Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 120(47): e2308355120, 2023 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-37963251

RESUMO

A detailed understanding of the innate immune mechanisms involved in restricting SARS-CoV-2 infection and how the virus disrupts these processes could reveal new strategies to boost antiviral mechanisms and develop therapeutics for COVID-19. Here, we identify cellular nucleic acid-binding protein (CNBP) as a key host factor controlling SARS-CoV-2 infection. In response to RNA-sensing pathways, CNBP is phosphorylated and translocates from the cytosol to the nucleus where it binds to the interferon-ß enhancer to initiate transcription. Because SARS-CoV-2 evades immune detection by the host's RNA-sensing pathways, CNBP is largely retained in the cytosol where it restricts SARS-CoV-2 directly, leading to a battle between the host and SARS-CoV-2 that extends beyond antiviral immune signaling pathways. We further demonstrated that CNBP binds SARS-CoV-2 viral RNA directly and competes with the viral nucleocapsid protein to prevent viral RNA and nucleocapsid protein from forming liquid-liquid phase separation (LLPS) condensates critical for viral replication. Consequently, cells and animals lacking CNBP have higher viral loads, and CNBP-deficient mice succumb rapidly to infection. Altogether, these findings identify CNBP as a key antiviral factor for SARS-CoV-2, functioning both as a regulator of antiviral IFN gene expression and a cell-intrinsic restriction factor that disrupts LLPS to limit viral replication and spread. In addition, our studies also highlight viral condensates as important targets and strategies for the development of drugs to combat COVID-19.


Assuntos
COVID-19 , Interferons , Animais , Camundongos , Proteínas do Nucleocapsídeo , RNA Viral/genética , RNA Viral/metabolismo , SARS-CoV-2/fisiologia , Fatores de Transcrição , Replicação Viral
2.
J Clin Invest ; 132(11)2022 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-35642634

RESUMO

Plasmodium falciparum (P. falciparum) induces trained innate immune responses in vitro, where initial stimulation of adherent PBMCs with P. falciparum-infected RBCs (iRBCs) results in hyperresponsiveness to subsequent ligation of TLR2. This response correlates with the presence of T and B lymphocytes in adherent PBMCs, suggesting that innate immune training is partially due to adaptive immunity. We found that T cell-depleted PBMCs and purified monocytes alone did not elicit hyperproduction of IL-6 and TNF-α under training conditions. Analysis of P. falciparum-trained PBMCs showed that DCs did not develop under control conditions, and IL-6 and TNF-α were primarily produced by monocytes and DCs. Transwell experiments isolating purified monocytes from either PBMCs or purified CD4+ T cells, but allowing diffusion of secreted proteins, enabled monocytes trained with iRBCs to hyperproduce IL-6 and TNF-α after TLR restimulation. Purified monocytes stimulated with IFN-γ hyperproduced IL-6 and TNF-α, whereas blockade of IFN-γ in P. falciparum-trained PBMCs inhibited trained responses. Assay for transposase-accessible chromatin with high-throughput sequencing (ATAC-Seq) on monocytes from patients with malaria showed persistently open chromatin at genes that appeared to be trained in vitro. Together, these findings indicate that the trained immune response of monocytes to P. falciparum is not completely cell intrinsic but depends on soluble signals from lymphocytes.


Assuntos
Linfócitos , Malária Falciparum , Monócitos , Cromatina , Humanos , Interleucina-6/genética , Linfócitos/imunologia , Malária Falciparum/imunologia , Monócitos/imunologia , Plasmodium falciparum , Fator de Necrose Tumoral alfa/metabolismo
3.
PLoS One ; 16(1): e0238753, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33481821

RESUMO

PFRED a software application for the design, analysis, and visualization of antisense oligonucleotides and siRNA is described. The software provides an intuitive user-interface for scientists to design a library of siRNA or antisense oligonucleotides that target a specific gene of interest. Moreover, the tool facilitates the incorporation of various design criteria that have been shown to be important for stability and potency. PFRED has been made available as an open-source project so the code can be easily modified to address the future needs of the oligonucleotide research community. A compiled version is available for downloading at https://github.com/pfred/pfred-gui/releases/tag/v1.0 as a java Jar file. The source code and the links for downloading the precompiled version can be found at https://github.com/pfred.


Assuntos
Biologia Computacional/métodos , Primers do DNA/genética , Oligonucleotídeos Antissenso/genética , Algoritmos , Biblioteca Gênica , Genômica , Oligonucleotídeos/genética , RNA Interferente Pequeno/genética , Software , Interface Usuário-Computador
4.
Proc Natl Acad Sci U S A ; 115(36): 9002-9007, 2018 09 04.
Artigo em Inglês | MEDLINE | ID: mdl-30127003

RESUMO

Alzheimer's disease (AD) is characterized by the progressive destruction and dysfunction of central neurons. AD patients commonly have unprovoked seizures compared with age-matched controls. Amyloid peptide-related inflammation is thought to be an important aspect of AD pathogenesis. We previously reported that NLRP3 inflammasome KO mice, when bred into APPswe/PS1ΔE9 (APP/PS1) mice, are completely protected from amyloid-induced AD-like disease, presumably because they cannot produce mature IL1ß or IL18. To test the role of IL18, we bred IL18KO mice with APP/PS1 mice. Surprisingly, IL18KO/APP/PS1 mice developed a lethal seizure disorder that was completely reversed by the anticonvulsant levetiracetam. IL18-deficient AD mice showed a lower threshold in chemically induced seizures and a selective increase in gene expression related to increased neuronal activity. IL18-deficient AD mice exhibited increased excitatory synaptic proteins, spine density, and basal excitatory synaptic transmission that contributed to seizure activity. This study identifies a role for IL18 in suppressing aberrant neuronal transmission in AD.


Assuntos
Doença de Alzheimer/metabolismo , Amiloide/metabolismo , Inflamassomos/metabolismo , Interleucina-18/metabolismo , Convulsões/metabolismo , Transmissão Sináptica , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Amiloide/genética , Animais , Inflamassomos/genética , Interleucina-18/genética , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Levetiracetam , Camundongos , Camundongos Knockout , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Piracetam/análogos & derivados , Piracetam/farmacologia , Convulsões/tratamento farmacológico , Convulsões/genética , Convulsões/patologia
5.
J Mol Biol ; 430(8): 1098-1115, 2018 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-29466705

RESUMO

The fitness effects of synonymous mutations can provide insights into biological and evolutionary mechanisms. We analyzed the experimental fitness effects of all single-nucleotide mutations, including synonymous substitutions, at the beginning of the influenza A virus hemagglutinin (HA) gene. Many synonymous substitutions were deleterious both in bulk competition and for individually isolated clones. Investigating protein and RNA levels of a subset of individually expressed HA variants revealed that multiple biochemical properties contribute to the observed experimental fitness effects. Our results indicate that a structural element in the HA segment viral RNA may influence fitness. Examination of naturally evolved sequences in human hosts indicates a preference for the unfolded state of this structural element compared to that found in swine hosts. Our overall results reveal that synonymous mutations may have greater fitness consequences than indicated by simple models of sequence conservation, and we discuss the implications of this finding for commonly used evolutionary tests and analyses.


Assuntos
Aptidão Genética , Glicoproteínas de Hemaglutininação de Vírus da Influenza/química , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Vírus da Influenza A Subtipo H1N1/crescimento & desenvolvimento , Mutação Silenciosa , Substituição de Aminoácidos , Animais , Cães , Evolução Molecular , Células HEK293 , Humanos , Vírus da Influenza A Subtipo H1N1/genética , Células Madin Darby de Rim Canino , Modelos Moleculares , Filogenia , Dobramento de RNA , Suínos , Replicação Viral
6.
J Immunol ; 199(1): 263-270, 2017 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-28539433

RESUMO

Tracheal cytotoxin (TCT), a monomer of DAP-type peptidoglycan from Bordetella pertussis, causes cytopathology in the respiratory epithelia of mammals and robustly triggers the Drosophila Imd pathway. PGRP-LE, a cytosolic innate immune sensor in Drosophila, directly recognizes TCT and triggers the Imd pathway, yet the mechanisms by which TCT accesses the cytosol are poorly understood. In this study, we report that CG8046, a Drosophila SLC46 family transporter, is a novel transporter facilitating cytosolic recognition of TCT, and plays a crucial role in protecting flies against systemic Escherichia coli infection. In addition, mammalian SLC46A2s promote TCT-triggered NOD1 activation in human epithelial cell lines, indicating that SLC46As is a conserved group of peptidoglycan transporter contributing to cytosolic immune recognition.


Assuntos
Citosol/imunologia , Proteínas de Drosophila/metabolismo , Imunidade Inata , Peptidoglicano/imunologia , Simportadores/metabolismo , Fatores de Virulência de Bordetella/imunologia , Animais , Linhagem Celular , Parede Celular/imunologia , Parede Celular/metabolismo , Citosol/metabolismo , Drosophila/imunologia , Drosophila/microbiologia , Escherichia coli/fisiologia , Células HEK293 , Humanos , Peptidoglicano/química , Peptidoglicano/metabolismo , Transdução de Sinais , Fatores de Virulência de Bordetella/química , Fatores de Virulência de Bordetella/metabolismo
7.
J Biol Chem ; 292(14): 5634-5644, 2017 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-28209713

RESUMO

Bacterial sepsis involves a complex interaction between the host immune response and bacterial LPS. LPS binds Toll-like receptor (TLR) 4, which leads to the release of proinflammatory cytokines that are essential for a potent innate immune response against pathogens. The innate immune system is tightly regulated, as excessive inflammation can lead to organ failure and death. MicroRNAs have recently emerged as important regulators of the innate immune system. Here we determined the function of miR-718, which is conserved across mammals and overlaps with the 5' UTR of the interleukin 1 receptor-associated kinase (IRAK1) gene. As IRAK1 is a key component of innate immune signaling pathways that are downstream of most TLRs, we hypothesized that miR-718 helps regulate the innate immune response. Activation of TLR4, but not TLR3, induced the expression of miR-718 in macrophages. miR-718 expression was also induced in the spleens of mice upon LPS injection. miR-718 modulates PI3K/Akt signaling by directly down-regulating phosphatase and tensin homolog (PTEN), thereby promoting phosphorylation of Akt, which leads to a decrease in proinflammatory cytokine production. Phosphorylated Akt induces let-7e expression, which, in turn, down-regulates TLR4 and further diminishes TLR4-mediated proinflammatory signals. Decreased miR-718 expression is associated with bacterial burden during Neisseria gonorrhoeae infection and alters the infection dynamics of N. gonorrhoeae in vitro Furthermore, miR-718 regulates the induction of LPS tolerance in macrophages. We propose a role for miR-718 in controlling TLR4 signaling and inflammatory cytokine signaling through a negative feedback regulation loop involving down-regulation of TLR4, IRAK1, and NF-κB.


Assuntos
Regiões 5' não Traduzidas , Citocinas/metabolismo , Macrófagos/metabolismo , MicroRNAs/metabolismo , PTEN Fosfo-Hidrolase/metabolismo , Transdução de Sinais , Animais , Citocinas/genética , Gonorreia/genética , Gonorreia/metabolismo , Humanos , Quinases Associadas a Receptores de Interleucina-1/genética , Quinases Associadas a Receptores de Interleucina-1/metabolismo , Macrófagos/microbiologia , Macrófagos/patologia , Camundongos , Camundongos Knockout , MicroRNAs/genética , Neisseria gonorrhoeae/metabolismo , PTEN Fosfo-Hidrolase/genética , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor 3 Toll-Like/genética , Receptor 3 Toll-Like/metabolismo , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/metabolismo
8.
Annu Rev Immunol ; 35: 177-198, 2017 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-28125358

RESUMO

The discovery of long noncoding RNAs (lncRNA) has provided a new perspective on gene regulation in diverse biological contexts. lncRNAs are remarkably versatile molecules that interact with RNA, DNA, or proteins to promote or restrain the expression of protein-coding genes. Activation of immune cells is associated with dynamic changes in expression of genes, the products of which combat infectious microorganisms, initiate repair, and resolve inflammatory responses in cells and tissues. Recent evidence indicates that lncRNAs play important roles in directing the development of diverse immune cells and controlling the dynamic transcriptional programs that are a hallmark of immune cell activation. The importance of these molecules is underscored by their newly recognized roles in inflammatory diseases. In this review, we discuss the contribution of lncRNAs in the development and activation of immune cells and their roles in immune-related diseases. We also discuss challenges faced in identifying biological functions for this large and complex class of genes.


Assuntos
Doenças do Sistema Imunitário/genética , Imunidade/genética , RNA Longo não Codificante/imunologia , Animais , Regulação da Expressão Gênica , Humanos
9.
Cell ; 165(7): 1672-1685, 2016 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-27315481

RESUMO

Long intergenic noncoding RNAs (lincRNAs) are important regulators of gene expression. Although lincRNAs are expressed in immune cells, their functions in immunity are largely unexplored. Here, we identify an immunoregulatory lincRNA, lincRNA-EPS, that is precisely regulated in macrophages to control the expression of immune response genes (IRGs). Transcriptome analysis of macrophages from lincRNA-EPS-deficient mice, combined with gain-of-function and rescue experiments, revealed a specific role for this lincRNA in restraining IRG expression. Consistently, lincRNA-EPS-deficient mice manifest enhanced inflammation and lethality following endotoxin challenge in vivo. lincRNA-EPS localizes at regulatory regions of IRGs to control nucleosome positioning and repress transcription. Further, lincRNA-EPS mediates these effects by interacting with heterogeneous nuclear ribonucleoprotein L via a CANACA motif located in its 3' end. Together, these findings identify lincRNA-EPS as a repressor of inflammatory responses, highlighting the importance of lincRNAs in the immune system.


Assuntos
Regulação da Expressão Gênica , Inflamação/genética , Macrófagos/imunologia , RNA Longo não Codificante/metabolismo , Animais , Cromátides/metabolismo , Deleção de Genes , Humanos , Listeria monocytogenes/fisiologia , Listeriose/imunologia , Macrófagos/metabolismo , Macrófagos/microbiologia , Macrófagos/virologia , Camundongos , Camundongos Endogâmicos C57BL , RNA Longo não Codificante/genética , Infecções por Respirovirus/imunologia , Vírus Sendai/fisiologia , Receptores Toll-Like/metabolismo , Transcriptoma
10.
Nat Commun ; 7: 10507, 2016 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-26856261

RESUMO

Ticks transmit more pathogens to humans and animals than any other arthropod. We describe the 2.1 Gbp nuclear genome of the tick, Ixodes scapularis (Say), which vectors pathogens that cause Lyme disease, human granulocytic anaplasmosis, babesiosis and other diseases. The large genome reflects accumulation of repetitive DNA, new lineages of retro-transposons, and gene architecture patterns resembling ancient metazoans rather than pancrustaceans. Annotation of scaffolds representing ∼57% of the genome, reveals 20,486 protein-coding genes and expansions of gene families associated with tick-host interactions. We report insights from genome analyses into parasitic processes unique to ticks, including host 'questing', prolonged feeding, cuticle synthesis, blood meal concentration, novel methods of haemoglobin digestion, haem detoxification, vitellogenesis and prolonged off-host survival. We identify proteins associated with the agent of human granulocytic anaplasmosis, an emerging disease, and the encephalitis-causing Langat virus, and a population structure correlated to life-history traits and transmission of the Lyme disease agent.


Assuntos
Anaplasma phagocytophilum , Vetores Aracnídeos/genética , Genoma/genética , Ixodes/genética , Canais Iônicos de Abertura Ativada por Ligante/genética , Animais , Perfilação da Expressão Gênica , Genômica , Doença de Lyme/transmissão , Oócitos , Xenopus laevis
11.
J Mol Biol ; 428(3): 538-553, 2016 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-26656922

RESUMO

The therapeutic benefits of the neuraminidase (NA) inhibitor oseltamivir are dampened by the emergence of drug resistance mutations in influenza A virus (IAV). To investigate the mechanistic features that underlie resistance, we developed an approach to quantify the effects of all possible single-nucleotide substitutions introduced into important regions of NA. We determined the experimental fitness effects of 450 nucleotide mutations encoding positions both surrounding the active site and at more distant sites in an N1 strain of IAV in the presence and absence of oseltamivir. NA mutations previously known to confer oseltamivir resistance in N1 strains, including H275Y and N295S, were adaptive in the presence of drug, indicating that our experimental system captured salient features of real-world selection pressures acting on NA. We identified mutations, including several at position 223, that reduce the apparent affinity for oseltamivir in vitro. Position 223 of NA is located adjacent to a hydrophobic portion of oseltamivir that is chemically distinct from the substrate, making it a hotspot for substitutions that preferentially impact drug binding relative to substrate processing. Furthermore, two NA mutations, K221N and Y276F, each reduce susceptibility to oseltamivir by increasing NA activity without altering drug binding. These results indicate that competitive expansion of IAV in the face of drug pressure is mediated by a balance between inhibitor binding and substrate processing.


Assuntos
Antivirais/farmacologia , Farmacorresistência Viral , Vírus da Influenza A/efeitos dos fármacos , Influenza Humana/tratamento farmacológico , Neuraminidase/genética , Oseltamivir/farmacologia , Proteínas Virais/genética , Animais , Linhagem Celular , Cães , Inibidores Enzimáticos/farmacologia , Humanos , Vírus da Influenza A/enzimologia , Vírus da Influenza A/genética , Influenza Humana/virologia , Modelos Moleculares , Neuraminidase/metabolismo , Infecções por Orthomyxoviridae/tratamento farmacológico , Infecções por Orthomyxoviridae/virologia , Mutação Puntual , Proteínas Virais/metabolismo
12.
RNA ; 22(2): 254-64, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26654912

RESUMO

Approximately 75% of the human genome is transcribed and many of these spliced transcripts contain primate-specific Alu elements, the most abundant mobile element in the human genome. The majority of exonized Alu elements are located in long noncoding RNAs (lncRNAs) and the untranslated regions of mRNA, with some performing molecular functions. To further assess the potential for Alu elements to be repurposed as functional RNA domains, we investigated the distribution and evolution of Alu elements in spliced transcripts. Our analysis revealed that Alu elements are underrepresented in mRNAs and lncRNAs, suggesting that most exonized Alu elements arising in the population are rare or deleterious to RNA function. When mRNAs and lncRNAs retain exonized Alu elements, they have a clear preference for Alu dimers, left monomers, and right monomers. mRNAs often acquire Alu elements when their genes are duplicated within Alu-rich regions. In lncRNAs, reverse-oriented Alu elements are significantly enriched and are not restricted to the 3' and 5' ends. Both lncRNAs and mRNAs primarily contain the Alu J and S subfamilies that were amplified relatively early in primate evolution. Alu J subfamilies are typically overrepresented in lncRNAs, whereas the Alu S dimer is overrepresented in mRNAs. The sequences of Alu dimers tend to be constrained in both lncRNAs and mRNAs, whereas the left and right monomers are constrained within particular Alu subfamilies and classes of RNA. Collectively, these findings suggest that Alu-containing RNAs are capable of forming stable structures and that some of these Alu domains might have novel biological functions.


Assuntos
Regiões 3' não Traduzidas , Regiões 5' não Traduzidas , Elementos Alu , Genoma Humano , RNA Longo não Codificante/química , Biologia Computacional/métodos , Evolução Molecular , Éxons , Humanos , RNA Longo não Codificante/genética
13.
J Immunol ; 195(4): 1359-63, 2015 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-26179904

RESUMO

Natural antisense transcripts (NATs) are a class of long noncoding RNAs (lncRNAs) that are complementary to other protein-coding genes. Although thousands of NATs are encoded by mammalian genomes, their functions in innate immunity are unknown. In this study, we identified and characterized a novel NAT, AS-IL1α, which is partially complementary to IL-1α. Similar to IL-1α, AS-IL1α is expressed at low levels in resting macrophages and is induced following infection with Listeria monocytogenes or stimulation with TLR ligands (Pam3CSK4, LPS, polyinosinic-polycytidylic acid). Inducible expression of IL-1α mRNA and protein were significantly reduced in macrophages expressing shRNA that target AS-IL1α. AS-IL1α is located in the nucleus and did not alter the stability of IL-1α mRNA. Instead, AS-IL1α was required for the recruitment of RNA polymerase II to the IL-1α promoter. In summary, our studies identify AS-IL1α as an important regulator of IL-1α transcription during the innate immune response.


Assuntos
Regulação da Expressão Gênica , Mediadores da Inflamação , Interleucina-1alfa/genética , RNA Antissenso/genética , RNA não Traduzido/genética , Transcrição Gênica , Animais , Linhagem Celular , Análise por Conglomerados , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Loci Gênicos , Ligantes , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , NF-kappa B/metabolismo , Interferência de RNA , Receptores Toll-Like/metabolismo
14.
Mol Biol Evol ; 32(6): 1519-32, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25713211

RESUMO

Influenza A virus (IAV) has a segmented genome that allows for the exchange of genome segments between different strains. This reassortment accelerates evolution by breaking linkage, helping IAV cross species barriers to potentially create highly virulent strains. Challenges associated with monitoring the process of reassortment in molecular detail have limited our understanding of its evolutionary implications. We applied a novel deep sequencing approach with quantitative analysis to assess the in vitro temporal evolution of genomic reassortment in IAV. The combination of H1N1 and H3N2 strains reproducibly generated a new H1N2 strain with the hemagglutinin and nucleoprotein segments originating from H1N1 and the remaining six segments from H3N2. By deep sequencing the entire viral genome, we monitored the evolution of reassortment, quantifying the relative abundance of all IAV genome segments from the two parent strains over time and measuring the selection coefficients of the reassorting segments. Additionally, we observed several mutations coemerging with reassortment that were not found during passaging of pure parental IAV strains. Our results demonstrate how reassortment of the segmented genome can accelerate viral evolution in IAV, potentially enabled by the emergence of a small number of individual mutations.


Assuntos
Alphainfluenzavirus/genética , Genoma Viral , Vírus Reordenados/genética , Seleção Genética , Animais , Biologia Computacional , Cães , Evolução Molecular , Frequência do Gene , Genótipo , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Sequenciamento de Nucleotídeos em Larga Escala , Vírus da Influenza A Subtipo H1N1/genética , Vírus da Influenza A Subtipo H1N2/genética , Vírus da Influenza A Subtipo H3N2/genética , Limite de Detecção , Células Madin Darby de Rim Canino , Nucleoproteínas/genética , Análise de Sequência de RNA
15.
PLoS Genet ; 10(2): e1004185, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24586206

RESUMO

The challenge of distinguishing genetic drift from selection remains a central focus of population genetics. Time-sampled data may provide a powerful tool for distinguishing these processes, and we here propose approximate Bayesian, maximum likelihood, and analytical methods for the inference of demography and selection from time course data. Utilizing these novel statistical and computational tools, we evaluate whole-genome datasets of an influenza A H1N1 strain in the presence and absence of oseltamivir (an inhibitor of neuraminidase) collected at thirteen time points. Results reveal a striking consistency amongst the three estimation procedures developed, showing strongly increased selection pressure in the presence of drug treatment. Importantly, these approaches re-identify the known oseltamivir resistance site, successfully validating the approaches used. Enticingly, a number of previously unknown variants have also been identified as being positively selected. Results are interpreted in the light of Fisher's Geometric Model, allowing for a quantification of the increased distance to optimum exerted by the presence of drug, and theoretical predictions regarding the distribution of beneficial fitness effects of contending mutations are empirically tested. Further, given the fit to expectations of the Geometric Model, results suggest the ability to predict certain aspects of viral evolution in response to changing host environments and novel selective pressures.


Assuntos
Farmacorresistência Viral/genética , Genética Populacional , Vírus da Influenza A Subtipo H1N1/genética , Seleção Genética , Teorema de Bayes , Deriva Genética , Humanos , Vírus da Influenza A Subtipo H1N1/efeitos dos fármacos , Influenza Humana/genética , Influenza Humana/virologia , Mutação , Oseltamivir/farmacologia
16.
Curr Opin Immunol ; 26: 140-6, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24556411

RESUMO

The differentiation and activation of both innate and adaptive immune cells is highly dependent on a coordinated set of transcriptional and post-transcriptional events. Chromatin-modifiers and transcription factors regulate the accessibility and transcription of immune genes, respectively. Immune cells also express miRNA and RNA-binding proteins that provide an additional layer of regulation at the mRNA level. However, long noncoding RNAs (lncRNAs), which have been primarily studied in the context of genomic imprinting, cancer, and cell differentiation, are now emerging as important regulators of immune cell differentiation and activation. In this review, we provide a brief overview of lncRNAs, their known functions in immunity, and discuss their potential to be more broadly involved in other aspects of the immune response.


Assuntos
Imunidade Adaptativa/genética , Imunidade Inata/genética , RNA Longo não Codificante/genética , RNA Longo não Codificante/imunologia , Animais , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Impressão Genômica/imunologia , Humanos , Íntrons/genética , Íntrons/imunologia , MicroRNAs/genética , MicroRNAs/imunologia , MicroRNAs/metabolismo , Neoplasias/genética , Neoplasias/imunologia , Splicing de RNA/genética , Splicing de RNA/imunologia , RNA Longo não Codificante/metabolismo , Transcrição Gênica/imunologia
17.
J Immunol ; 192(5): 2291-304, 2014 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-24477914

RESUMO

The transcriptional repressor B lymphocyte-induced maturation protein 1 (BLIMP1) is a master regulator of B and T cell differentiation. To examine the role of BLIMP1 in innate immunity, we used a conditional knockout (CKO) of Blimp1 in myeloid cells and found that Blimp1 CKO mice were protected from lethal infection induced by Listeria monocytogenes. Transcriptome analysis of Blimp1 CKO macrophages identified the murine chemokine (C-C motif) ligand 8, CCL8, as a direct target of Blimp1-mediated transcriptional repression in these cells. BLIMP1-deficient macrophages expressed elevated levels of Ccl8, and consequently Blimp1 CKO mice had higher levels of circulating CCL8, resulting in increased neutrophils in the peripheral blood, promoting a more aggressive antibacterial response. Mice lacking the Ccl8 gene were more susceptible to L. monocytogenes infection than were wild-type mice. Although CCL8 failed to recruit neutrophils directly, it was chemotactic for γ/δ T cells, and CCL8-responsive γ/δ T cells were enriched for IL-17F. Finally, CCL8-mediated enhanced clearance of L. monocytogenes was dependent on γ/δ T cells. Collectively, these data reveal an important role for BLIMP1 in modulating host defenses by suppressing expression of the chemokine CCL8.


Assuntos
Quimiocina CCL8/imunologia , Regulação da Expressão Gênica/imunologia , Listeria monocytogenes/imunologia , Listeriose/imunologia , Macrófagos/imunologia , Fatores de Transcrição/imunologia , Animais , Quimiocina CCL8/genética , Regulação da Expressão Gênica/genética , Listeriose/genética , Macrófagos/patologia , Camundongos , Camundongos Knockout , Neutrófilos/imunologia , Neutrófilos/patologia , Fator 1 de Ligação ao Domínio I Regulador Positivo , Receptores de Antígenos de Linfócitos T gama-delta/genética , Receptores de Antígenos de Linfócitos T gama-delta/imunologia , Linfócitos T/imunologia , Linfócitos T/patologia , Fatores de Transcrição/genética , Transcrição Gênica/genética , Transcrição Gênica/imunologia
18.
J Virol ; 88(1): 272-81, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24155392

RESUMO

Influenza A virus (IAV) is a major cause of morbidity and mortality throughout the world. Current antiviral therapies include oseltamivir, a neuraminidase inhibitor that prevents the release of nascent viral particles from infected cells. However, the IAV genome can evolve rapidly, and oseltamivir resistance mutations have been detected in numerous clinical samples. Using an in vitro evolution platform and whole-genome population sequencing, we investigated the population genomics of IAV during the development of oseltamivir resistance. Strain A/Brisbane/59/2007 (H1N1) was grown in Madin-Darby canine kidney cells with or without escalating concentrations of oseltamivir over serial passages. Following drug treatment, the H274Y resistance mutation fixed reproducibly within the population. The presence of the H274Y mutation in the viral population, at either a low or a high frequency, led to measurable changes in the neuraminidase inhibition assay. Surprisingly, fixation of the resistance mutation was not accompanied by alterations of viral population diversity or differentiation, and oseltamivir did not alter the selective environment. While the neighboring K248E mutation was also a target of positive selection prior to H274Y fixation, H274Y was the primary beneficial mutation in the population. In addition, once evolved, the H274Y mutation persisted after the withdrawal of the drug, even when not fixed in viral populations. We conclude that only selection of H274Y is required for oseltamivir resistance and that H274Y is not deleterious in the absence of the drug. These collective results could offer an explanation for the recent reproducible rise in oseltamivir resistance in seasonal H1N1 IAV strains in humans.


Assuntos
Antivirais/farmacologia , Farmacorresistência Viral/genética , Evolução Molecular , Genoma Viral , Vírus da Influenza A Subtipo H1N1/genética , Oseltamivir/farmacologia , Animais , Linhagem Celular , Cães , Ensaios de Triagem em Larga Escala , Técnicas In Vitro , Vírus da Influenza A Subtipo H1N1/crescimento & desenvolvimento , Concentração Inibidora 50 , Mutação , Ensaio de Placa Viral
19.
Gastroenterology ; 146(3): 754-764.e3, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24316261

RESUMO

BACKGROUND & AIMS: The alimentary tract contains a diffuse endocrine system comprising enteroendocrine cells that secrete peptides or biogenic amines to regulate digestion, insulin secretion, food intake, and energy homeostasis. Lineage analysis in the stomach revealed that a significant fraction of endocrine cells in the gastric corpus did not arise from Neurogenin3 (Neurog3)-expressing cells, unlike enteroendocrine cells elsewhere in the digestive tract. We aimed to isolate enriched serotonin-secreting and enterochromaffin-like (ECL) cells from the stomach and to clarify their cellular origin. METHODS: We used Neurogenic differentiation 1 (NeuroD1) and Neurog3 lineage analysis and examined the differentiation of serotonin-producing and ECL cells in stomach tissues of NeuroD1-cre;ROSA(tdTom), tryptophan hydroxylase 1 (Tph1)-cyan fluorescent protein (CFP), c-Kit(wsh/wsh), and Neurog3Cre;ROSA(tdTom) mice by immunohistochemistry. We used fluorescence-activated cell sorting to isolate each cell type for gene expression analysis. We also performed RNA sequencing analysis of ECL cells. RESULTS: Neither serotonin-secreting nor ECL cells of the corpus arose from cells expressing NeuroD1. Serotonin-secreting cells expressed a number of mast cell genes but not genes associated with endocrine differentiation; they did not develop in c-Kit(wsh/wsh) mice and were labeled with transplanted bone marrow cells. RNA sequencing analysis of ECL cells revealed high expression levels of many genes common to endocrine cells, including transcription factors, hormones, ion channels, and solute transporters but not markers of bone marrow cells. CONCLUSIONS: Serotonin-expressing cells of the gastric corpus of mice appear to be bone marrow-derived mucosal mast cells. Gene expression analysis of ECL cells indicated that they are endocrine cells of epithelial origin that do not express the same transcription factors as their intestinal enteroendocrine cell counterparts.


Assuntos
Linhagem da Célula , Células Enterocromafins/patologia , Células Enteroendócrinas/patologia , Serotonina/metabolismo , Estômago/patologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Células da Medula Óssea/metabolismo , Células da Medula Óssea/patologia , Diferenciação Celular , Células Enterocromafins/metabolismo , Células Enteroendócrinas/metabolismo , Mucosa Gástrica/metabolismo , Mastócitos/metabolismo , Mastócitos/patologia , Camundongos , Camundongos Transgênicos , Modelos Animais , Proteínas do Tecido Nervoso/metabolismo
20.
Science ; 341(6147): 789-92, 2013 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-23907535

RESUMO

An inducible program of inflammatory gene expression is central to antimicrobial defenses. This response is controlled by a collaboration involving signal-dependent activation of transcription factors, transcriptional co-regulators, and chromatin-modifying factors. We have identified a long noncoding RNA (lncRNA) that acts as a key regulator of this inflammatory response. Pattern recognition receptors such as the Toll-like receptors induce the expression of numerous lncRNAs. One of these, lincRNA-Cox2, mediates both the activation and repression of distinct classes of immune genes. Transcriptional repression of target genes is dependent on interactions of lincRNA-Cox2 with heterogeneous nuclear ribonucleoprotein A/B and A2/B1. Collectively, these studies unveil a central role of lincRNA-Cox2 as a broad-acting regulatory component of the circuit that controls the inflammatory response.


Assuntos
Regulação da Expressão Gênica , Imunidade Inata/genética , Inflamação/genética , Macrófagos/imunologia , Macrófagos/metabolismo , RNA Longo não Codificante/genética , Animais , Linhagem Celular , Núcleo Celular/metabolismo , Ciclo-Oxigenase 2/genética , Citocinas/genética , Citocinas/metabolismo , Citosol/metabolismo , Ribonucleoproteínas Nucleares Heterogêneas/metabolismo , Ativação de Macrófagos , Camundongos , Modelos Imunológicos , Interferência de RNA , RNA Longo não Codificante/metabolismo , Receptores Toll-Like/genética , Receptores Toll-Like/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transcrição Gênica , Ativação Transcricional
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...