Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Virology ; 589: 109921, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-37939648

RESUMO

Human norovirus is the leading cause of acute gastroenteritis worldwide, however despite the significance of this pathogen, we have a limited understanding of how noroviruses cause disease, and modulate the innate immune response. Programmed cell death (PCD) is an important part of the innate response to invading pathogens, but little is known about how specific PCD pathways contribute to norovirus replication. Here, we reveal that murine norovirus (MNV) virus-induced PCD in macrophages correlates with the release of infectious virus. We subsequently show, genetically and chemically, that MNV-induced cell death and viral replication occurs independent of the activity of inflammatory mediators. Further analysis revealed that MNV infection promotes the cleavage of apoptotic caspase-3 and PARP. Correspondingly, pan-caspase inhibition, or BAX and BAK deficiency, perturbed viral replication rates and delayed virus release and cell death. These results provide new insights into how MNV harnesses cell death to increase viral burden.


Assuntos
Infecções por Caliciviridae , Norovirus , Camundongos , Humanos , Animais , Macrófagos , Apoptose , Imunidade Inata , Norovirus/fisiologia , Replicação Viral
2.
PLoS Pathog ; 15(4): e1007709, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-31017981

RESUMO

Norovirus infection is the leading cause of food-borne gastroenteritis worldwide, being responsible for over 200,000 deaths annually. Studies with murine norovirus (MNV) showed that protective STAT1 signaling controls viral replication and pathogenesis, but the immune mechanisms that noroviruses exploit to induce pathology are elusive. Here, we show that gastrointestinal MNV infection leads to widespread IL-1ß maturation in MNV-susceptible STAT1-deficient mice. MNV activates the canonical Nlrp3 inflammasome in macrophages, leading to maturation of IL-1ß and to Gasdermin D (GSDMD)-dependent pyroptosis. STAT1-deficient macrophages displayed increased MAVS-mediated expression of pro-IL-1ß, facilitating elevated Nlrp3-dependent release of mature IL-1ß upon MNV infection. Accordingly, MNV-infected Stat1-/- mice showed Nlrp3-dependent maturation of IL-1ß as well as Nlrp3-dependent pyroptosis as assessed by in vivo cleavage of GSDMD to its active N-terminal fragment. While MNV-induced diarrheic responses were not affected, Stat1-/- mice additionally lacking either Nlrp3 or GSDMD displayed lower levels of the fecal inflammatory marker Lipocalin-2 as well as delayed lethality after gastrointestinal MNV infection. Together, these results uncover new insights into the mechanisms of norovirus-induced inflammation and cell death, thereby revealing Nlrp3 inflammasome activation and ensuing GSDMD-driven pyroptosis as contributors to MNV-induced immunopathology in susceptible STAT1-deficient mice.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Infecções por Caliciviridae/patologia , Trato Gastrointestinal/patologia , Inflamassomos/imunologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Piroptose/fisiologia , Fator de Transcrição STAT1/fisiologia , Animais , Proteínas Reguladoras de Apoptose/genética , Infecções por Caliciviridae/imunologia , Infecções por Caliciviridae/metabolismo , Infecções por Caliciviridae/virologia , Células Cultivadas , Trato Gastrointestinal/imunologia , Trato Gastrointestinal/metabolismo , Trato Gastrointestinal/virologia , Inflamassomos/metabolismo , Interleucina-1beta/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/patologia , Macrófagos/virologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Norovirus/imunologia , Norovirus/patogenicidade , Proteínas de Ligação a Fosfato
3.
Antiviral Res ; 148: 32-42, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29097227

RESUMO

The innate immune system provides the host with both a dynamic barrier to prevent infection and a means to which rapid anti-microbial responses can be mounted. The inflammasome pathway is a critical host early response mechanism that enables detection of pathogens and initiates production of inflammatory cytokines, inducing recruitment of effector cells to the site of infection. The complete mechanism of inflammasome activation requires two signals: an initial priming step upon detection of pathogen, followed by activation of intracellular pattern recognition receptors critical to the formation of the inflammasome complex. The inflammasome complex is made of intracellular multiprotein oligomers which includes a sensor protein such as the nucleotide-binding oligomerization domain (NOD) like receptor proteins (NLRP), and an adapter protein, ASC, which critically activates pro-caspase-1. The mature caspase-1 then proteolytically cleaves cytosolic pro-IL-1ß and pro-IL-18, which are then secreted as inflammatory cytokines that activate the inflammatory arm of the immune response to infection. Active caspase-1 also results in pyroptosis, which is a form of cell death triggered by inflammation. The induction and activation of IL-1ß and IL-18 are considered critical signatures for inflammasome activation. With focus upon influenza A virus infection, this review will address present knowledge on the mechanisms of inflammasome complex activation, particularly how the viral components modulate activation of the cytosolic NOD-like receptor protein-3 (NLRP3)-dependent inflammasome complex. We also discuss potential therapeutic strategies that target the inflammasome to ameliorate illness, as well as novel methods of vaccination that target inflammasome stimulation with the aim to increase efficacy.


Assuntos
Inflamassomos/imunologia , Vírus da Influenza A/imunologia , Influenza Humana/imunologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/imunologia , Imunidade Adaptativa , Animais , Interações Hospedeiro-Patógeno , Humanos , Imunidade Inata , Influenza Humana/virologia , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/virologia
4.
J Virol ; 91(3)2017 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-27881660

RESUMO

Norovirus (NoV) infections are a significant health burden to society, yet the lack of reliable tissue culture systems has hampered the development of appropriate antiviral therapies. Here we show that the NoV NS3 protein, derived from murine NoV (MNV), is intimately associated with the MNV replication complex and the viral replication intermediate double-stranded RNA (dsRNA). We observed that when expressed individually, MNV NS3 and NS3 encoded by human Norwalk virus (NV) induced the formation of distinct vesicle-like structures that did not colocalize with any particular protein markers to cellular organelles but localized to cellular membranes, in particular those with a high cholesterol content. Both proteins also showed some degree of colocalization with the cytoskeleton marker ß-tubulin. Although the distribution of MNV and NV NS3s were similar, NV NS3 displayed a higher level of colocalization with the Golgi apparatus and the endoplasmic reticulum (ER). However, we observed that although both proteins colocalized in membranes counterstained with filipin, an indicator of cholesterol content, MNV NS3 displayed a greater association with flotillin and stomatin, proteins known to associate with sphingolipid- and cholesterol-rich microdomains. Utilizing time-lapse epifluorescence microscopy, we observed that the membrane-derived vesicular structures induced by MNV NS3 were highly motile and dynamic in nature, and their movement was dependent on intact microtubules. These results begin to interrogate the functions of NoV proteins during virus replication and highlight the conserved properties of the NoV NS3 proteins among the seven Norovirus genogroups. IMPORTANCE: Many mechanisms involved in the replication of norovirus still remain unclear, including the role for the NS3 protein, one of seven nonstructural viral proteins, which remains to be elucidated. This study reveals that murine norovirus (MNV) NS3 is intimately associated with the viral replication complex and dsRNA. We observed that the NS3 proteins of both MNV and Norwalk virus (NV) induce prominent vesicular structures and that this formation is dependent on microtubules and cellular cholesterol. Thus, this study contributes to our understanding of protein function within different Norovirus genogroups and expands a growing knowledge base on the interaction between positive-strand RNA [(+)RNA] viruses and cellular membranes that contribute to the biogenesis of virus-induced membrane organelles. This study contributes to our understanding of viral protein function and the ability of a viral protein to recruit specific cellular organelles and lipids that enable replication.


Assuntos
Infecções por Caliciviridae/metabolismo , Infecções por Caliciviridae/virologia , Metabolismo dos Lipídeos , Microtúbulos/metabolismo , Norovirus/fisiologia , Proteínas não Estruturais Virais/metabolismo , Replicação Viral , Sequência de Aminoácidos , Animais , Linhagem Celular , Chlorocebus aethiops , Colesterol/metabolismo , Interações Hospedeiro-Patógeno , Espaço Intracelular , Microdomínios da Membrana/metabolismo , Proteínas de Membrana/metabolismo , Camundongos , Ligação Proteica , Transporte Proteico , RNA de Cadeia Dupla/metabolismo , RNA Viral/metabolismo , Células Vero , Proteínas não Estruturais Virais/química
5.
J Interferon Cytokine Res ; 36(4): 215-25, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27046239

RESUMO

Noroviruses (NoVs) belong to the Caliciviridae family of viruses and are responsible for causing the majority of gastroenteritis outbreaks worldwide. In the past decade, research on NoV biology has intensified because of the discovery of murine NoV and subsequently the first cell culture system and small animal model for NoV replication and pathogenesis. In this review, we discuss the current literature on NoV biology, focusing particularly on NoV replication and the interaction between NoV and the host immune response. Understanding the NoV replication cycle and its interaction with cellular processes and innate immune immunity will help develop molecular targets to control human NoV infection and prevent outbreaks. In addition to the innate immune response, we have documented the current efforts to develop NoV vaccines to control outbreaks.


Assuntos
Infecções por Caliciviridae/imunologia , Gastroenterite/imunologia , Norovirus/fisiologia , Animais , Infecções por Caliciviridae/complicações , Infecções por Caliciviridae/prevenção & controle , Modelos Animais de Doenças , Surtos de Doenças , Gastroenterite/etiologia , Gastroenterite/prevenção & controle , Interações Hospedeiro-Patógeno , Humanos , Imunidade Inata , Camundongos , Vacinas Virais , Replicação Viral
6.
Nat Commun ; 6: 6795, 2015 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-25865065

RESUMO

To date, the activities of protein kinases have formed the core of our understanding of cell signal transduction. Comprehension of the extent of protein acetylation has raised expectations that this alternate post-transcriptional modification will be shown to rival phosphorylation in its importance in mediating cellular responses. However, limited instances have been identified. Here we show that signalling from Toll-like or TNF-α receptors triggers the calcium/calmodulin-dependent protein kinase (CaMK2) to activate histone acetyltransferase-1 (HAT1), which then acetylates the transcriptional regulator PLZF. Acetylation of PLZF promotes the assembly of a repressor complex incorporating HDAC3 and the NF-κB p50 subunit that limits the NF-κB response. Accordingly, diminishing the activity of CaMK2, the expression levels of PLZF or HAT1, or mutating key residues that are covalently modified in PLZF and HAT1, curtails control of the production of inflammatory cytokines. These results identify a central role for acetylation in controlling the inflammatory NF-κB transcriptional programme.


Assuntos
Histona Acetiltransferases/genética , Fatores de Transcrição Kruppel-Like/genética , NF-kappa B/genética , Processamento de Proteína Pós-Traducional , Transcrição Gênica , Acetilação , Animais , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/genética , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/imunologia , Histona Acetiltransferases/imunologia , Histona Desacetilases/genética , Histona Desacetilases/imunologia , Imunidade Inata , Inflamação/induzido quimicamente , Inflamação/genética , Inflamação/imunologia , Inflamação/patologia , Fatores de Transcrição Kruppel-Like/deficiência , Fatores de Transcrição Kruppel-Like/imunologia , Lipopolissacarídeos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação , NF-kappa B/imunologia , Proteína com Dedos de Zinco da Leucemia Promielocítica , Receptores do Fator de Necrose Tumoral/genética , Receptores do Fator de Necrose Tumoral/imunologia , Transdução de Sinais
7.
Nucleic Acids Res ; 43(2): 1177-88, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25539920

RESUMO

Anti-microRNA (miRNA) oligonucleotides (AMOs) with 2'-O-Methyl (2'OMe) residues are commonly used to study miRNA function and can achieve high potency, with low cytotoxicity. Not withstanding this, we demonstrate the sequence-dependent capacity of 2'OMe AMOs to inhibit Toll-like receptor (TLR) 7 and 8 sensing of immunostimulatory RNA, independent of their miRNA-targeting function. Through a screen of 29 AMOs targeting common miRNAs, we found a subset of sequences highly inhibitory to TLR7 sensing in mouse macrophages. Interspecies conservation of this inhibitory activity was confirmed on TLR7/8 activity in human peripheral blood mononuclear cells. Significantly, we identified a core motif governing the inhibitory activity of these AMOs, which is present in more than 50 AMOs targeted to human miRNAs in miRBaseV20. DNA/locked nucleic acids (LNA) AMOs synthesized with a phosphorothioate backbone also inhibited TLR7 sensing in a sequence-dependent manner, demonstrating that the off-target effects of AMOs are not restricted to 2'OMe modification. Taken together, our work establishes the potential for off-target effects of AMOs on TLR7/8 function, which should be taken into account in their therapeutic development and in vivo application.


Assuntos
MicroRNAs/antagonistas & inibidores , Oligonucleotídeos/química , Receptor 7 Toll-Like/antagonistas & inibidores , Receptor 8 Toll-Like/antagonistas & inibidores , Adjuvantes Imunológicos/farmacologia , Animais , Sequência de Bases , Células HEK293 , Humanos , Camundongos , Motivos de Nucleotídeos , RNA/farmacologia
8.
J Biol Chem ; 289(40): 27776-93, 2014 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-25100717

RESUMO

Integrin-linked kinase (ILK) is a ubiquitously expressed and highly conserved serine-threonine protein kinase that regulates cellular responses to a wide variety of extracellular stimuli. ILK is involved in cell-matrix interactions, cytoskeletal organization, and cell signaling. ILK signaling has also been implicated in oncogenesis and progression of cancers. However, its role in the innate immune system remains unknown. Here, we show that ILK mediates pro-inflammatory signaling in response to lipopolysaccharide (LPS). Pharmacological or genetic inhibition of ILK in mouse embryonic fibroblasts and macrophages selectively blocks LPS-induced production of the pro-inflammatory cytokine tumor necrosis factor α (TNF-α). ILK is required for LPS-induced activation of nuclear factor κB (NF-κB) and transcriptional induction of TNF-α. The modulation of LPS-induced TNF-α synthesis by ILK does not involve the classical NF-κB pathway, because IκB-α degradation and p65 nuclear translocation are both unaffected by ILK inhibition. Instead, ILK is involved in an alternative activation of NF-κB signaling by modulating the phosphorylation of p65 at Ser-536. Furthermore, ILK-mediated alternative NF-κB activation through p65 Ser-536 phosphorylation also occurs during Helicobacter pylori infection in macrophages and gastric cancer cells. Moreover, ILK is required for H. pylori-induced TNF-α secretion in macrophages. Although ILK-mediated phosphorylation of p65 at Ser-536 is independent of the phosphatidylinositol 3-kinase (PI3K)/Akt pathway during LPS stimulation, upon H. pylori infection this event is dependent on the PI3K/Akt pathway. Our findings implicate ILK as a critical regulatory molecule for the NF-κB-mediated pro-inflammatory signaling pathway, which is essential for innate immune responses against pathogenic microorganisms.


Assuntos
Infecções por Helicobacter/enzimologia , Helicobacter pylori/fisiologia , Lipopolissacarídeos/metabolismo , NF-kappa B/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , eIF-2 Quinase/química , eIF-2 Quinase/metabolismo , Motivos de Aminoácidos , Animais , Regulação da Expressão Gênica , Infecções por Helicobacter/genética , Infecções por Helicobacter/metabolismo , Infecções por Helicobacter/microbiologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/genética , Fosforilação , Proteínas Serina-Treonina Quinases/genética , Serina/metabolismo , Fator de Necrose Tumoral alfa/genética , eIF-2 Quinase/genética
9.
J Virol ; 88(2): 799-810, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24227841

RESUMO

RNA-specific adenosine deaminase (ADAR)-mediated adenosine-to-inosine (A-to-I) editing is a critical arm of the antiviral response. However, mechanistic insights into how A-to-I RNA editing affects viral infection are lacking. We posited that inosine incorporation into RNA facilitates sensing of nonself RNA by innate immune sensors and accordingly investigated the impact of inosine-modified RNA on Toll-like receptor 7 and 8 (TLR7/8) sensing. Inosine incorporation into synthetic single-stranded RNA (ssRNA) potentiated tumor necrosis factor alpha (TNF-α) or alpha interferon (IFN-α) production in human peripheral blood mononuclear cells (PBMCs) in a sequence-dependent manner, indicative of TLR7/8 recruitment. The effect of inosine incorporation on TLR7/8 sensing was restricted to immunostimulatory ssRNAs and was not seen with inosine-containing short double-stranded RNAs or with a deoxy-inosine-modified ssRNA. Inosine-mediated increase of self-secondary structure of an ssRNA resulted in potentiated IFN-α production in human PBMCs through TLR7 recruitment, as established through the use of a TLR7 antagonist and Tlr7-deficient cells. There was a correlation between hyperediting of influenza A viral ssRNA and its ability to stimulate TNF-α, independent of 5'-triphosphate residues, and involving Adar-1. Furthermore, A-to-I editing of viral ssRNA directly enhanced mouse Tlr7 sensing, when present in proportions reproducing biologically relevant levels of RNA editing. Thus, we demonstrate for the first time that inosine incorporation into immunostimulatory ssRNA can potentiate TLR7/8 activation. Our results suggest a novel function of A-to-I RNA editing, which is to facilitate TLR7/8 sensing of phagocytosed viral RNA.


Assuntos
Vírus da Influenza A Subtipo H1N1/genética , Influenza Humana/imunologia , Influenza Humana/virologia , Inosina/genética , Edição de RNA , RNA Viral/genética , Receptor 7 Toll-Like/imunologia , Receptor 8 Toll-Like/imunologia , Adenosina/genética , Adenosina/imunologia , Adenosina Desaminase/genética , Adenosina Desaminase/imunologia , Animais , Sequência de Bases , Feminino , Humanos , Vírus da Influenza A Subtipo H1N1/imunologia , Influenza Humana/genética , Inosina/imunologia , Interferon-alfa/genética , Interferon-alfa/imunologia , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/virologia , Masculino , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Conformação de Ácido Nucleico , RNA Viral/química , RNA Viral/imunologia , Receptor 7 Toll-Like/genética , Receptor 8 Toll-Like/genética
10.
J Interferon Cytokine Res ; 32(8): 350-61, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22817608

RESUMO

Recent advances in our understanding of foreign nucleic acid sensing indicate an important role for the human Toll-like receptor (TLR) 8 in the initiation of immune responses to certain pathogens. However, TLR8, far too often grouped together with TLR7 for its common ability to detect RNA, has a function on its own in the initiation of specific proinflammatory responses to viruses and bacteria. Here, we present an overview of what is currently known of human TLR8 biology, from genetic regulation to its function in innate immunity, and discuss how TLR8 could present novel therapeutic opportunities in viral and cancer diseases.


Assuntos
Imunidade Inata/fisiologia , RNA/imunologia , Receptor 8 Toll-Like/fisiologia , Animais , Doenças Autoimunes/genética , Doenças Autoimunes/imunologia , Doenças Autoimunes/metabolismo , Regulação da Expressão Gênica , Humanos , Ligantes , RNA Bacteriano/imunologia , RNA Viral/imunologia , Receptor 8 Toll-Like/genética , Receptor 8 Toll-Like/metabolismo
11.
Nucleic Acids Res ; 40(16): 8048-58, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22684508

RESUMO

Fine-tuning of inflammatory responses by microRNAs (miRNAs) is complex, as they can both enhance and repress expression of pro-inflammatory mediators. In this study, we investigate inflammatory responses following global miRNA depletion, to better define the overall contribution of miRNAs to inflammation. We demonstrate that miRNAs positively regulate Toll-like receptor signaling using inducible Dicer1 deletion and global miRNA depletion. We establish an important contribution of miR-19b in this effect, which potentiates nuclear factor-κB (NF-κB) activity in human and mouse cells. Positive regulation of NF-κB signaling by miR-19b involves the coordinated suppression of a regulon of negative regulators of NF-κB signaling (including A20/Tnfaip3, Rnf11, Fbxl11/Kdm2a and Zbtb16). Transfection of miR-19b mimics exacerbated the inflammatory activation of rheumatoid arthritis primary fibroblast-like synoviocytes, demonstrating its physiological importance in the pathology of this disease. This study constitutes, to our knowledge, the first description of a miR-19 regulon that controls NF-κB signaling, and suggests that targeting this miRNA and linked family members could regulate the activity of NF-κB signaling in inflammation.


Assuntos
MicroRNAs/metabolismo , NF-kappa B/metabolismo , Regulon , Transdução de Sinais , Animais , Células da Medula Óssea/metabolismo , Células Cultivadas , Fibroblastos/metabolismo , Humanos , Inflamação/genética , Mediadores da Inflamação/metabolismo , Macrófagos/metabolismo , Camundongos , Membrana Sinovial/citologia , Membrana Sinovial/metabolismo , Receptores Toll-Like/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA