Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Int J Mol Sci ; 24(13)2023 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-37445672

RESUMO

There is an urgent need to identify efficient antiviral compounds to combat existing and emerging RNA virus infections, particularly those related to seasonal and pandemic influenza outbreaks. While inhibitors of the influenza viral integral membrane proton channel protein (M2), neuraminidase (NA), and cap-dependent endonuclease are available, circulating influenza viruses acquire resistance over time. Thus, the need for the development of additional anti-influenza drugs with novel mechanisms of action exists. In the present study, a cell-based screening assay and a small molecule library were used to screen for activities that antagonized influenza A non-structural protein 1 (NS1), a highly conserved, multifunctional accessory protein that inhibits the type I interferon response against influenza. Two potential anti-influenza agents, compounds 157 and 164, were identified with anti-NS1 activity, resulting in the reduction of A/PR/8/34(H1N1) influenza A virus replication and the restoration of IFN-ß expression in human lung epithelial A549 cells. A 3D pharmacophore modeling study of the active compounds provided a glimpse of the structural motifs that may contribute to anti-influenza virus activity. This screening approach is amenable to a broader analysis of small molecule compounds to inhibit other viral targets.


Assuntos
Vírus da Influenza A Subtipo H1N1 , Vírus da Influenza A , Influenza Humana , Interferon Tipo I , Humanos , Vírus da Influenza A Subtipo H1N1/genética , Interferon Tipo I/metabolismo , Proteínas não Estruturais Virais/metabolismo , Influenza Humana/tratamento farmacológico , Vírus da Influenza A/genética , Antivirais/farmacologia , Antivirais/metabolismo , Replicação Viral
2.
J Virol ; 93(18)2019 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-31243131

RESUMO

Current combination antiretroviral therapies (cART) are unable to eradicate HIV-1 from infected individuals because of the establishment of proviral latency in long-lived cellular reservoirs. The shock-and-kill approach aims to reactivate viral replication from the latent state (shock) using latency-reversing agents (LRAs), followed by the elimination of reactivated virus-producing cells (kill) by specific therapeutics. The NF-κB RelA/p50 heterodimer has been characterized as an essential component of reactivation of the latent HIV-1 long terminal repeat (LTR). Nevertheless, prolonged NF-κB activation contributes to the development of various autoimmune, inflammatory, and malignant disorders. In the present study, we established a cellular model of HIV-1 latency in J-Lat CD4+ T cells that stably expressed the NF-κB superrepressor IκB-α 2NΔ4 and demonstrate that conventional treatments with bryostatin-1 and hexamethylenebisacetamide (HMBA) or ionomycin synergistically reactivated HIV-1 from latency, even under conditions where NF-κB activation was repressed. Using specific calcineurin phosphatase, p38, and MEK1/MEK2 kinase inhibitors or specific short hairpin RNAs, c-Jun was identified to be an essential factor binding to the LTR enhancer κB sites and mediating the combined synergistic reactivation effect. Furthermore, acetylsalicylic acid (ASA), a potent inhibitor of the NF-κB activator kinase IκB kinase ß (IKK-ß), did not significantly diminish reactivation in a primary CD4+ T central memory (TCM) cell latency model. The present work demonstrates that the shock phase of the shock-and-kill approach to reverse HIV-1 latency may be achieved in the absence of NF-κB, with the potential to avoid unwanted autoimmune- and or inflammation-related side effects associated with latency-reversing strategies.IMPORTANCE The shock-and-kill approach consists of the reactivation of HIV-1 replication from latency using latency-reversing agents (LRAs), followed by the elimination of reactivated virus-producing cells. The cellular transcription factor NF-κB is considered a master mediator of HIV-1 escape from latency induced by LRAs. Nevertheless, a systemic activation of NF-κB in HIV-1-infected patients resulting from the combined administration of different LRAs could represent a potential risk, especially in the case of a prolonged treatment. We demonstrate here that conventional treatments with bryostatin-1 and hexamethylenebisacetamide (HMBA) or ionomycin synergistically reactivate HIV-1 from latency, even under conditions where NF-κB activation is repressed. Our study provides a molecular proof of concept for the use of anti-inflammatory drugs, like aspirin, capable of inhibiting NF-κB in patients under combination antiretroviral therapy during the shock-and-kill approach, to avoid potential autoimmune and inflammatory disorders that can be elicited by combinations of LRAs.


Assuntos
HIV-1/efeitos dos fármacos , Ativação Viral/efeitos dos fármacos , Latência Viral/efeitos dos fármacos , Antirretrovirais/uso terapêutico , Linfócitos T CD4-Positivos/imunologia , Regulação Viral da Expressão Gênica/genética , Infecções por HIV/virologia , Soropositividade para HIV/imunologia , HIV-1/fisiologia , Humanos , Células Jurkat , Inibidor de NF-kappaB alfa/metabolismo , NF-kappa B/metabolismo , Provírus/efeitos dos fármacos , Provírus/fisiologia , Receptores Imunológicos/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fator de Transcrição RelA/metabolismo , Replicação Viral/efeitos dos fármacos
3.
J Immunol ; 186(4): 1951-62, 2011 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-21220691

RESUMO

Despite extensive studies that unraveled ligands and signal transduction pathways triggered by TLRs, little is known about the regulation of TLR gene expression. TLR3 plays a crucial role in the recognition of viral pathogens and induction of immune responses by myeloid DCs. IFN regulatory factor (IRF)-8, a member of the IRF family, is a transcriptional regulator that plays essential roles in the development and function of myeloid lineage, affecting different subsets of myeloid DCs. In this study, we show that IRF-8 negatively controls TLR3 gene expression by suppressing IRF-1- and/or polyinosinic-polycytidylic acid-stimulated TLR3 expression in primary human monocyte-derived DCs (MDDCs). MDDCs expressed TLR3 increasingly during their differentiation from monocytes to DCs with a peak at day 5, when TLR3 expression was further enhanced upon stimulation with polyinosinic-polycytidylic acid and then was promptly downregulated. We found that both IRF-1 and IRF-8 bind the human TLR3 promoter during MDDC differentiation in vitro and in vivo but with different kinetic and functional effects. We demonstrate that IRF-8-induced repression of TLR3 is specifically mediated by ligand-activated Src homology 2 domain-containing protein tyrosine phosphatase association. Indeed, Src homology 2 domain-containing protein tyrosine phosphatase-dephosphorylated IRF-8 bound to the human TLR3 promoter competing with IRF-1 and quashing its activity by recruitment of histone deacetylase 3. Our findings identify IRF-8 as a key player in the control of intracellular viral dsRNA-induced responses and highlight a new mechanism for negative regulation of TLR3 expression that can be exploited to block excessive TLR activation.


Assuntos
Células Dendríticas/imunologia , Regulação para Baixo/imunologia , Fatores Reguladores de Interferon/fisiologia , Células Mieloides/imunologia , Proteína Tirosina Fosfatase não Receptora Tipo 2/metabolismo , Receptor 3 Toll-Like/antagonistas & inibidores , Receptor 3 Toll-Like/genética , Domínios de Homologia de src/imunologia , Células Dendríticas/enzimologia , Células Dendríticas/virologia , Regulação para Baixo/genética , Regulação da Expressão Gênica/imunologia , Humanos , Fatores Reguladores de Interferon/genética , Fatores Reguladores de Interferon/metabolismo , Líquido Intracelular/imunologia , Líquido Intracelular/metabolismo , Líquido Intracelular/virologia , Ligantes , Células Mieloides/enzimologia , Células Mieloides/virologia , Poli I-C/farmacologia , Ligação Proteica/genética , Ligação Proteica/imunologia , Proteína Tirosina Fosfatase não Receptora Tipo 2/fisiologia , RNA Viral/farmacologia , Receptor 3 Toll-Like/metabolismo , Domínios de Homologia de src/genética
4.
Neoplasia ; 22(10): 459-469, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32784074

RESUMO

Interferon Regulatory Factors (IRFs) are key regulators of immunity, cell survival and apoptosis. IRF transcriptional activity and subcellular localization are tightly regulated by posttranscriptional modifications including phosphorylation. The IκB kinase family member IKK-ε is essential in regulating antiviral innate immunity mediated by IRFs but is now also recognized as an oncoprotein amplified and overexpressed in breast cancer cell lines and patient-derived tumors. In the present study, we report that the tumor suppressor IRF-1 is a specific target of IKK-ε in breast cancer cells. IKK-ε-mediated phosphorylation of IRF-1 dramatically decreases IRF-1 protein stability, accelerating IRF-1 degradation and quenching IRF-1 transcriptional activity. Chemical inhibition of IKK-ε activity, fully restores IRF-1 levels and function and positively correlates with inhibition of cell growth and proliferation of breast cancer cells. By using a breast cancer cell line stably expressing a dominant negative version of IRF-1 we were able to demonstrate that IKK-ε preferentially exerts its oncogenic potential in breast cancer through the regulation of IRF-1 and point to the IKK-ε-mediated phosphorylation of IRF-1 as a therapeutic target to overcome IKK-ε-mediated tumorigenesis.


Assuntos
Neoplasias da Mama/patologia , Quinase I-kappa B/metabolismo , Fator Regulador 1 de Interferon/metabolismo , Ubiquitina/metabolismo , Apoptose , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Proliferação de Células , Feminino , Humanos , Quinase I-kappa B/genética , Fator Regulador 1 de Interferon/genética , Fosforilação , Proteólise , Transdução de Sinais , Células Tumorais Cultivadas , Ubiquitinação
5.
J Virol ; 82(7): 3632-41, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18216101

RESUMO

Human immunodeficiency virus type 1 (HIV-1) gene expression is controlled by a complex interplay between viral and host factors. We have previously shown that interferon-regulatory factor 1 (IRF-1) is stimulated early after HIV-1 infection and regulates promoter transcriptional activity even in the absence of the viral transactivator Tat. In this work we demonstrate that IRF-1 is also required for full NF-kappaB transcriptional activity. We provide evidence that IRF-1 and NF-kappaB form a functional complex at the long terminal repeat (LTR) kappaB sites, which is abolished by specific mutations in the two adjacent kappaB sites in the enhancer region. Silencing IRF-1 with small interfering RNA resulted in impaired NF-kappaB-mediated transcriptional activity and in repressed HIV-1 transcription early in de novo-infected T cells. These data indicate that in early phases of HIV-1 infection or during virus reactivation from latency, when the viral transactivator is absent or present at very low levels, IRF-1 is an additional component of the p50/p65 heterodimer binding the LTR enhancer, absolutely required for efficient HIV-1 replication.


Assuntos
Ampliador HIV/genética , Repetição Terminal Longa de HIV/genética , HIV-1/fisiologia , Fator Regulador 1 de Interferon/metabolismo , NF-kappa B/metabolismo , Sítios de Ligação , Linhagem Celular , Ensaio de Desvio de Mobilidade Eletroforética , Inativação Gênica , HIV-1/genética , Humanos , Imunoprecipitação , Fator Regulador 1 de Interferon/antagonistas & inibidores , Mutação Puntual , Ligação Proteica , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Linfócitos T/virologia
6.
Viruses ; 10(2)2018 02 24.
Artigo em Inglês | MEDLINE | ID: mdl-29495311

RESUMO

The interferon (IFN) system is the first line of defense against viral infections. Evasion of IFN signaling by Ebola viral protein 24 (VP24) is a critical event in the pathogenesis of the infection and, hence, VP24 is a potential target for drug development. Since no drugs target VP24, the identification of molecules able to inhibit VP24, restoring and possibly enhancing the IFN response, is a goal of concern. Accordingly, we developed a dual signal firefly and Renilla luciferase cell-based drug screening assay able to quantify IFN-mediated induction of Interferon Stimulated Genes (ISGs) and its inhibition by VP24. Human Embryonic Kidney 293T (HEK293T) cells were transiently transfected with a luciferase reporter gene construct driven by the promoter of ISGs, Interferon-Stimulated Response Element (ISRE). Stimulation of cells with IFN-α activated the IFN cascade leading to the expression of ISRE. Cotransfection of cells with a plasmid expressing VP24 cloned from a virus isolated during the last 2014 outbreak led to the inhibition of ISRE transcription, quantified by a luminescent signal. To adapt this system to test a large number of compounds, we performed it in 96-well plates; optimized the assay analyzing different parameters; and validated the system by calculating the Z'- and Z-factor, which showed values of 0.62 and 0.53 for IFN-α stimulation assay and VP24 inhibition assay, respectively, indicative of robust assay performance.


Assuntos
Ebolavirus/genética , Genes Reporter/genética , Interferon beta/genética , Luciferases/genética , Proteínas Virais/genética , Proteínas Virais/metabolismo , Descoberta de Drogas , Células HEK293 , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Humanos , Interferon-alfa/farmacologia , Regiões Promotoras Genéticas/efeitos dos fármacos , Reprodutibilidade dos Testes , Transdução de Sinais/efeitos dos fármacos , Transfecção , Proteínas Virais/efeitos dos fármacos
7.
Ann N Y Acad Sci ; 1095: 325-33, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17404045

RESUMO

The interferon regulatory factor 7 (IRF-7), a member of the IRF family of transcription factors, is a key player in the innate immune response against viral infections. Constitutive expression of IRF-7 is limited to peripheral blood lymphocytes and dendritic cells while in most cell types its expression can be induced by type I interferon (INF). IRF-7 is sequestered in the cytoplasm of uninfected cells and following viral infection, double-stranded RNA (dsRNA), or toll-like receptor (TLR) signaling, it becomes phosphorylated by TBK and IKK-i kinases. Phosphorylated IRF-7 migrates in the nucleus where it can activate IFN type I genes and other interferon-stimulated genes (ISGs). Here we report that the overexpression of a constitutively active form of IRF-7 binds and positively regulates the transcriptional activity of the promotor of IRF-1 and low molecular mass polypeptide-2 (LMP-2), two proteins that play a key role in adaptive immunity. The so far unrecognized role of IRF-7 in LMP-2 stimulation points to IRF-7 as a transcriptional regulator that bridges innate and adaptive immunity.


Assuntos
Regulação da Expressão Gênica/imunologia , Imunidade Ativa/genética , Fator Regulador 7 de Interferon/fisiologia , Linhagem Celular , Cisteína Endopeptidases/metabolismo , Humanos , Imunidade Inata/genética , Fator Regulador 1 de Interferon/biossíntese , Fator Regulador 1 de Interferon/genética
8.
Cell Signal ; 14(6): 537-45, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11897494

RESUMO

Macrophage cell lines exhibit different responses to IFN-gamma depending on their maturation stage. We investigated the mechanisms underlying the differential IFN-gamma responsiveness in the less mature P388.D1 and in mature RAW264.7 cells. A reduction in the binding activity of the signal transducer and activator of transcription-1 (STAT-1) to different STAT binding elements (SBEs) was observed in P388.D1. This reduced binding activity was not due to an impaired STAT-1 activation. Studies on the expression of a negative regulator of cytokine signalling, protein-inhibiting activated STAT-1 (PIAS-1), showed that this protein was expressed constitutively at high levels in P388.D1. Forced expression of a PIAS-1 homologue, the Gu binding protein (GBP), inhibited the STAT-1-mediated gene activation in RAW264.7 cells, whereas a construct expressing the 5' portion of GBP in the antisense orientation reverts the IFN-gamma-resistant phenotype of P388.D1. Thus, our results indicate that PIAS-1 may account for the differential IFN-gamma responsiveness in macrophage cell lines at different stages of maturation.


Assuntos
Interferon gama/antagonistas & inibidores , Macrófagos/imunologia , Proteínas/fisiologia , Animais , Diferenciação Celular , Linhagem Celular , DNA Antissenso/farmacologia , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/metabolismo , Interferon gama/farmacologia , Macrófagos/efeitos dos fármacos , Camundongos , Proteínas Inibidoras de STAT Ativados , Proteínas/genética , RNA Mensageiro/biossíntese , Fator de Transcrição STAT1 , Transdução de Sinais , Transativadores/antagonistas & inibidores , Transativadores/metabolismo
9.
Biochem J ; 377(Pt 2): 367-78, 2004 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-14505489

RESUMO

IRFs [IFN (interferon) regulatory factors] constitute a family of transcription factors involved in IFN signalling and in the development and differentiation of the immune system. IRF-2 has generally been described as an antagonist of IRF-1-mediated transcription of IFN and IFN-inducible genes; however, it has been recently identified as a transcriptional activator of some genes, such as those encoding histone H4, VCAM-1 (vascular cell adhesion molecule-1) and Fas ligand. Biologically, IRF-2 plays an important role in cell growth regulation and has been shown to be a potential oncogene. Studies in knock-out mice have also implicated IRF-2 in the differentiation and functionality of haematopoietic cells. Here we show that IRF-2 expression in a myeloid progenitor cell line leads to reprogramming of these cells towards the megakaryocytic lineage and enables them to respond to thrombopoietin, as assessed by cell morphology and expression of specific differentiation markers. Up-regulation of transcription factors involved in the development of the megakaryocytic lineage, such as GATA-1, GATA-2, FOG-1 (friend of GATA-1) and NF-E2 (nuclear factor-erythroid-2), and transcriptional stimulation of the thrombopoietin receptor were also demonstrated. Our results provide evidence for a key role for IRF-2 in the induction of a programme of megakaryocytic differentiation, and reveal a remarkable functional diversity of this transcription factor in the regulation of cellular responses.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Megacariócitos/metabolismo , Proteínas Repressoras , Apoptose , Diferenciação Celular , Divisão Celular , Linhagem Celular , Linhagem da Célula , Humanos , Fator Regulador 2 de Interferon , Megacariócitos/citologia , Células Progenitoras Mieloides/fisiologia , Fatores de Transcrição/metabolismo , Ativação Transcricional
10.
Mol Cell Biol ; 34(6): 1054-65, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24396068

RESUMO

IκB kinase ε (IKK-ε) has an essential role as a regulator of innate immunity, functioning downstream of pattern recognition receptors to modulate NF-κB and interferon (IFN) signaling. In the present study, we investigated IKK-ε activation following T cell receptor (TCR)/CD28 stimulation of primary CD4(+) T cells and its role in the stimulation of a type I IFN response. IKK-ε was activated following TCR/CD28 stimulation of primary CD4(+) T cells; however, in T cells treated with poly(I·C), TCR/CD28 costimulation blocked induction of IFN-ß transcription. We demonstrated that IKK-ε phosphorylated the transcription factor IFN regulatory factor 1 (IRF-1) at amino acid (aa) 215/219/221 in primary CD4(+) T cells and blocked its transcriptional activity. At the mechanistic level, IRF-1 phosphorylation impaired the physical interaction between IRF-1 and the NF-κB RelA subunit and interfered with PCAF-mediated acetylation of NF-κB RelA. These results demonstrate that TCR/CD28 stimulation of primary T cells stimulates IKK-ε activation, which in turn contributes to suppression of IFN-ß production.


Assuntos
Linfócitos T CD4-Positivos/metabolismo , Quinase I-kappa B/genética , Fator Regulador 1 de Interferon/genética , Ativação Linfocitária/genética , Acetilação , Antígenos CD28/genética , Antígenos CD28/metabolismo , Complexo CD3/genética , Complexo CD3/metabolismo , Linhagem Celular , Células HEK293 , Humanos , Quinase I-kappa B/metabolismo , Fator Regulador 1 de Interferon/metabolismo , Interferon beta/genética , Interferon beta/metabolismo , Processamento de Proteína Pós-Traducional/genética , Fator de Transcrição RelA/genética , Fator de Transcrição RelA/metabolismo , Transcrição Gênica/genética , Fatores de Transcrição de p300-CBP/genética , Fatores de Transcrição de p300-CBP/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa