Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Bioorg Med Chem Lett ; 30(24): 127613, 2020 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-33075488

RESUMO

Type I Interferon (IFN) signaling plays an important role in the immune defense system against virus infection and in the innate immune response, thus IFNs are widely used as anti-viral agents and treatment for immune disorder or cancer. However, there is a growing demand for novel small-molecule IFN inducer due to tolerance, toxicity, or short duration of action following direct administration of IFNs. In this study, we assessed arylpiperazine (ARP) as a new core skeleton of IFN inducer. To investigate structure-activity relationship, we designed and synthesized a series of ARP analogues and evaluated the ability to stimulate IFN response in THP-1 human monocyte cells. Compound 5i was identified as a potent type I IFN inducer as it significantly increased cytokine secretion and increased expression of various IFN-stimulating genes which are representative biomarkers of type I IFN pathway. Our results suggested a beneficial therapeutic potential of 5i as an anti-viral agent.


Assuntos
Indutores de Interferon/química , Indutores de Interferon/farmacologia , Monócitos/efeitos dos fármacos , Piperazinas/química , Piperazinas/farmacologia , Desenho de Fármacos , Humanos , Imunidade Inata/efeitos dos fármacos , Indutores de Interferon/síntese química , Interferon Tipo I/agonistas , Interferon Tipo I/imunologia , Monócitos/imunologia , Piperazinas/síntese química , Células THP-1
2.
J Dent Res ; 98(10): 1073-1080, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31340724

RESUMO

The success of immune checkpoint receptor blockade has brought exciting promises for the treatment of head and neck squamous cell carcinoma (HNSCC). While patients who respond to checkpoint inhibitors tend to develop a durable response, <15% of patients with HNSCC respond to immune checkpoint inhibitors, underscoring the critical need to alleviate cancer resistance to immunotherapy. Major advances have been made to elucidate the intrinsic and adaptive resistance mechanisms to immunotherapy. Central genomic events in HNSCC have been found to possess previously unknown roles in suppressing immune sensing. Such inhibitory function affects both the innate and adaptive arms of tumor-specific immunity. While checkpoint blockade effectively reinvigorates adaptive T-cell responses, additional targeting of the oncogenic inhibitors of innate immune sensing likely informs a novel and potent strategy for immune priming. This review discusses the recent advances on the identification of key HNSCC oncogenes that impair antitumor immunity and emerging immune-priming approaches that sensitize poorly immunogenic HNSCCs to checkpoint blockade. These approaches include but are not limited to cancer vaccine systems utilizing novel type I interferon agonists as immune adjuvants, radiation, DNA damage-inducing agents, and metabolic reprogramming. The goal of these multipronged approaches is to expand tumor-specific effector T-cells, break checkpoint receptor-mediated tolerance, and metabolically support sustained T-cell activation. The translation of therapeutics that reverses oncogenic inhibition of immune sensing requires thorough characterization of the HNSCC regulators of innate immune sensors, development of additional immunocompetent HNSCC mouse models, as well as engineering of more robust immune adjuvant delivery systems. Built on the success of checkpoint blockade, validation of novel immune-priming approaches holds key promises to expand the pool of responders to immunotherapy.


Assuntos
Antineoplásicos Imunológicos/uso terapêutico , Neoplasias de Cabeça e Pescoço/terapia , Imunoterapia , Carcinoma de Células Escamosas de Cabeça e Pescoço/terapia , Animais , Vacinas Anticâncer , Reprogramação Celular , Humanos , Interferon Tipo I/agonistas , Camundongos , Linfócitos T/imunologia
3.
J Biol Chem ; 293(8): 2701-2710, 2018 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-29301940

RESUMO

Mammalian Nod-like receptor (NLR) proteins contribute to the regulation and induction of innate and adaptive immunity in mammals, although the function of about half of the currently identified NLR proteins remains poorly characterized. Here we analyzed the function of the primate-specific NLRP11 gene product. We show that NLRP11 is highly expressed in immune cells, including myeloid cells, B cells, and some B cell lymphoma lines. Overexpression of NLRP11 in human cells did not trigger key innate immune signaling pathways, including NF-κB and type I interferon responses. NLRP11 harbors a pyrin domain, which is responsible for inflammasome formation in related NLR proteins. However, NLRP11 did not interact with the inflammasome adaptor protein ASC, and it did not trigger caspase-1 activation. By contrast, expression of NLRP11 specifically repressed NF-κB and type I interferon responses, two key innate immune pathways involved in inflammation. This effect was independent of the pyrin domain and ATPase activity of NLRP11. siRNA-mediated knockdown of NLRP11 in human myeloid THP1 cells validated these findings and revealed enhanced lipopolysaccharide and Sendai virus-induced cytokine and interferon responses, respectively, in cells with reduced NLRP11 expression. In summary, our work identifies a novel role of NLRP11 in the regulation of inflammatory responses in human cells.


Assuntos
Linfócitos B/metabolismo , Regulação para Baixo , Regulação da Expressão Gênica , Imunidade Inata , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Células Mieloides/metabolismo , Proteínas NLR/metabolismo , Substituição de Aminoácidos , Linfócitos B/citologia , Linfócitos B/efeitos dos fármacos , Linfócitos B/imunologia , Linhagem Celular Transformada , Linhagem Celular Tumoral , Regulação para Baixo/efeitos dos fármacos , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Genes Reporter/efeitos dos fármacos , Humanos , Imunidade Inata/efeitos dos fármacos , Interferon Tipo I/agonistas , Interferon Tipo I/antagonistas & inibidores , Interferon Tipo I/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Peptídeos e Proteínas de Sinalização Intracelular/química , Peptídeos e Proteínas de Sinalização Intracelular/genética , Lipopolissacarídeos/toxicidade , Masculino , Mutação , Células Mieloides/citologia , Células Mieloides/efeitos dos fármacos , Células Mieloides/imunologia , NF-kappa B/agonistas , NF-kappa B/antagonistas & inibidores , NF-kappa B/metabolismo , Proteínas NLR/genética , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Especificidade de Órgãos , Fragmentos de Peptídeos/antagonistas & inibidores , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Interferência de RNA , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo
4.
Neurochem Res ; 42(9): 2625-2638, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28540600

RESUMO

Microglia, the resident immune cells of the CNS, are primary regulators of the neuroimmune response to injury. Type I interferons (IFNs), including the IFNαs and IFNß, are key cytokines in the innate immune system. Their activity is implicated in the regulation of microglial function both during development and in response to neuroinflammation, ischemia, and neurodegeneration. Data from numerous studies in multiple sclerosis (MS) and stroke suggest that type I IFNs can modulate the microglial phenotype, influence the overall neuroimmune milieu, regulate phagocytosis, and affect blood-brain barrier integrity. All of these IFN-induced effects result in numerous downstream consequences on white matter pathology and microglial reactivity. Dysregulation of IFN signaling in mouse models with genetic deficiency in ubiquitin specific protease 18 (USP18) leads to a severe neurological phenotype and neuropathological changes that include white matter microgliosis and pro-inflammatory gene expression in dystrophic microglia. A class of genetic disorders in humans, referred to as pseudo-TORCH syndrome (PTS) for the clinical resemblance to infection-induced TORCH syndrome, also show dysregulation of IFN signaling, which leads to severe neurological developmental disease. In these disorders, the excessive activation of IFN signaling during CNS development results in a destructive interferonopathy with similar induction of microglial dysfunction as seen in USP18 deficient mice. Other recent studies implicate "microgliopathies" more broadly in neurological disorders including Alzheimer's disease (AD) and MS, suggesting that microglia are a potential therapeutic target for disease prevention and/or treatment, with interferon signaling playing a key role in regulating the microglial phenotype.


Assuntos
Interferon Tipo I/metabolismo , Microglia/metabolismo , Transdução de Sinais/fisiologia , Substância Branca/metabolismo , Animais , Humanos , Indutores de Interferon/farmacologia , Interferon Tipo I/agonistas , Interferon Tipo I/imunologia , Microglia/efeitos dos fármacos , Microglia/imunologia , Doenças do Sistema Nervoso/imunologia , Doenças do Sistema Nervoso/metabolismo , Poli I-C/farmacologia , Transdução de Sinais/efeitos dos fármacos , Substância Branca/efeitos dos fármacos , Substância Branca/imunologia
5.
PLoS Pathog ; 11(4): e1004863, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25927359

RESUMO

Alphaviruses are a group of widely distributed human and animal pathogens. It is well established that their replication is sensitive to type I IFN treatment, but the mechanism of IFN inhibitory function remains poorly understood. Using a new experimental system, we demonstrate that in the presence of IFN-ß, activation of interferon-stimulated genes (ISGs) does not interfere with either attachment of alphavirus virions to the cells, or their entry and nucleocapsid disassembly. However, it strongly affects translation of the virion-delivered virus-specific RNAs. One of the ISG products, IFIT1 protein, plays a major role in this translation block, although an IFIT1-independent mechanism is also involved. The 5'UTRs of the alphavirus genomes were found to differ significantly in their ability to drive translation in the presence of increased concentration of IFIT1. Prior studies have shown that adaptation of naturally circulating alphaviruses to replication in tissue culture results in accumulation of mutations in the 5'UTR, which increase the efficiency of the promoter located in the 5'end of the genome. Here, we show that these mutations also decrease resistance of viral RNA to IFIT1-induced translation inhibition. In the presence of higher levels of IFIT1, alphaviruses with wt 5'UTRs became potent inducers of type I IFN, suggesting a new mechanism of type I IFN induction. We applied this knowledge of IFIT1 interaction with alphaviruses to develop new attenuated variants of Venezuelan equine encephalitis and chikungunya viruses that are more sensitive to the antiviral effects of IFIT1, and thus could serve as novel vaccine candidates.


Assuntos
Alphavirus/fisiologia , Proteínas de Transporte/metabolismo , Regulação Viral da Expressão Gênica , Genoma Viral , Interações Hospedeiro-Patógeno , Interferon Tipo I/agonistas , Replicação Viral , Regiões 5' não Traduzidas , Proteínas Adaptadoras de Transdução de Sinal , Aedes , Alphavirus/genética , Alphavirus/imunologia , Animais , Linhagem Celular , Células Cultivadas , Vírus Chikungunya/genética , Vírus Chikungunya/imunologia , Vírus Chikungunya/fisiologia , Regulação para Baixo , Vírus da Encefalite Equina Venezuelana/genética , Vírus da Encefalite Equina Venezuelana/imunologia , Vírus da Encefalite Equina Venezuelana/fisiologia , Vacinas Fúngicas/metabolismo , Interferon Tipo I/genética , Interferon Tipo I/metabolismo , Camundongos , Células-Tronco Embrionárias Murinas/citologia , Células-Tronco Embrionárias Murinas/imunologia , Células-Tronco Embrionárias Murinas/metabolismo , Células-Tronco Embrionárias Murinas/virologia , Mutação , Células NIH 3T3 , RNA/metabolismo , Proteínas de Ligação a RNA , Tropismo Viral
6.
J Biol Chem ; 289(42): 29014-29, 2014 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-25193661

RESUMO

IFNß is a common therapeutic option to treat multiple sclerosis. It is unique among the family of type I IFNs in that it binds to the interferon receptors with high affinity, conferring exceptional biological properties. We have previously reported the generation of an interferon superagonist (dubbed YNSα8) that is built on the backbone of a low affinity IFNα but modified to exhibit higher receptor affinity than even for IFNß. Here, YNSα8 was fused with a 600-residue hydrophilic, unstructured N-terminal polypeptide chain comprising proline, alanine, and serine (PAS) to prolong its plasma half-life via "PASylation." PAS-YNSα8 exhibited a 10-fold increased half-life in both pharmacodynamic and pharmacokinetic assays in a transgenic mouse model harboring the human receptors, notably without any detectable loss in biological potency or bioavailability. This long-lived superagonist conferred significantly improved protection from MOG35-55-induced experimental autoimmune encephalomyelitis compared with IFNß, despite being injected with a 4-fold less frequency and at an overall 16-fold lower dosage. These data were corroborated by FACS measurements showing a decrease of CD11b(+)/CD45(hi) myeloid lineage cells detectable in the CNS, as well as a decrease in IBA(+) cells in spinal cord sections determined by immunohistochemistry for PAS-YNSα8-treated animals. Importantly, PAS-YNSα8 did not induce antibodies upon repeated administration, and its biological efficacy remained unchanged after 21 days of treatment. A striking correlation between increased levels of CD274 (PD-L1) transcripts from spleen-derived CD4(+) cells and improved clinical response to autoimmune encephalomyelitis was observed, indicating that, at least in this mouse model of multiple sclerosis, CD274 may serve as a biomarker to predict the effectiveness of IFN therapy to treat this complex disease.


Assuntos
Encefalomielite Autoimune Experimental/tratamento farmacológico , Interferon Tipo I/agonistas , Interferon Tipo I/farmacologia , Peptídeos/química , Animais , Separação Celular , Encefalomielite Autoimune Experimental/metabolismo , Feminino , Citometria de Fluxo , Humanos , Interferon beta/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Esclerose Múltipla/tratamento farmacológico , Engenharia de Proteínas/métodos , Proteínas Recombinantes/química , Ressonância de Plasmônio de Superfície , Resultado do Tratamento
7.
Blood ; 119(24): 5750-7, 2012 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-22550346

RESUMO

A key feature differentiating nonpathogenic SIV infection of sooty mangabeys (SMs) from pathogenic HIV/SIV infections is the rapid resolution of type I IFN (IFN-I) responses and IFN-stimulated gene expression during the acute-to-chronic phase transition and the establishment of an immune quiescent state that persists throughout the chronic infection. We hypothesized that low levels of IFN-I signaling may help to prevent chronic immune activation and disease progression in SIV-infected SMs. To assess the effects of IFN-I signaling in this setting, in the present study, we administered recombinant rhesus macaque IFNα2-IgFc (rmIFNα2) to 8 naturally SIV-infected SMs weekly for 16 weeks. Gene-expression profiling revealed a strong up-regulation of IFN-stimulated genes in the blood of treated animals, confirming the reagent's bioactivity. Interestingly, we observed an approximately 1-log decrease in viral load that persisted through day 35 of treatment. Flow cytometric analysis of lymphocytes in the blood, lymph nodes, and rectal biopsies did not reveal a significant decline of CD4(+) T cells, a robust increase in lymphocyte activation, or change in the level of SIV-specific CD8(+) T cells. The results of the present study indicate that administration of type I IFNs in SIV-infected SMs induces a significant anti-viral effect that is not associated with a detectable increase in chronic immune activation.


Assuntos
Cercocebus atys/virologia , Interferon Tipo I/agonistas , Ativação Linfocitária/efeitos dos fármacos , Proteínas Recombinantes/farmacologia , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Vírus da Imunodeficiência Símia/fisiologia , Replicação Viral/efeitos dos fármacos , Animais , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/imunologia , Proliferação de Células/efeitos dos fármacos , Cercocebus atys/sangue , Cercocebus atys/imunologia , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Interferon Tipo I/administração & dosagem , Interferon Tipo I/farmacologia , Depleção Linfocítica , Macaca mulatta/imunologia , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/uso terapêutico , Síndrome de Imunodeficiência Adquirida dos Símios/sangue , Síndrome de Imunodeficiência Adquirida dos Símios/tratamento farmacológico , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Vírus da Imunodeficiência Símia/efeitos dos fármacos , Especificidade da Espécie , Carga Viral/efeitos dos fármacos , Viremia/sangue , Viremia/imunologia , Viremia/virologia
8.
Future Med Chem ; 3(9): 1199-208, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21806381

RESUMO

Prothymosin α (ProTα) is a 12.5-kDa, highly acidic protein widely distributed in different cell types expressed intracellularly and extracellularly. ProTα does not contain a secretion-signal sequence and is released by a nonclassical secretory pathway with a cargo protein. New findings on the extracellular function of ProTα have yielded exciting insights into the cytokine-like functions of this host protein that stimulates type I interferon via Toll-like receptor 4. Here, we discuss the intracellular function of ProTα, how new findings of cytokine-like activities of ProTα aid our understanding of mechanisms that direct ProTα functions, and the potential application of these new insights to the development of immunotherapies.


Assuntos
Imunoproteínas/farmacologia , Imunoterapia/métodos , Precursores de Proteínas/farmacologia , Timosina/análogos & derivados , Animais , Citocinas/farmacologia , Citocinas/uso terapêutico , Humanos , Imunoproteínas/metabolismo , Imunoproteínas/uso terapêutico , Interferon Tipo I/agonistas , Interferon Tipo I/metabolismo , Precursores de Proteínas/uso terapêutico , Proteínas/metabolismo , Proteínas/fisiologia , Timosina/farmacologia , Timosina/uso terapêutico , Receptor 4 Toll-Like/agonistas , Receptor 4 Toll-Like/metabolismo
9.
Arthritis Rheum ; 56(3): 1010-20, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17328080

RESUMO

OBJECTIVE: Microarray analyses of peripheral blood leukocytes have shown that patients with systemic lupus erythematosus express increased levels of type I interferon (IFN)-regulated genes. In this study we examined gene expression by peripheral blood mononuclear cells (PBMCs) from patients with systemic sclerosis (SSc) to better understand the dysregulation of the immune system in this disease. METHODS: PBMC gene expression was analyzed by microarray and confirmed by real-time polymerase chain reaction (PCR). Surface protein expression of Siglec-1 was analyzed by flow cytometry in PBMCs from healthy control subjects and patients with SSc, and in control PBMCs that were cultured in vitro with Toll-like receptor (TLR) agonists. RESULTS: SSc patients showed increased expression of a cluster of IFN-regulated genes, including Siglec-1 (CD169, sialoadhesin). This result was verified and extended by real-time PCR, showing that a subset of the SSc patients expressed strikingly increased levels of Siglec-1 messenger RNA (mRNA). Flow cytometry of PBMCs from SSc patients and healthy controls showed increased Siglec-1 surface protein expression, which was restricted to CD14+ monocytes. In vitro studies showed that type I IFN and certain TLR agonists, including TLR-7 and TLR-9, induced Siglec-1 mRNA and protein expression. Moreover, TLR induction of surface Siglec-1 was shown to be type I IFN-dependent. Increased numbers of Siglec-1+ cells were observed by immunohistochemistry in the skin of SSc patients compared with healthy controls. CONCLUSION: Increased expression of Siglec-1 in circulating SSc monocytes and tissue macrophages suggests that type I IFN-mediated activation of monocytes occurs in SSc, possibly through TLR activation of IFN secretion. These observations indicate a potential role for type I IFN-activated monocyte/macrophages in the pathogenesis of SSc.


Assuntos
Interferon Tipo I/agonistas , Glicoproteínas de Membrana/sangue , Monócitos/metabolismo , Receptores Imunológicos/sangue , Escleroderma Sistêmico/sangue , Receptores Toll-Like/agonistas , Adolescente , Adulto , Idoso , Biomarcadores/sangue , Estudos de Casos e Controles , Células Cultivadas , Feminino , Regulação da Expressão Gênica , Humanos , Interferon Tipo I/fisiologia , Receptores de Lipopolissacarídeos/metabolismo , Masculino , Glicoproteínas de Membrana/genética , Análise em Microsséries , Pessoa de Meia-Idade , Monócitos/imunologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores Imunológicos/genética , Escleroderma Sistêmico/metabolismo , Lectina 1 Semelhante a Ig de Ligação ao Ácido Siálico , Receptores Toll-Like/fisiologia
10.
Blood ; 97(2): 473-82, 2001 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-11154225

RESUMO

The ability to modify responses to type I interferons (IFNs) could alter processes such as hematopoiesis and immunity, which involve endogenous IFNs and responses to exogenous IFNs. The data presented here support a significant role for a recently identified soluble isoform of the murine type I IFN receptor, muIfnar-2a, as an efficient regulator of IFN responses. The messenger RNA (mRNA) transcript encoding muIfnar-2a is generally more abundant than that encoding the transmembrane isoform, muIfnar-2c. Furthermore, the ratio of muIfnar-2a:2c transcripts varied from more than 10:1 in the liver and other organs to less than 1:1 in bone-marrow macrophages, indicating independent regulation of the 2 transcripts encoding receptor isoforms and suggesting that the soluble muIfnar-2a levels are biologically relevant in some organs. Western blot analysis showed that soluble muIfnar-2 was present at high levels in murine serum and other biologic fluids and bound type I IFN. Recombinant muIfnar-2a competitively inhibited the activity of both IFNalpha and beta in reporter assays using the L929 cell line and in antiproliferative and antiviral assays using primary cells. Surprisingly, using primary thymocytes from Ifnar-2(-/-) mice, recombinant muIfnar-2a formed a complex with IFN alpha or beta and muIfnar-1 at the cell surface and transmitted an antiproliferative signal. These data indicate potential dual actions of soluble muIfnar-2 and imply that a signal can be transduced through the Ifnar-1 chain of the receptor complex in the absence of the cytoplasmic domain of Ifnar-2. Therefore, our results suggest that soluble Ifnar-2 is an important regulator of endogenous and systemically administered type I IFN.


Assuntos
Interferon Tipo I/metabolismo , Receptores de Interferon/metabolismo , Fatores Etários , Animais , Western Blotting , Células COS , Técnicas de Cultura de Células , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Fatores Imunológicos/metabolismo , Interferon Tipo I/agonistas , Interferon Tipo I/antagonistas & inibidores , Proteínas de Membrana , Camundongos , Modelos Animais , Peso Molecular , Isoformas de Proteínas/genética , RNA Mensageiro/metabolismo , Receptor de Interferon alfa e beta , Receptores de Interferon/genética , Receptores de Interferon/fisiologia , Proteínas Recombinantes/sangue , Proteínas Recombinantes/metabolismo , Transdução de Sinais/efeitos dos fármacos , Solubilidade , Timo/citologia , Timo/efeitos dos fármacos , Distribuição Tecidual , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA