitos Vetores , Receptor de Interferon alfa e beta/agonistas
3.
Hum Antibodies ; 26(1): 17-22, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28282804

RESUMO

BACKGROUND: Multiple sclerosis (MS) as an auto-immune disease is an inflammatory, demyelinating disease of the central nervous system. Certain genes have shown to be involved in the initiation of MS but the specific role of some of them, e.g. IFNAR1 has not been identified in certain populations yet. OBJECTIVE: The IFNAR1 as a type I membrane protein shapes one of the two chains of a receptor for interferons alpha and beta. METHODS: To find out how IFNAR1 functions in the Iranian population, the researchers compared the expression level of this gene in relapsing-remitting MS (RR-MS) samples with normal individuals. RNA from the whole blood of 50 RR-MS patients and 50 normal controls were extracted. All patients were HLA-DRB1*15 negative and were responders to interferon-beta with a normal vitamin D level. The level of IFNAR1 gene expression was measured using quantitative RT-PCR. RESULTS: According to the results the RR-MS patients manifested a statistically higher expression level of IFNAR1 than their normal counterparts (p= 0.012). Age-wise, females between the ages, 30 to 40 had a significant increase (p= 0.046) but males under 30 showed a statistically meaningful decrease in the expression level (p= 0.04). In terms of sex, only the female patients manifested a statistically significant increase in IFNAR1 (p= 0.004). CONCLUSIONS: The overall results show an increase in IFNAR1 level in MS patients treated with IFN-B.


Assuntos
Fatores Imunológicos/uso terapêutico , Interferon beta/uso terapêutico , Esclerose Múltipla Recidivante-Remitente/terapia , Receptor de Interferon alfa e beta/genética , Adulto , Estudos de Casos e Controles , Feminino , Regulação da Expressão Gênica , Humanos , Masculino , Pessoa de Meia-Idade , Esclerose Múltipla Recidivante-Remitente/genética , Esclerose Múltipla Recidivante-Remitente/imunologia , Esclerose Múltipla Recidivante-Remitente/patologia , Receptor de Interferon alfa e beta/agonistas , Receptor de Interferon alfa e beta/imunologia , Transdução de Sinais , Vitamina D/sangue , Vitamina D/imunologia
4.
J Interferon Cytokine Res ; 36(3): 180-91, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26700737

RESUMO

Type I interferons (IFNs) exhibit broad-spectrum antiviral activity, with potential utility against emerging acute virus infections that pose a threat to global health. Recombinant IFN-αs that have been approved for clinical use require cold storage and are administered through intramuscular or subcutaneous injection, features that are problematic for global distribution, storage, and administration. Cognizant that the biological potency of an IFN-α subtype is determined by its binding affinity to the type I IFN receptor, IFNAR, we identified a panel of small molecule nonpeptide compounds using an in silico screening strategy that incorporated specific structural features of amino acids in the receptor-binding domains of the most potent IFN-α, IFN alfacon-1. Hit compounds were selected based on ease of synthesis and formulation properties. In preliminary biological assays, we provide evidence that these compounds exhibit antiviral activity. This proof-of-concept study validates the strategy of in silico design and development for IFN mimetics.


Assuntos
Antivirais/farmacologia , Vírus da Encefalomiocardite/efeitos dos fármacos , Interferon-alfa/química , Peptidomiméticos/farmacologia , Receptor de Interferon alfa e beta/agonistas , Bibliotecas de Moléculas Pequenas/farmacologia , Antivirais/síntese química , Linfócitos B/efeitos dos fármacos , Linfócitos B/patologia , Linfócitos B/virologia , Linhagem Celular Tumoral , Simulação por Computador , Desenho de Fármacos , Vírus da Encefalomiocardite/crescimento & desenvolvimento , Expressão Gênica , Ensaios de Triagem em Larga Escala , Humanos , Ligantes , Modelos Moleculares , Peptidomiméticos/síntese química , Estrutura Secundária de Proteína , Receptor de Interferon alfa e beta/química , Receptor de Interferon alfa e beta/genética , Receptor de Interferon alfa e beta/metabolismo , Proteínas Recombinantes/química , Bibliotecas de Moléculas Pequenas/síntese química , Relação Estrutura-Atividade , Interface Usuário-Computador
5.
J Biol Chem ; 291(7): 3371-84, 2016 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-26679999

RESUMO

Type I interferons serve as the first line of defense against pathogen invasion. Binding of IFNs to its receptors, IFNAR1 and IFNAR2, is leading to activation of the IFN response. To determine whether structural perturbations observed during binding are propagated to the cytoplasmic domain, multiple mutations were introduced into the transmembrane helix and its surroundings. Insertion of one to five alanine residues near either the N or C terminus of the transmembrane domain (TMD) likely promotes a rotation of 100° and a translation of 1.5 Å per added residue. Surprisingly, the added alanines had little effect on the binding affinity of IFN to the cell surface receptors, STAT phosphorylation, or gene induction. Similarly, substitution of the juxtamembrane residues of the TMD with alanines, or replacement of the TMD of IFNAR1 with that of IFNAR2, did not affect IFN binding or activity. Finally, only the addition of 10 serine residues (but not 2 or 4) between the extracellular domain of IFNAR1 and the TMD had some effect on signaling. Bioinformatic analysis shows a correlation between high sequence conservation of TMDs of cytokine receptors and the ability to transmit structural signals. Sequence conservation near the TMD of IFNAR1 is low, suggesting limited functional importance for this region. Our results suggest that IFN binding to the extracellular domains of IFNAR1 and IFNAR2 promotes proximity between the intracellular domains and that differential signaling is a function of duration of activation and affinity of binding rather than specific conformational changes transmitted from the outside to the inside of the cell.


Assuntos
Interferon-alfa/metabolismo , Modelos Moleculares , Receptor de Interferon alfa e beta/agonistas , Transdução de Sinais , Sequência de Aminoácidos , Linhagem Celular , Biologia Computacional , Sequência Conservada , Técnicas de Inativação de Genes , Humanos , Cinética , Mutagênese Insercional , Proteínas Mutantes/agonistas , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Fragmentos de Peptídeos/agonistas , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Conformação Proteica , Domínios e Motivos de Interação entre Proteínas , Estrutura Terciária de Proteína , Receptor de Interferon alfa e beta/química , Receptor de Interferon alfa e beta/genética , Receptor de Interferon alfa e beta/metabolismo , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo
6.
PLoS Pathog ; 11(5): e1004897, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25954804

RESUMO

In mice, plasmacytoid dendritic cells (pDC) and natural killer (NK) cells both contribute to resistance to systemic infections with herpes viruses including mouse Cytomegalovirus (MCMV). pDCs are the major source of type I IFN (IFN-I) during MCMV infection. This response requires pDC-intrinsic MyD88-dependent signaling by Toll-Like Receptors 7 and 9. Provided that they express appropriate recognition receptors such as Ly49H, NK cells can directly sense and kill MCMV-infected cells. The loss of any one of these responses increases susceptibility to infection. However, the relative importance of these antiviral immune responses and how they are related remain unclear. In humans, while IFN-I responses are essential, MyD88 is dispensable for antiviral immunity. Hence, a higher redundancy has been proposed in the mechanisms promoting protective immune responses against systemic infections by herpes viruses during natural infections in humans. It has been assumed, but not proven, that mice fail to mount protective MyD88-independent IFN-I responses. In humans, the mechanism that compensates MyD88 deficiency has not been elucidated. To address these issues, we compared resistance to MCMV infection and immune responses between mouse strains deficient for MyD88, the IFN-I receptor and/or Ly49H. We show that selective depletion of pDC or genetic deficiencies for MyD88 or TLR9 drastically decreased production of IFN-I, but not the protective antiviral responses. Moreover, MyD88, but not IFN-I receptor, deficiency could largely be compensated by Ly49H-mediated antiviral NK cell responses. Thus, contrary to the current dogma but consistent with the situation in humans, we conclude that, in mice, in our experimental settings, MyD88 is redundant for IFN-I responses and overall defense against a systemic herpes virus infection. Moreover, we identified direct NK cell sensing of infected cells as one mechanism able to compensate for MyD88 deficiency in mice. Similar mechanisms likely contribute to protect MyD88- or IRAK4-deficient patients from viral infections.


Assuntos
Infecções por Herpesviridae/imunologia , Interações Hospedeiro-Patógeno , Interferon Tipo I/metabolismo , Células Matadoras Naturais/imunologia , Muromegalovirus/imunologia , Fator 88 de Diferenciação Mieloide/metabolismo , Receptor de Interferon alfa e beta/agonistas , Animais , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Células Dendríticas/virologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Infecções por Herpesviridae/sangue , Infecções por Herpesviridae/metabolismo , Infecções por Herpesviridae/virologia , Imunidade Inata , Síndromes de Imunodeficiência/imunologia , Síndromes de Imunodeficiência/metabolismo , Síndromes de Imunodeficiência/virologia , Interferon Tipo I/sangue , Interleucina-12/metabolismo , Células Matadoras Naturais/metabolismo , Células Matadoras Naturais/virologia , Camundongos Endogâmicos BALB C , Camundongos Knockout , Camundongos Mutantes , Muromegalovirus/fisiologia , Fator 88 de Diferenciação Mieloide/deficiência , Fator 88 de Diferenciação Mieloide/genética , Subfamília A de Receptores Semelhantes a Lectina de Células NK/deficiência , Subfamília A de Receptores Semelhantes a Lectina de Células NK/genética , Subfamília A de Receptores Semelhantes a Lectina de Células NK/metabolismo , Doenças da Imunodeficiência Primária , Receptor de Interferon alfa e beta/deficiência , Receptor de Interferon alfa e beta/genética , Receptor de Interferon alfa e beta/metabolismo , Transdução de Sinais , Organismos Livres de Patógenos Específicos , Baço/imunologia , Baço/metabolismo , Baço/virologia , Receptor Toll-Like 9/deficiência , Receptor Toll-Like 9/genética , Receptor Toll-Like 9/metabolismo
7.
Eur J Immunol ; 41(6): 1687-95, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21469099

RESUMO

Autoimmune diseases including rheumatoid arthritis (RA) involve immune reactions against specific antigens. The type I IFN system is suspected to promote autoimmunity in systemic lupus erythematosus, but may also dampen immune reactions in e.g. inflammatory bowel disease. This prompted us to investigate the role of type I IFN in antigen-induced arthritis (AIA). The importance of type I IFN in methylated (m) BSA-induced arthritis was studied by using mice deficient for the type I IFN receptor (IFNAR) and by administration of the IFN-α activator viral double-stranded (ds) RNA or recombinant IFN-α at antigen sensitization. In IFNAR knock-out mice, arthritis severity was significantly higher than in WT mice. Administration of dsRNA at antigen sensitization protected WT but not IFNAR KO mice from arthritis. Also, addition of recombinant IFN-α during the immunization, but not the induction phase of arthritis, almost abolished arthritis. Protection mediated by IFN-α was accompanied by delayed and decreased antigen-specific proliferative responses, including impaired lymph node recall responses after intra-articular antigenic challenge. In conclusion, we demonstrate that type I IFN can prevent joint inflammation by downregulating antigen-specific cellular immunity.


Assuntos
Artrite Experimental/imunologia , Artrite Reumatoide/imunologia , Interferon-alfa , Receptor de Interferon alfa e beta/agonistas , Linfócitos T/metabolismo , Animais , Artrite Experimental/induzido quimicamente , Bovinos , Processos de Crescimento Celular/efeitos dos fármacos , Células Cultivadas , Modelos Animais de Doenças , Progressão da Doença , Humanos , Imunidade Ativa/efeitos dos fármacos , Imunidade Ativa/genética , Interferon-alfa/administração & dosagem , Interferon-alfa/farmacologia , Ativação Linfocitária/efeitos dos fármacos , Ativação Linfocitária/genética , Camundongos , Camundongos Knockout , RNA Viral/administração & dosagem , Receptor de Interferon alfa e beta/genética , Soroalbumina Bovina/administração & dosagem , Soroalbumina Bovina/química , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Linfócitos T/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA