Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
País de afiliação
Intervalo de ano de publicação
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.
Acta Trop ; 245: 106975, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37348801

RESUMO

Like most countries in sub-Saharan African countries, Benin continues to bear a heavy malaria burden. In 2014, the National Malaria Control Programme (NMCP) changed its treatment policy, and recommended the use of artemisinin-based combination therapy (ACT) as first-line treatment for uncomplicated Plasmodium falciparum cases. The study presented here was conducted to investigate the impact of current antimalarial drug resistance on the country. Molecular surveillance targeting the Pfcrt, Pfmdr1, Pfkelch13, dhfr, and dhps genes was carried out on samples from patients positive for P. falciparum malaria by microscopy, LAMP and PCR diagnostic test. Molecular analysis was performed using targeted amplicon deep sequencing (TADS). In addition, the frequency of parasites with dual deletion of the histidine-rich protein 2 and 3 genes (pfhrp2 and pfhrp3), known to be responsible of the performance of HRP-based malaria rapid diagnostic tests (HRP-RDT), was estimated. Fifty-three falciparum samples collected at the Saint Jean de Dieu hospital in Tanguiéta, Benin, were tested. No Pfkelch13 validated or candidate artemisinin partial resistant variants were identified. A marked prevalence of Asn51Ile (N51I), Cys59Arg (C59R), and Ser108Asn (S108N) mutant alleles was found in the dhfr gene, representing the most frequent genotype (64%). Five-point mutations were detected in dhps, Ile431Val (I431V), Ser436Ala (S436A), Ala437Gly (A437G), Ala581Gly (A581G), Ala613Ser (A613S) of which the third was the most common (92%). No mutation was identified in dhps Lys540Glu (K540E). The quintuple mutant genotype resulting from the combination of the dhfr triple mutant (51I/59R/108N) with the dhps double mutant 436A/437G was detected at a frequency of 30%. Low levels of mutations in Pfcrt and no mutation at codon 86 in the Pfmdr1 DNA fragment were observed, whereas a high level of Tyr184Phe (Y184F) polymorphism in the Pfmdr1 gene was found. These results could be indicative, over a decade after the implementation of ACT therapy, of the return of chloroquine-sensitive but artemether-lumefantrine resistant falciparum genotypes in Benin. There was no evidence of HRP2 and HRP3 deletions. Data from the present study support the need for routine monitoring of molecular markers of antimalarial drug resistance as part of surveillance activities aimed to make informed treatment policy decisions at the national level.


Assuntos
Antimaláricos , Malária Falciparum , Malária , Humanos , Antimaláricos/farmacologia , Antimaláricos/uso terapêutico , Plasmodium falciparum , Pirimetamina/uso terapêutico , Sulfadoxina , Benin/epidemiologia , Artemeter , Combinação Arteméter e Lumefantrina , Malária Falciparum/tratamento farmacológico , Malária Falciparum/epidemiologia , Malária Falciparum/parasitologia , Malária/tratamento farmacológico , Resistência a Medicamentos/genética , Tetra-Hidrofolato Desidrogenase/genética , Proteínas de Protozoários/genética , Combinação de Medicamentos
5.
Parasit Vectors ; 16(1): 238, 2023 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-37461081

RESUMO

BACKGROUND: ETRAMP11.2 (PVX_003565) is a well-characterized protein with antigenic potential. It is considered to be a serological marker for diagnostic tools, and it has been suggested as a potential vaccine candidate. Despite its immunological relevance, the polymorphism of the P. vivax ETRAMP11.2 gene (pvetramp11.2) remains undefined. The genetic variability of an antigen may limit the effectiveness of its application as a serological surveillance tool and in vaccine development and, therefore, the aim of this study was to investigate the genetic diversity of pvetramp11.2 in parasite populations from Amazonian regions and worldwide. We also evaluated amino acid polymorphism on predicted B-cell epitopes. The low variability of the sequence encoding PvETRAMP11.2 protein suggests that it would be a suitable marker in prospective serodiagnostic assays for surveillance strategies or in vaccine design against P. vivax malaria. METHODS: The pvetramp11.2 of P. vivax isolates collected from Brazil (n = 68) and Peru (n = 36) were sequenced and analyzed to assess nucleotide polymorphisms, allele distributions, population differentiation, genetic diversity and signature of selection. In addition, sequences (n = 104) of seven populations from different geographical regions were retrieved from the PlasmoDB database and included in the analysis to study the worldwide allele distribution. Potential linear B-cell epitopes and their polymorphisms were also explored. RESULTS: The multiple alignments of 208 pvetramp11.2 sequences revealed a low polymorphism and a marked geographical variation in allele diversity. Seven polymorphic sites and 11 alleles were identified. All of the alleles were detected in isolates from the Latin American region and five alleles were detected in isolates from the Southeast Asia/Papua New Guinea (SEA/PNG) region. Three alleles were shared by all Latin American populations (H1, H6 and H7). The H1 allele (reference allele from Salvador-1 strain), which was absent in the SEA/PNG populations, was the most represented allele in populations from Brazil (54%) and was also detected at high frequencies in populations from all other Latin America countries (range: 13.0% to 33.3%). The H2 allele was the major allele in SEA/PNG populations, but was poorly represented in Latin America populations (only in Brazil: 7.3%). Plasmodium vivax populations from Latin America showed a marked inter-population genetic differentiation (fixation index [Fst]) in contrast to SEA/PNG populations. Codon bias measures (effective number of codons [ENC] and Codon bias index [CBI]) indicated preferential use of synonymous codons, suggesting selective pressure at the translation level. Only three amino acid substitutions, located in the C-terminus, were detected. Linear B-cell epitope mapping predicted two epitopes in the Sal-1 PvETRAMP11.2 protein, one of which was fully conserved in all of the parasite populations analyzed. CONCLUSIONS: We provide an overview of the allele distribution and genetic differentiation of ETRAMP11.2 antigen in P. vivax populations from different endemic areas of the world. The reduced polymorphism and the high degree of protein conservation supports the application of PvETRAMP11.2 protein as a reliable antigen for application in serological assays or vaccine design. Our findings provide useful information that can be used to inform future study designs.


Assuntos
Malária Vivax , Plasmodium vivax , Humanos , Antígenos de Protozoários/genética , Epitopos de Linfócito B/genética , Variação Genética , Malária Vivax/parasitologia , Proteínas de Membrana/genética , Estudos Prospectivos , Proteínas de Protozoários/genética , Análise de Sequência de DNA
6.
J Virol ; 85(10): 5070-80, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21389130

RESUMO

Crucial steps in high-risk human papillomavirus (HR-HPV)-related carcinogenesis are the integration of HR-HPV into the host genome and loss of viral episomes. The mechanisms that promote cervical neoplastic progression are, however, not clearly understood. During HR-HPV infection, the HPV E5 protein is expressed in precancerous stages but not after viral integration. Given that it has been reported that loss of HPV16 episomes and cervical tumor progression are associated with increased expression of antiviral genes that are inducible by type I interferon (IFN), we asked whether E5, expressed in early phases of cervical carcinogenesis, affects IFN-ß signaling. We show that the HPV type 16 (HPV16) E5 protein expression per se stimulates IFN-ß expression. This stimulation is specifically mediated by the induction of interferon regulatory factor 1 (IRF-1) which, in turn, induces transcriptional activation of IRF-1-targeted interferon-stimulated genes (ISGs) as double-stranded RNA-dependent protein kinase R (PKR) and caspase 8. Our data show a new and unexpected role for HR-HPV E5 protein and indicate that HPV16 E5 may contribute to the mechanisms responsible for cervical carcinogenesis in part via stimulation of IFN-ß and an IFN signature, with IRF-1 playing a pivotal role. HPV16 E5 and IRF-1 may thus serve as potential therapeutic targets in HPV-associated premalignant lesions.


Assuntos
Interações Hospedeiro-Patógeno , Papillomavirus Humano 16/imunologia , Fator Regulador 1 de Interferon/metabolismo , Interferon beta/biossíntese , Queratinócitos/imunologia , Proteínas Oncogênicas Virais/metabolismo , Linhagem Celular , Papillomavirus Humano 16/patogenicidade , Humanos , Queratinócitos/virologia
7.
Int J Infect Dis ; 107: 234-241, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33940188

RESUMO

BACKGROUND: Recent studies showed the first emergence of the R561H artemisinin-associated resistance marker in Africa, which highlights the importance of continued molecular surveillance to assess the selection and spread of this and other drug resistance markers in the region. METHOD: In this study, we used targeted amplicon deep sequencing of 116 isolates collected in two areas of Cameroon to genotype the major drug resistance genes, k13, crt, mdr1, dhfr, and dhps, and the cytochrome b gene (cytb) in Plasmodium falciparum. RESULTS: No confirmed or associated artemisinin resistance markers were observed in Pfk13. In comparison, both major and minor alleles associated with drug resistance were found in Pfcrt, Pfmdr1, Pfdhfr, and Pfdhps. Notably, a high frequency of other nonsynonymous mutations was observed across all the genes, except for Pfcytb, suggesting continued selection pressure. CONCLUSIONS: The results from this study supported the continued use of artemisinin-based combination therapy and administration of sulfadoxine-pyrimethamine for intermittent preventive therapy in pregnant women, and for seasonal chemoprevention in these study sites in Cameroon.


Assuntos
Antimaláricos/farmacologia , Resistência a Medicamentos/genética , Marcadores Genéticos/genética , Sequenciamento de Nucleotídeos em Larga Escala , Plasmodium falciparum/efeitos dos fármacos , Plasmodium falciparum/genética , Alelos , Camarões , Feminino , Genótipo , Humanos , Mutação , Plasmodium falciparum/isolamento & purificação , Gravidez
8.
J Travel Med ; 28(5)2021 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-33295621

RESUMO

BACKGROUND: Plasmodium falciparum (P. falciparum) malaria is a significant public health problem in returning travellers, and artemisinin combination therapy (ACT) remains the first choice for treatment. Several single nucleotide polymorphisms (SNPs) in the P. falciparum kelch 13 (Pfk13) gene have been associated with artemisinin (ART) resistance. Moreover, the increase in the P. falciparum plasmepsin 2 (Pfpm2) gene copy number was shown to be linked with reduced susceptibility of P. falciparum to piperaquine (PPQ), a partner drug in an ACT regimen. Active molecular surveillance for imported drug-resistant malaria parasites is a pivotal activity to provide adequate chemoprophylaxis and treatment guidelines. METHODS: A retrospective study to review imported P. falciparum malaria in patients admitted to Spallanzani Institute between 2014 and 2015 was conducted. Information collected included clinic and epidemiological characteristics such as age, gender, country of origin, time since arrival to our country, travel history. All P.falciparum isolates were analysed for SNPs in the Pfk13 gene and for copy number variations in the Pfpm2 gene. RESULTS: P. falciparum malaria was identified in 54 travellers. The mean age was 37 years, 44 were males. All cases were imported from non-EU countries. In the Pfk13 gene two mutations (R561R and F673L) were detected. Six P. falciparum isolates carried two copies of Pfpm2 gene, and one three copies, representing ≈16% of the analysed isolates. CONCLUSIONS: None of the SNPs known to be associated with ART resistance were detected in the examined parasites. Our results provide evidence that Pfpm2 duplications (associated with piperaquine resistance) occur in Africa, emphasizing the necessity to better decode the genetic background associated with PPQ resistance. Further epidemiological investigations in Pfpm2 amplification along with mutations in the Pfk13 gene will be useful for developing and updating anti-malarial guidance in travellers.


Assuntos
Antimaláricos , Artemisininas , Malária Falciparum , Malária , Adulto , Antimaláricos/farmacologia , Antimaláricos/uso terapêutico , Artemisininas/uso terapêutico , Variações do Número de Cópias de DNA , Resistência a Medicamentos/genética , Humanos , Itália , Malária/tratamento farmacológico , Malária Falciparum/tratamento farmacológico , Malária Falciparum/epidemiologia , Masculino , Plasmodium falciparum/genética , Proteínas de Protozoários/genética , Proteínas de Protozoários/uso terapêutico , Estudos Retrospectivos
9.
J Immunol ; 181(3): 1673-82, 2008 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-18641303

RESUMO

Regulatory T (Treg) cells are critical in inducing and maintaining tolerance. Despite progress in understanding the basis of immune tolerance, mechanisms and molecules involved in the generation of Treg cells remain poorly understood. IFN regulatory factor (IRF)-1 is a pleiotropic transcription factor implicated in the regulation of various immune processes. In this study, we report that IRF-1 negatively regulates CD4(+)CD25(+) Treg cell development and function by specifically repressing Foxp3 expression. IRF-1-deficient (IRF-1(-/-)) mice showed a selective and marked increase of highly activated and differentiated CD4(+)CD25(+)Foxp3(+) Treg cells in thymus and in all peripheral lymphoid organs. Furthermore, IRF-1(-/-) CD4(+)CD25(-) T cells showed extremely high bent to differentiate into CD4(+)CD25(+)Foxp3(+) Treg cells, whereas restoring IRF-1 expression in IRF-1(-/-) CD4(+)CD25(-) T cells impaired their differentiation into CD25(+)Foxp3(+) cells. Functionally, both isolated and TGF-beta-induced CD4(+)CD25(+) Treg cells from IRF-1(-/-) mice exhibited more increased suppressive activity than wild-type Treg cells. Such phenotype and functional characteristics were explained at a mechanistic level by the finding that IRF-1 binds a highly conserved IRF consensus element sequence (IRF-E) in the foxp3 gene promoter in vivo and negatively regulates its transcriptional activity. We conclude that IRF-1 is a key negative regulator of CD4(+)CD25(+) Treg cells through direct repression of Foxp3 expression.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Diferenciação Celular/imunologia , Fatores de Transcrição Forkhead/metabolismo , Fator Regulador 1 de Interferon/metabolismo , Subunidade alfa de Receptor de Interleucina-2/imunologia , Animais , Linfócitos T CD4-Positivos/citologia , Células Cultivadas , Sequência Consenso , Regulação para Baixo , Fatores de Transcrição Forkhead/genética , Humanos , Fator Regulador 1 de Interferon/deficiência , Fator Regulador 1 de Interferon/genética , Camundongos , Camundongos Knockout , Regiões Promotoras Genéticas/genética , Ligação Proteica , Transcrição Gênica/genética
10.
Ann Ist Super Sanita ; 56(4): 409-418, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33346166

RESUMO

Plasmodium vivax has been considered for years as responsible for a mild form of malaria, due to the absence in the majority of its infections of the severe form of the disease, typical instead of the deadly human parasite P. falciparum. In the last decade, studies on vivax malaria have had a partial step ahead especially after the completion of the whole genome project, but there is still a gap of knowledge in the biology of this parasite. Moreover, the emergence of P. vivax antimalarial resistance in 1980s and its subsequent spread in the Southeast Asia have indicated new concerns about the possibility to control this parasite. P. vivax drug resistance poses a major threat to endemic countries and without important international efforts, we could assist in a near future to the paradox of seeing different malaria co-endemic countries, that have successfully controlled/eliminated P. falciparum, still fighting against P. vivax.


Assuntos
Antimaláricos/farmacologia , Antimaláricos/uso terapêutico , Malária Vivax/tratamento farmacológico , Plasmodium vivax/efeitos dos fármacos , Resistência a Medicamentos , Humanos
11.
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
12.
Parasit Vectors ; 13(1): 137, 2020 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-32171330

RESUMO

BACKGROUND: Routine molecular surveillance for imported drug-resistant malaria parasites to the USA and European Union is an important public health activity. The obtained molecular data are used to help keep chemoprophylaxis and treatment guidelines up to date for persons traveling to malaria endemic countries. Recent advances in next-generation sequencing (NGS) technologies provide a new and effective way of tracking malaria drug-resistant parasites. METHODS: As part of a technology transfer arrangement between the CDC Malaria Branch and the Istituto Superiore di Sanità (ISS), Rome, Italy, the recently described Malaria Resistance Surveillance (MaRS) protocol was used to genotype 148 Plasmodium falciparum isolates from Eritrea for kelch 13 (k13) and cytochrome b (cytb) genes, molecular markers associated with resistance to artemisinin (ART) and atovaquone/proguanil (AP), respectively. RESULTS: Spanning the full-length k13 gene, seven non-synonymous single nucleotide polymorphisms (SNPs) were found (K189N, K189T, E208K, D281V, E401Q, R622I and T535M), of which none have been associated with artemisinin resistance. No mutations were found in cytochrome b. CONCLUSION: All patients successfully genotyped carried parasites susceptible to ART and AP treatment. Future studies between CDC Malaria Branch and ISS are planned to expand the MaRS system, including data sharing, in an effort to maintain up to date treatment guidelines for travelers to malaria endemic countries.


Assuntos
Citocromos b/genética , Resistência a Medicamentos/genética , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Plasmodium falciparum/genética , Plasmodium falciparum/isolamento & purificação , Proteínas de Protozoários/genética , África , Anti-Infecciosos/farmacologia , Antimaláricos/farmacologia , Artemisininas , Atovaquona/farmacologia , DNA de Protozoário/genética , Proteínas de Drosophila , Combinação de Medicamentos , Genótipo , Humanos , Itália , Malária Falciparum/parasitologia , Malária Falciparum/prevenção & controle , Proteínas dos Microfilamentos/genética , Plasmodium falciparum/efeitos dos fármacos , Polimorfismo de Nucleotídeo Único , Prevalência , Proguanil/farmacologia , Viagem
13.
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
14.
Biochem J ; 396(2): 371-80, 2006 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-16512786

RESUMO

The Tat protein is the transcriptional activator of HIV-1 gene expression, which is not only essential for viral replication, but also important in the complex HIV-induced pathogenesis of AIDS, as both an intracellular and an extracellular released protein. Accordingly, Tat is able to profoundly affect cellular gene expression, regulating several cellular functions, also in non-infected cells. We showed recently that Tat induces modification of immunoproteasomes in that it up-regulates LMP7 (low-molecular-mass polypeptide 7) and MECL1 (multicatalytic endopeptidase complex-like 1) subunits and down-modulates the LMP2 subunit, resulting in a change in the generation and presentation of epitopes in the context of MHC class I. In particular, Tat increases presentation of subdominant and cryptic epitopes. In the present study, we investigated the molecular mechanism responsible for the Tat-induced LMP2 down-regulation and show that intracellular Tat represses transcription of the LMP2 gene by competing with STAT1 (signal transducer and activator of transcription 1) for binding to IRF-1 (interferon-regulatory factor-1) on the overlapping ICS-2 (interferon consensus sequence-2)-GAS (gamma-interferon-activated sequence) present in the LMP2 promoter. This element is constitutively occupied in vivo by the unphosphorylated STAT1-IRF-1 complex, which is responsible for the basal transcription of the gene. Sequestration of IRF-1 by intracellular Tat impairs the formation of the complex resulting in lower LMP2 gene transcription and LMP2 protein expression, which is associated with increased proteolytic activity. On the other hand, extracellular Tat induces the expression of LMP2. These effects of Tat provide another effective mechanism by which HIV-1 affects antigen presentation in the context of the MHC class I complex and may have important implications in the use of Tat for vaccination strategies.


Assuntos
Cisteína Endopeptidases/metabolismo , Repressão Enzimática , Produtos do Gene tat/metabolismo , Produtos do Gene tat/farmacologia , Fator Regulador 1 de Interferon/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Fator de Transcrição STAT1/metabolismo , Transcrição Gênica , Cisteína Endopeptidases/genética , Regulação da Expressão Gênica , Produtos do Gene tat/genética , Humanos , Fator Regulador 1 de Interferon/antagonistas & inibidores , Fator Regulador 1 de Interferon/genética , Interferon gama/metabolismo , Células Jurkat , Modelos Biológicos , Complexos Multienzimáticos/genética , Complexos Multienzimáticos/metabolismo , Regiões Promotoras Genéticas , Complexo de Endopeptidases do Proteassoma/genética , Ligação Proteica , Fator de Transcrição STAT1/antagonistas & inibidores , Fator de Transcrição STAT1/genética , Fatores de Tempo , Transfecção
15.
J Interferon Cytokine Res ; 36(7): 414-32, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27379864

RESUMO

The interferon regulatory factor (IRF) family consists of transcriptional regulators that exert multifaceted and versatile functions in multiple biological processes. Their crucial role as central mediators in the establishment and execution of host immunity in response to pathogen-derived signals downstream pattern recognition receptors (PRRs) makes IRFs a hallmark of the host antiviral response. They function as hub molecules at the crossroad of different signaling pathways for the induction of interferon (IFN) and inflammatory cytokines, as well as of antiviral and immunomodulatory genes even in an IFN-independent manner. By regulating the development and activity of immune cells, IRFs also function as a bridge between innate and adaptive responses. As such, IRFs represent attractive and compulsive targets in viral strategies to subvert antiviral signaling. In this study, we discuss current knowledge on the wide array of strategies put in place by pathogenic viruses to evade, subvert, and/or hijack these essential components of host antiviral immunity.


Assuntos
Interações Hospedeiro-Patógeno/imunologia , Imunidade Inata , Fatores Reguladores de Interferon/metabolismo , Viroses/imunologia , Viroses/metabolismo , Vírus/imunologia , Animais , Regulação da Expressão Gênica , Interações Hospedeiro-Patógeno/genética , Humanos , Fatores Reguladores de Interferon/genética , Família Multigênica , Transdução de Sinais , Viroses/genética , Viroses/virologia
16.
mBio ; 7(5)2016 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-27795392

RESUMO

In addition to its ability to regulate HIV-1 promoter activation, the viral transactivator Tat also functions as a determinant of pathogenesis and disease progression by directly and indirectly modulating the host anti-HIV response, largely through the capacity of Tat to interact with and modulate the activities of multiple host proteins. We previously demonstrated that Tat modulated both viral and host transcriptional machinery by interacting with the cellular transcription factor interferon regulatory factor 1 (IRF-1). In the present study, we investigated the mechanistic basis and functional significance of Tat-IRF-1 interaction and demonstrate that Tat dramatically decreased IRF-1 protein stability. To accomplish this, Tat exploited the cellular HDM2 (human double minute 2 protein) ubiquitin ligase to accelerate IRF-1 proteasome-mediated degradation, resulting in a quenching of IRF-1 transcriptional activity during HIV-1 infection. These data identify IRF-1 as a new target of Tat-induced modulation of the cellular protein machinery and reveal a new strategy developed by HIV-1 to evade host immune responses. IMPORTANCE: Current therapies have dramatically reduced morbidity and mortality associated with HIV infection and have converted infection from a fatal pathology to a chronic disease that is manageable via antiretroviral therapy. Nevertheless, HIV-1 infection remains a challenge, and the identification of useful cellular targets for therapeutic intervention remains a major goal. The cellular transcription factor IRF-1 impacts various physiological functions, including the immune response to viral infection. In this study, we have identified a unique mechanism by which HIV-1 evades IRF-1-mediated host immune responses and show that the viral protein Tat accelerates IRF-1 proteasome-mediated degradation and inactivates IRF-1 function. Restoration of IRF-1 functionality may thus be regarded as a potential strategy to reinstate both a direct antiviral response and a more broadly acting immune regulatory circuit.


Assuntos
HIV-1/patogenicidade , Interações Hospedeiro-Patógeno , Fator Regulador 1 de Interferon/metabolismo , Processamento de Proteína Pós-Traducional , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Produtos do Gene tat do Vírus da Imunodeficiência Humana/metabolismo , Linhagem Celular , Humanos , Ligação Proteica , Ubiquitinação
17.
Oncogene ; 21(52): 7933-44, 2002 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-12439744

RESUMO

High expression of transferrin receptor (TfR) on the membrane of erythroid cells accounts for the high level of iron required to sustain heme synthesis. Several studies indicate that during erythroid differentiation TfR expression is highly dependent on transcriptional regulation. In this study we characterized the minimal region able to confer transcriptional regulation during erythroid differentiation in Friend leukemia cells (FLC). This region of 120 bp, upstream the transcription start site, contains an overlapping consensus recognition sequence for AP1/CREB/ATF transcription factors and for proteins of the Ets family and a GC rich region. Here, we report that both the Ets and the Ap1/CRE like sites are essential for promoter activity during erythroid differentiation. We showed that Ets-1 binds to the EBS-TfR and its binding activity decreases in FLC induced to differentiate and during normal erythroid differentiation. Consistent with this, FLC constitutively expressing Ets-1 show a decrease in TfR gene expression, globin mRNA and hemoglobin synthesis. We conclude that Ets-1 binding activity is modulated during erythroid maturation and that a deregulated expression of this transcription factor interferes with terminal erythroid differentiation.


Assuntos
Diferenciação Celular/fisiologia , Eritrócitos/química , Regulação da Expressão Gênica/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Receptores da Transferrina/genética , Fatores de Transcrição/fisiologia , Transcrição Gênica/fisiologia , Sequência de Bases , Células Cultivadas , DNA , Humanos , Dados de Sequência Molecular , Regiões Promotoras Genéticas , Ligação Proteica , Proteína Proto-Oncogênica c-ets-1 , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-ets , Fatores de Transcrição/metabolismo , Células Tumorais Cultivadas
18.
Cytokine Growth Factor Rev ; 26(2): 143-58, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25466629

RESUMO

For more than 50 years, Type I Interferon (IFN) has been recognized as critical in controlling viral infections. IFN is produced downstream germ-line encoded pattern recognition receptors (PRRs) upon engagement by pathogen-associated molecular patterns (PAMPs). As a result, hundreds of different interferon-stimulated genes (ISGs) are rapidly induced, acting in both autocrine and paracrine manner to build a barrier against viral replication and spread. ISGs encode proteins with direct antiviral and immunomodulatory activities affecting both innate and adaptive immune responses. During infection with viruses, as HIV-1, that can establish a persistent infection, IFN although produced, is not able to block the initial infection and a chronic IFN-mediated immune activation/inflammation becomes a pathogenic mechanism of disease progression. This review will briefly summarize when and how IFN is produced during HIV-1 infection and the way this innate immune response is manipulated by the virus to its own advantage to drive chronic immune activation and progression to AIDS.


Assuntos
Infecções por HIV/imunologia , HIV-1/imunologia , HIV-1/patogenicidade , Interferon Tipo I/imunologia , Progressão da Doença , Infecções por HIV/complicações , Infecções por HIV/virologia , Humanos , Evasão da Resposta Imune , Imunidade Inata , Inflamação , Fatores Reguladores de Interferon/imunologia , Interferon Tipo I/biossíntese , Moléculas com Motivos Associados a Patógenos/metabolismo , Proteínas Virais/imunologia , Replicação Viral
19.
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
20.
Cytokine Growth Factor Rev ; 23(4-5): 255-70, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22748237

RESUMO

Thirty years after the first isolation of the etiological agent of AIDS, the virus HIV-1 is still a major threat worldwide with millions of individuals currently infected. Although current combination therapies allow viral replication to be controlled, HIV-1 is not eradicated and persists in drug- and immune system-insensitive reservoirs and a cure is still lacking. Pathogens such as HIV-1 that cause chronic infections are able to adapt to the host in a manner that ensures long term residence and survival, via the evolution of numerous mechanisms that evade various aspects of the innate and adaptive immune response. One such mechanism is targeted to members of the interferon (IFN) regulatory factor (IRF) family of proteins. These transcription factors regulate a variety of biological processes including interferon induction, immune cell activation and downstream pattern recognition receptors (PRRs). HIV-1 renders IRFs harmless and hijacks them to its own advantage in order to facilitate its replication and evasion of immune responses. Type I interferon (IFN), the canonical antiviral innate response, can be induced in both acute and chronic HIV-1 infection in vivo, but in the majority of individuals this initial response is not protective and can contribute to disease progression. Type I IFN expression is largely inhibited in T cells and macrophages in order to successfully establish productive infection, whereas sustained IFN production by plasmacytoid dendritic cells is considered an important source of chronic immune activation, a hallmark to AIDS progression.


Assuntos
Infecções por HIV/imunologia , HIV-1/imunologia , Fatores Reguladores de Interferon/imunologia , Interferons/imunologia , Infecções por HIV/metabolismo , Infecções por HIV/virologia , HIV-1/fisiologia , Interações Hospedeiro-Patógeno/imunologia , Humanos , Evasão da Resposta Imune/imunologia , Imunidade Inata/imunologia , Fatores Reguladores de Interferon/metabolismo , Interferons/metabolismo , Modelos Imunológicos , Transdução de Sinais/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA