Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
1.
Front Immunol ; 13: 918551, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36248901

RESUMO

The complement system is an ancient and critical part of innate immunity. Recent studies have highlighted novel roles of complement beyond lysis of invading pathogens with implications in regulating the innate immune response, as well as contributing to metabolic reprogramming of T-cells, synoviocytes as well as cells in the CNS. These findings hint that complement can be an immunometabolic regulator, but whether this is also the case for the terminal step of the complement pathway, the membrane attack complex (MAC) is not clear. In this study we focused on determining whether MAC is an immunometabolic regulator of the innate immune response in human monocyte-derived macrophages. Here, we uncover previously uncharacterized metabolic changes and mitochondrial dysfunction occurring downstream of MAC deposition. These alterations in glycolytic flux and mitochondrial morphology and function mediate NLRP3 inflammasome activation, pro-inflammatory cytokine release and gasdermin D formation. Together, these data elucidate a novel signalling cascade, with metabolic alterations at its center, in MAC-stimulated human macrophages that drives an inflammatory consequence in an immunologically relevant cell type.


Assuntos
Inflamassomos , Proteína 3 que Contém Domínio de Pirina da Família NLR , Complexo de Ataque à Membrana do Sistema Complemento/metabolismo , Humanos , Inflamassomos/metabolismo , Interleucina-18/metabolismo , Interleucina-1beta/metabolismo , Macrófagos , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo
2.
Nat Commun ; 13(1): 1406, 2022 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-35301296

RESUMO

Human rhinovirus (HRV), like coronavirus (HCoV), are positive-strand RNA viruses that cause both upper and lower respiratory tract illness, with their replication facilitated by concentrating RNA-synthesizing machinery in intracellular compartments made of modified host membranes, referred to as replication organelles (ROs). Here we report a non-canonical, essential function for stimulator of interferon genes (STING) during HRV infections. While the canonical function of STING is to detect cytosolic DNA and activate inflammatory responses, HRV infection triggers the release of STIM1-bound STING in the ER by lowering Ca2+, thereby allowing STING to interact with phosphatidylinositol 4-phosphate (PI4P) and traffic to ROs to facilitates viral replication and transmission via autophagy. Our results thus hint a critical function of STING in HRV viral replication and transmission, with possible implications for other RO-mediated RNA viruses.


Assuntos
Enterovirus , Vírus de RNA , Humanos , Organelas , Rhinovirus , Replicação Viral/fisiologia
3.
PLoS Pathog ; 17(4): e1009417, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33861800

RESUMO

Macrophages are important drivers of pathogenesis and progression to AIDS in HIV infection. The virus in the later phases of the infection is often predominantly macrophage-tropic and this tropism contributes to a chronic inflammatory and immune activation state that is observed in HIV patients. Pattern recognition receptors of the innate immune system are the key molecules that recognise HIV and mount the inflammatory responses in macrophages. The innate immune response against HIV-1 is potent and elicits caspase-1-dependent pro-inflammatory cytokine production of IL-1ß and IL-18. Although, NLRP3 has been reported as an inflammasome sensor dictating this response little is known about the pattern recognition receptors that trigger the "priming" signal for inflammasome activation, the NLRs involved or the HIV components that trigger the response. Using a combination of siRNA knockdowns in monocyte derived macrophages (MDMs) of different TLRs and NLRs as well as chemical inhibition, it was demonstrated that HIV Vpu could trigger inflammasome activation via TLR4/NLRP3 leading to IL-1ß/IL-18 secretion. The priming signal is triggered via TLR4, whereas the activation signal is triggered by direct effects on Kv1.3 channels, causing K+ efflux. In contrast, HIV gp41 could trigger IL-18 production via NAIP/NLRC4, independently of priming, as a one-step inflammasome activation. NAIP binds directly to the cytoplasmic tail of HIV envelope protein gp41 and represents the first non-bacterial ligand for the NAIP/NLRC4 inflammasome. These divergent pathways represent novel targets to resolve specific inflammatory pathologies associated with HIV-1 infection in macrophages.


Assuntos
Infecções por HIV/virologia , Inflamassomos/imunologia , Interleucina-18/metabolismo , Interleucina-1beta/metabolismo , Macrófagos/virologia , Fragmentos de Peptídeos/metabolismo , Comunicação Celular/genética , Comunicação Celular/imunologia , Expressão Gênica/genética , Expressão Gênica/imunologia , Infecções por HIV/metabolismo , Humanos , Imunidade Inata/genética , Imunidade Inata/imunologia , Inflamassomos/metabolismo , Macrófagos/imunologia , Proteína Inibidora de Apoptose Neuronal/genética , Transdução de Sinais/imunologia
4.
Immunology ; 163(4): 348-362, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33682108

RESUMO

Nucleotide-binding domain and leucine-rich repeat receptor (NLR)-mediated inflammasome activation is important in host response to microbes, danger-associated molecular patterns (DAMPs) and metabolic disease. Some NLRs have been shown to interact with distinct cell metabolic pathways and cause negative regulation, tumorigenesis and autoimmune disorders, interacting with multiple innate immune receptors to modulate disease. NLR activation is therefore crucial in host response and in the regulation of metabolic pathways that can trigger a wide range of immunometabolic diseases or syndromes. However, the exact mode by which some of the less well-studied NLR inflammasomes are activated, interact with other metabolites and immune receptors, and the role they play in the progression of metabolic diseases is still not fully elucidated. In this study, we review up-to-date evidence regarding NLR function in metabolic pathways and the interplay with other immune receptors involved in GPCR signalling, gut microbiota and the complement system, in order to gain a better understanding of its link to disease processes.


Assuntos
Doenças Autoimunes/metabolismo , Proteínas do Sistema Complemento/metabolismo , Inflamassomos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animais , Doenças Autoimunes/imunologia , Microbioma Gastrointestinal , Humanos , Imunidade Inata , Receptor Cross-Talk , Transdução de Sinais
5.
J Biol Chem ; 293(15): 5509-5521, 2018 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-29463677

RESUMO

Different immune activation states require distinct metabolic features and activities in immune cells. For instance, inhibition of fatty acid synthase (FASN), which catalyzes the synthesis of long-chain fatty acids, prevents the proinflammatory response in macrophages; however, the precise role of this enzyme in this response remains poorly defined. Consistent with previous studies, we found here that FASN is essential for lipopolysaccharide-induced, Toll-like receptor (TLR)-mediated macrophage activation. Interestingly, only agents that block FASN upstream of acetoacetyl-CoA synthesis, including the well-characterized FASN inhibitor C75, inhibited TLR4 signaling, while those acting downstream had no effect. We found that acetoacetyl-CoA could overcome C75's inhibitory effect, whereas other FASN metabolites, including palmitate, did not prevent C75-mediated inhibition. This suggested an unexpected role for acetoacetyl-CoA in inflammation that is independent of its role in palmitate synthesis. Our evidence further suggested that acetoacetyl-CoA arising from FASN activity promotes cholesterol production, indicating a surprising link between fatty acid synthesis and cholesterol synthesis. We further demonstrate that this process is required for TLR4 to enter lipid rafts and facilitate TLR4 signaling. In conclusion, we have uncovered an unexpected link between FASN and cholesterol synthesis that appears to be required for TLR signal transduction and proinflammatory macrophage activation.


Assuntos
Colesterol/biossíntese , Ácido Graxo Sintase Tipo I/metabolismo , Ativação de Macrófagos , Macrófagos/enzimologia , Transdução de Sinais , Acil Coenzima A/metabolismo , Animais , Inflamação/enzimologia , Camundongos , Ácido Palmítico/metabolismo , Receptor 4 Toll-Like/metabolismo
6.
Trends Mol Med ; 23(2): 165-180, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-28109721

RESUMO

The inflammasome is a cytoplasmic protein complex that processes interleukins (IL)-1ß and IL-18, and drives a form of cell death known as pyroptosis. Oligomerization of this complex is actually the second step of activation, and a priming step must occur first. This involves transcriptional upregulation of pro-IL-1ß, inflammasome sensor NLRP3, or the non-canonical inflammasome sensor caspase-11. An additional aspect of priming is the post-translational modification of particular inflammasome constituents. Priming is typically accomplished in vitro using a microbial Toll-like receptor (TLR) ligand. However, it is now clear that inflammasomes are activated during the progression of sterile inflammatory diseases such as atherosclerosis, metabolic disease, and neuroinflammatory disorders. Therefore, it is time to consider the endogenous factors and mechanisms that may prime the inflammasome in these conditions.


Assuntos
Inflamassomos/imunologia , Inflamação/imunologia , Doença de Alzheimer/imunologia , Animais , Aterosclerose/imunologia , Doenças Hereditárias Autoinflamatórias/imunologia , Humanos , Imunidade Inata , Interleucina-1beta/imunologia , Doenças Metabólicas/imunologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/imunologia , Neuroimunomodulação , Obesidade/imunologia , Receptores Toll-Like/imunologia
7.
Immunology ; 147(2): 152-64, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26572245

RESUMO

The innate immune system is an ancient surveillance system able to sense microbial invaders as well as aberrations in normal cell function. No longer viewed as a static and non-specific part of immunity, the innate immune system employs a plethora of specialized pattern recognition sensors to monitor and achieve homeostasis; these include the Toll-like receptors, the retinoic acid-inducible gene-like receptors, the nucleotide-binding oligomerization domain receptors (NLRs), the C-type lectins and the complement system. In order to increase specificity and diversity, innate immunity uses homotypic and heterotypic associations among these different components. Multi-molecular assemblies are formed both on the cell surface and in the cytosol to respond to pathogen and danger signals. Diverse, but tailored, responses to a changing environment are orchestrated depending on the the nature of the challenge and the repertoire of interacting receptors and components available in the sensing cell. It is now emerging that innate immunity operates a system of 'checks and balances' where interaction among the sensors is key in maintaining normal cell function. Complement sits at the heart of this alarm system and it is becoming apparent that it is capable of interacting with all the other pathways to effect a tailored immune response. In this review, we will focus on complement interactions with NLRs, the so-called 'inflammasomes', describing the molecular mechanisms that have been revealed so far and discussing the circumstantial evidence that exists for these interactions in disease states.


Assuntos
Ativação do Complemento , Proteínas do Sistema Complemento/imunologia , Imunidade Inata , Inflamassomos/imunologia , Inflamação/imunologia , Animais , Anti-Inflamatórios/uso terapêutico , Ativação do Complemento/efeitos dos fármacos , Proteínas do Sistema Complemento/metabolismo , Humanos , Imunidade Inata/efeitos dos fármacos , Inflamassomos/efeitos dos fármacos , Inflamassomos/metabolismo , Inflamação/metabolismo , Inflamação/prevenção & controle , Mediadores da Inflamação/imunologia , Mediadores da Inflamação/metabolismo , Ligantes , Receptores Imunológicos/imunologia , Receptores Imunológicos/metabolismo , Transdução de Sinais
8.
Trends Microbiol ; 22(10): 580-8, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24986075

RESUMO

Innate immunity has a primary role in lung antimicrobial defenses. The inflammasome has evolved for this purpose and is an important surveillance system that, when triggered, fights infection and eliminates pathogens. However, there is growing evidence that the inflammasome also plays a role in the pathogenesis of acute and chronic respiratory disease. Inflammasomes contribute to both the clearance of the pathogen as well as its pathogenesis - depending on the amount of inflammation triggered. How respiratory viruses trigger inflammasome activation remains unclear. Emerging evidence shows that ion flux is responsible for triggering inflammasome activation in the lung, causing lung pathology and disease exacerbations. Viroporins, encoded by all common respiratory viruses, are responsible for the changes in intracellular ion homeostasis that modulate inflammasome activation. This is a novel mechanism by which respiratory viral infection activates inflammasomes, and identifies sensing of disturbances in intracellular ionic concentrations as a novel pathogen-recognition pathway in the lung.


Assuntos
Imunidade Inata/imunologia , Inflamassomos/imunologia , Ferro/metabolismo , Pulmão/imunologia , Humanos , Inflamação/imunologia , Ativação Viral
9.
Cell Host Microbe ; 15(6): 768-78, 2014 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-24922578

RESUMO

Certain low-abundance bacterial species, such as the periodontitis-associated oral bacterium Porphyromonas gingivalis, can subvert host immunity to remodel a normally symbiotic microbiota into a dysbiotic, disease-provoking state. However, such pathogens also exploit inflammation to thrive in dysbiotic conditions. How these bacteria evade immunity while maintaining inflammation is unclear. As previously reported, P. gingivalis remodels the oral microbiota into a dysbiotic state by exploiting complement. Now we show that in neutrophils P. gingivalis disarms a host-protective TLR2-MyD88 pathway via proteasomal degradation of MyD88, whereas it activates an alternate TLR2-Mal-PI3K pathway. This alternate TLR2-Mal-PI3K pathway blocks phagocytosis, provides "bystander" protection to otherwise susceptible bacteria, and promotes dysbiotic inflammation in vivo. This mechanism to disengage bacterial clearance from inflammation required an intimate crosstalk between TLR2 and the complement receptor C5aR and can contribute to the persistence of microbial communities that drive dysbiotic diseases.


Assuntos
Disbiose/microbiologia , Periodontite/microbiologia , Porphyromonas gingivalis/patogenicidade , Receptor da Anafilatoxina C5a/metabolismo , Receptor 2 Toll-Like/metabolismo , Animais , Infecções por Bacteroidaceae/imunologia , Proteínas do Sistema Complemento/imunologia , Disbiose/imunologia , Interações Hospedeiro-Patógeno/imunologia , Camundongos , Camundongos Mutantes , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/metabolismo , Neutrófilos/imunologia , Neutrófilos/microbiologia , Periodontite/imunologia , Fagocitose , Fosfatidilinositol 3-Quinases/metabolismo , Porphyromonas gingivalis/imunologia , Receptor da Anafilatoxina C5a/genética , Receptor da Anafilatoxina C5a/imunologia , Transdução de Sinais , Receptor 2 Toll-Like/genética , Receptor 2 Toll-Like/imunologia
10.
J Biol Chem ; 289(22): 15309-18, 2014 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-24737331

RESUMO

Inflammation is mediated mainly by leukocytes that express both Toll-like receptor 4 (TLR4) and Fc γ receptors (FcγR). Dysregulated activation of leukocytes via exogenous and endogenous ligands of TLR4 results in a large number of inflammatory disorders that underlie a variety of human diseases. Thus, differentially blocking inflammatory cells while sparing structural cells, which are FcγR-negative, represents an elegant strategy when targeting the underlying causes of human diseases. Here, we report a novel tethering mechanism of the Fv and Fc portions of anti-TLR4 blocking antibodies that achieves increased potency on inflammatory cells. In the presence of ligand (e.g. lipopolysaccharide (LPS)), TLR4 traffics into glycolipoprotein microdomains, forming concentrated protein platforms that include FcγRs. This clustering produces a microenvironment allowing anti-TLR4 antibodies to co-engage TLR4 and FcγRs, increasing their avidity and thus substantially increasing their inhibitory potency. Tethering of antibodies to both TLR4 and FcγRs proves valuable in ameliorating inflammation in vivo. This novel mechanism of action therefore has the potential to enable selective intervention of relevant cell types in TLR4-driven diseases.


Assuntos
Inflamação/imunologia , Macrófagos/imunologia , Receptores de IgG/imunologia , Receptor 4 Toll-Like/imunologia , Animais , Anticorpos Monoclonais/imunologia , Sítios de Ligação , Células CHO , Linhagem Celular , Cricetulus , Dimerização , Feminino , Humanos , Inflamação/metabolismo , Macrófagos/citologia , Microdomínios da Membrana/imunologia , Microdomínios da Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Receptores de IgG/metabolismo , Receptor 4 Toll-Like/química , Receptor 4 Toll-Like/metabolismo , Células U937
11.
Am J Obstet Gynecol ; 210(2): 122.e1-122.e10, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24080302

RESUMO

OBJECTIVE: The pathway by which herpes simplex virus 2 (HSV2) triggers the innate immune system in the urogenital system has not as yet been fully elucidated. In this study, we aimed to determine which pattern recognition receptors (PRRs) recognize HSV2 in primary vaginal epithelial cells. Once we deciphered the receptors involved, we aimed to target them to immunomodulate innate responses as a prophylactic or therapeutic intervention for early HSV2 infection. STUDY DESIGN: To determine which PRRs are involved, receptor silencing as well as confocal microscopy was utilized. For immunomodulation, PRR agonists were utilized to induce a strong, local response to limit the infection, and we used 2 quantitative methods, flow cytometry and plaque assays, to determine their effect on HSV2 replication. RESULTS: Our results show that HSV2 is detected by a plethora of PRRs: Toll-like receptors (TLR) 2 as well as deoxyribonucleic acid (DNA) sensors TLR9, DNA-dependent activator of interferon regulatory factors, and to a lesser extent interferon-inducible 16, which trigger cytokine secretion to protect the host. Using PRR agonists, such as lipoproteins, CpG DNA, and cyclic dinucleotides, we could significantly limit HSV2 replication. CONCLUSION: Different PRRs are strategically placed in different cell locations to detect virus invasion. Use of agonists that target and activate these PRRs appeared to be effective in preventing primary HSV2 infection in vaginal cells and could provide new insights in defense against HSV2 urogenital infections.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Herpes Genital/virologia , Herpesvirus Humano 2/fisiologia , Proteínas Nucleares/fisiologia , Fosfoproteínas/fisiologia , Receptor 2 Toll-Like/fisiologia , Ativação Viral/fisiologia , Feminino , Herpes Genital/imunologia , Humanos , Imunidade Inata , Proteínas de Ligação a RNA , Receptor Toll-Like 9/fisiologia , Vagina/imunologia , Vagina/virologia
12.
Am J Respir Cell Mol Biol ; 49(6): 923-34, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23815151

RESUMO

Human rhinoviruses have been linked with underlying lung disorders, such as asthma and chronic obstructive pulmonary disease, in children and adults. However, the mechanism of virus-induced airway inflammation is poorly understood. In this study, using virus deletion mutants and silencing for nucleotide-binding oligomerization domain-like receptors (NLRs), we show that the rhinovirus ion channel protein 2B triggers NLRP3 and NLRC5 inflammasome activation and IL-1ß secretion in bronchial cells. 2B protein targets the endoplasmic reticulum and Golgi and induces Ca(2+) reduction in these organelles, thereby disturbing the intracellular calcium homeostasis. NLRP3 and NLRC5 act in a cooperative manner during the inflammasome assembly by sensing intracellular Ca(2+) fluxes and trigger IL-1ß secretion. These results reveal for the first time that human rhinovirus infection in primary bronchial cells triggers inflammasome activation.


Assuntos
Brônquios/metabolismo , Brônquios/virologia , Sinalização do Cálcio/fisiologia , Proteínas de Transporte/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Rhinovirus/patogenicidade , Asma/etiologia , Asma/metabolismo , Asma/virologia , Proteínas de Transporte/antagonistas & inibidores , Proteínas de Transporte/genética , Células Cultivadas , Retículo Endoplasmático/metabolismo , Retículo Endoplasmático/virologia , Técnicas de Silenciamento de Genes , Complexo de Golgi/metabolismo , Complexo de Golgi/virologia , Humanos , Inflamassomos/metabolismo , Interleucina-1beta/biossíntese , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR , Infecções por Picornaviridae/complicações , Infecções por Picornaviridae/metabolismo , Infecções por Picornaviridae/virologia , Doença Pulmonar Obstrutiva Crônica/etiologia , Doença Pulmonar Obstrutiva Crônica/metabolismo , Doença Pulmonar Obstrutiva Crônica/virologia , Rhinovirus/genética , Rhinovirus/metabolismo , Proteínas Virais/metabolismo
13.
J Cell Sci ; 126(Pt 13): 2903-13, 2013 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-23613465

RESUMO

The membrane attack complex of complement (MAC), apart from its classical role of lysing cells, can also trigger a range of non-lethal effects on cells, acting as a drive to inflammation. In the present study, we chose to investigate these non-lethal effects on inflammasome activation. We found that, following sublytic MAC attack, there is increased cytosolic Ca(2+) concentration, at least partly through Ca(2+) release from the endoplasmic reticulum lumen via the inositol 1,4,5-triphosphate receptor (IP3R) and ryanodine receptor (RyR) channels. This increase in intracellular Ca(2+) concentration leads to Ca(2+) accumulation in the mitochondrial matrix via the 'mitochondrial calcium uniporter' (MCU), and loss of mitochondrial transmembrane potential, triggering NLRP3 inflammasome activation and IL-1ß release. NLRP3 co-localises with the mitochondria, probably sensing the increase in calcium and the resultant mitochondrial dysfunction, leading to caspase activation and apoptosis. This is the first study that links non-lethal effects of sublytic MAC attack with inflammasome activation and provides a mechanism by which sublytic MAC can drive inflammation and apoptosis.


Assuntos
Cálcio/metabolismo , Proteínas de Transporte/genética , Membrana Celular/efeitos dos fármacos , Complexo de Ataque à Membrana do Sistema Complemento/farmacologia , Células Epiteliais/efeitos dos fármacos , Inflamassomos/efeitos dos fármacos , Mucosa Respiratória/efeitos dos fármacos , Cálcio/agonistas , Canais de Cálcio/genética , Canais de Cálcio/metabolismo , Proteínas de Transporte/imunologia , Membrana Celular/imunologia , Retículo Endoplasmático/metabolismo , Células Epiteliais/citologia , Células Epiteliais/imunologia , Humanos , Inflamassomos/metabolismo , Receptores de Inositol 1,4,5-Trifosfato/genética , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Interleucina-1beta/imunologia , Interleucina-1beta/metabolismo , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Potencial da Membrana Mitocondrial/imunologia , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR , Cultura Primária de Células , Mucosa Respiratória/citologia , Mucosa Respiratória/imunologia , Canal de Liberação de Cálcio do Receptor de Rianodina/genética , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo
14.
PLoS One ; 8(4): e61199, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23593431

RESUMO

Ureaplasma species are the most frequently isolated microorganisms inside the amniotic cavity and have been associated with spontaneous abortion, chorioamnionitis, premature rupture of the membranes (PROM), preterm labour (PL) pneumonia in neonates and bronchopulmonary dysplasia in neonates. The mechanisms by which Ureaplasmas cause such diseases remain unclear, but it is believed that inappropriate induction of inflammatory responses is involved, triggered by the innate immune system. As part of its mechanism of activation, the innate immune system employs germ-lined encoded receptors, called pattern recognition receptors (PRRs) in order to "sense" pathogens. One such family of PRRs are the Toll like receptor family (TLR). In the current study we aimed to elucidate the role of TLRs in Ureaplasma-induced inflammation in human amniotic epithelial cells. Using silencing, as well as human embryonic kidney (HEK) transfected cell lines, we demonstrate that TLR2, TLR6 and TLR9 are involved in the inflammatory responses against Ureaplasma parvum and urealyticum serovars. Ureaplasma lipoproteins, such as Multiple Banded antigen (MBA), trigger responses via TLR2/TLR6, whereas the whole bacterium is required for TLR9 activation. No major differences were observed between the different serovars. Cell activation by Ureaplasma parvum and urealyticum seem to require lipid raft function and formation of heterotypic receptor complexes comprising of TLR2 and TLR6 on the cell surface and TLR9 intracellularly.


Assuntos
Âmnio/patologia , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Receptores Toll-Like/metabolismo , Infecções por Ureaplasma/metabolismo , Ureaplasma urealyticum/fisiologia , Ureaplasma/fisiologia , Membrana Celular/metabolismo , Citocinas/metabolismo , Endocitose , Endossomos/metabolismo , Endossomos/microbiologia , Transferência Ressonante de Energia de Fluorescência , Gangliosídeo G(M1)/metabolismo , Inativação Gênica , Células HEK293 , Humanos , Espaço Intracelular/microbiologia , Microdomínios da Membrana/metabolismo , Fator 88 de Diferenciação Mieloide/metabolismo , Receptor 2 Toll-Like/metabolismo , Receptor 6 Toll-Like/metabolismo , Receptor Toll-Like 9/metabolismo , Infecções por Ureaplasma/microbiologia
15.
Thorax ; 68(1): 66-75, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23229815

RESUMO

BACKGROUND: Respiratory syncytial virus (RSV) remains the leading cause of serious viral bronchiolitis and pneumonia in infants and young children throughout the world. The burden of disease is significant, with 70% of all infants being infected with RSV within the first year of their life. 40% of those children discharged from hospital have recurrent, repeated respiratory symptoms and wheezing for at least 10 years. The infection is also an important illness in the elderly and immunocompromised individuals. Ongoing symptoms relate to continued lung inflammation. One cytokine that is associated with RSV infection is IL-1ß, but the mechanism of activation remain unclear. OBJECTIVES: In the current study, we set out to decipher the molecular mechanisms of RSV-induced inflammasome activation. METHODS AND RESULTS: Using deletion mutants of the virus and measuring IL-1ß secretion, as well as caspase 1 expression via western blotting, we demonstrate that the NLRP3 inflammasome is activated through the small hydrophobic (SH) RSV viroporin which induces membrane permeability to ions or small molecules. Confocal microscopy revealed that during virus infection, SH seems to accumulate within lipid rafts in the Golgi compartments. CONCLUSIONS: Upon RSV infection, SH gets localised in the cell membranes and intracellular organelle membranes, and then induces permeability by disrupting membrane architecture, thus leading us to believe that formation of viral ion channels in lipid bilayers of cells is a viral recognition pathway used by the host to signal inflammasome activation.


Assuntos
Proteínas de Transporte/metabolismo , Inflamassomos/metabolismo , Vírus Sincicial Respiratório Humano/genética , Adulto , Bronquiolite Viral/genética , Bronquiolite Viral/fisiopatologia , Proteínas de Transporte/genética , Células Cultivadas , Células Epiteliais/metabolismo , Células Epiteliais/fisiologia , Humanos , Inflamassomos/genética , Pulmão/citologia , Masculino , Microscopia Confocal , Proteína 3 que Contém Domínio de Pirina da Família NLR , Pneumonia Viral/metabolismo , Pneumonia Viral/fisiopatologia , RNA Viral/genética , RNA Viral/metabolismo , Infecções por Vírus Respiratório Sincicial/genética , Infecções por Vírus Respiratório Sincicial/fisiopatologia , Vírus Sincicial Respiratório Humano/fisiologia , Sensibilidade e Especificidade , Transdução de Sinais/genética
17.
J Cell Sci ; 125(Pt 20): 4761-9, 2012 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-22797917

RESUMO

The innate immune system is a vital part of the body's defences against viral pathogens. The proteins retinoic acid-inducible gene-I (RIG-I) and melanoma differentiation associated gene 5 (MDA5) function as cytoplasmic pattern recognition receptors that are involved in the elimination of actively replicating RNA viruses. Their location and their differential responses to RNA viruses emphasises the complexity of the innate detection system. Despite the wealth of information on the types of RNA that trigger RIG-I, much less is known about the nature of the RNAs that act as agonists for MDA5. In order to identify which RNA species triggers MDA5 activation during infection, we isolated viral ssRNA and replicative intermediates of RNA from positive sense ssRNA viruses. We reveal that MDA5 recognises not the genomic ssRNA but the dsRNA generated by the replication of these viruses. Furthermore, using fluorescent imaging we present the first report of the visualisation of dsRNA and MDA5, which provides unique evidence of the relationship between viral dsRNA and MDA5 and proves without a doubt that MDA5 is the key sensor for the dsRNA replicative intermediate form of positive sense ssRNA viruses.


Assuntos
RNA Helicases DEAD-box , Vírus de RNA , RNA de Cadeia Dupla , Adulto , Proteína DEAD-box 58 , RNA Helicases DEAD-box/genética , RNA Helicases DEAD-box/imunologia , RNA Helicases DEAD-box/metabolismo , Enterovirus/genética , Enterovirus/metabolismo , Feminino , Células HEK293 , Humanos , Imunidade Inata/genética , Infecções/genética , Infecções/imunologia , Helicase IFIH1 Induzida por Interferon , Melanoma/genética , Melanoma/imunologia , Células Musculares/citologia , Células Musculares/metabolismo , Vírus de RNA/genética , Vírus de RNA/imunologia , Vírus de RNA/metabolismo , RNA de Cadeia Dupla/genética , RNA de Cadeia Dupla/imunologia , RNA de Cadeia Dupla/metabolismo , Receptores Imunológicos , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Replicação Viral/genética
18.
J Immunol ; 188(8): 3893-902, 2012 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-22427633

RESUMO

Myristoylated alanine-rich C kinase substrate (MARCKS) is an intrinsically unfolded protein with a conserved cationic effector domain, which mediates the cross-talk between several signal transduction pathways. Transcription of MARCKS is increased by stimulation with bacterial LPS. We determined that MARCKS and MARCKS-related protein specifically bind to LPS and that the addition of the MARCKS effector peptide inhibited LPS-induced production of TNF-α in mononuclear cells. The LPS binding site within the effector domain of MARCKS was narrowed down to a heptapeptide that binds to LPS in an extended conformation as determined by nuclear magnetic resonance spectroscopy. After LPS stimulation, MARCKS moved from the plasma membrane to FYVE-positive endosomes, where it colocalized with LPS. MARCKS-deficient mouse embryonic fibroblasts (MEFs) responded to LPS with increased IL-6 production compared with the matched wild-type MEFs. Similarly, small interfering RNA knockdown of MARCKS also increased LPS signaling, whereas overexpression of MARCKS inhibited LPS signaling. TLR4 signaling was enhanced by the ablation of MARCKS, which had no effect on stimulation by TLR2, TLR3, and TLR5 agonists. These findings demonstrate that MARCKS contributes to the negative regulation of the cellular response to LPS.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/imunologia , Leucócitos Mononucleares/imunologia , Lipopolissacarídeos/imunologia , Proteínas de Membrana/imunologia , Sequência de Aminoácidos , Animais , Sítios de Ligação , Endossomos/imunologia , Fibroblastos/imunologia , Regulação da Expressão Gênica , Células HEK293 , Humanos , Imunidade Inata , Interleucina-6/biossíntese , Interleucina-6/imunologia , Peptídeos e Proteínas de Sinalização Intracelular/química , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Lipopolissacarídeos/metabolismo , Lipopolissacarídeos/farmacologia , Proteínas de Membrana/química , Proteínas de Membrana/metabolismo , Camundongos , Dados de Sequência Molecular , Substrato Quinase C Rico em Alanina Miristoilada , Peptídeos/química , Peptídeos/farmacologia , Ligação Proteica , Transporte Proteico/imunologia , RNA Interferente Pequeno/genética , Transdução de Sinais , Receptores Toll-Like/genética , Receptores Toll-Like/imunologia , Fator de Necrose Tumoral alfa/biossíntese , Fator de Necrose Tumoral alfa/imunologia
19.
Virulence ; 3(2): 136-45, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22460642

RESUMO

Bacterial cell wall components, such LPS and LTA, are potent initiators of an inflammatory response that can lead to septic shock. The advances in the past were centered around membrane-bound receptors and intracellular events, but our understanding of the initial interactions of these bacterial components with serum proteins as they enter the bloodstream remain unclear. In this study we identified several serum proteins, which are involved in the innate recognition of bacterial products. Using affinity chromatography and mass spectrometry we performed proteomic analysis of LPS- and LTA-binding serum proteins. We isolated proteins from normal serum that can interact with LPS and LTA. Fluorescent binding experiments and cytokine assays revealed that serum proteins, such as apolipoprotein, LDL, transferrin and holotransferrin could neutralize LPS/LTA binding as well as the subsequent inflammatory response, suggesting that serum proteins modulate LPS/LTA-induced responses. When compared with the proteomic profile of serum from septic patients it was shown that these proteins were in lower abundance. Investigation of serum proteins in 25 critically ill patients with a mortality rate of 40% showed statistically higher levels of these proteins in survivors. Patients surviving sepsis had statistically significant higher levels of apolipoprotein, albumin, LDL, transferrin and holotransferrin than individuals that succumbed, suggesting that these proteins have an inhibitory effect on LPS/LTA-induced inflammatory responses and in their absence there might be an augmented inflammatory response in sepsis.


Assuntos
Proteínas Sanguíneas/metabolismo , Lipopolissacarídeos/imunologia , Lipopolissacarídeos/metabolismo , Sepse/imunologia , Sepse/patologia , Ácidos Teicoicos/imunologia , Ácidos Teicoicos/metabolismo , Adulto , Idoso , Proteínas Sanguíneas/química , Proteínas Sanguíneas/imunologia , Proteínas Sanguíneas/isolamento & purificação , Cromatografia de Afinidade , Estado Terminal , Citocinas/metabolismo , Feminino , Humanos , Masculino , Espectrometria de Massas , Pessoa de Meia-Idade , Ligação Proteica , Proteoma/análise , Análise de Sobrevida , Adulto Jovem
20.
Methods Mol Biol ; 799: 253-66, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-21993651

RESUMO

The innate immune system utilises a set of receptors, called pattern recognition receptors (PRRs), in order to recognise specific molecular patterns or motifs called pathogen-associated molecular patterns (PAMPs) on invading pathogens. The toll-like receptor (TLR) family of proteins is an integral part of the mammalian innate immune system. We are now beginning to decipher which TLRs are involved in the recognition of particular microbial patterns, but questions remain as to the homo- and heterotypic associations that TLRs form and how these associations affect their activation. Technical advances in fluorescence microscopy has enabled us to investigate the functional associations of TLRs and other PPRs in living cells in response to different pathogens using non-invasive fluorescence imaging methods. In this chapter, we will describe some of the fluorescent imaging techniques, such as FRET and FRAP, that we employ in order to study PAMP-PRR associations.


Assuntos
Recuperação de Fluorescência Após Fotodegradação/métodos , Transferência Ressonante de Energia de Fluorescência/métodos , Imunidade Inata/imunologia , Microscopia de Fluorescência/métodos , Receptores Toll-Like/ultraestrutura , Anticorpos/metabolismo , Linhagem Celular , Humanos , Coloração e Rotulagem/métodos , Receptores Toll-Like/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...