Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
1.
Cells ; 12(23)2023 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-38067135

RESUMEN

The complement system mediates diverse regulatory immunological functions. C5aR2, an enigmatic receptor for anaphylatoxin C5a, has been shown to modulate PRR-dependent pro-inflammatory cytokine secretion in human macrophages. However, the specific downstream targets and underlying molecular mechanisms are less clear. In this study, CRISPR-Cas9 was used to generate macrophage models lacking C5aR2, which were used to probe the role of C5aR2 in the context of PRR stimulation. cGAS and STING-induced IFN-ß secretion was significantly increased in C5aR2 KO THP-1 cells and C5aR2-edited primary human monocyte-derived macrophages, and STING and IRF3 expression were increased, albeit not significantly, in C5aR2 KO cell lines implicating C5aR2 as a regulator of the IFN-ß response to cGAS-STING pathway activation. Transcriptomic analysis by RNAseq revealed that nucleic acid sensing and antiviral signalling pathways were significantly up-regulated in C5aR2 KO THP-1 cells. Altogether, these data suggest a link between C5aR2 and nucleic acid sensing in human macrophages. With further characterisation, this relationship may yield therapeutic options in interferon-related pathologies.


Asunto(s)
Interferón beta , Macrófagos , Proteínas de la Membrana , Ácidos Nucleicos , Receptor de Anafilatoxina C5a , Humanos , Interferón beta/metabolismo , Macrófagos/metabolismo , Ácidos Nucleicos/metabolismo , Nucleotidiltransferasas/metabolismo , Transducción de Señal , Receptor de Anafilatoxina C5a/metabolismo , Proteínas de la Membrana/metabolismo
2.
Front Immunol ; 13: 918551, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36248901

RESUMEN

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.


Asunto(s)
Inflamasomas , Proteína con Dominio Pirina 3 de la Familia NLR , Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Humanos , Inflamasomas/metabolismo , Interleucina-18/metabolismo , Interleucina-1beta/metabolismo , Macrófagos , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo
3.
Nat Commun ; 13(1): 1406, 2022 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-35301296

RESUMEN

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.


Asunto(s)
Enterovirus , Virus ARN , Humanos , Orgánulos , Rhinovirus , Replicación Viral/fisiología
4.
Immunology ; 163(4): 345-347, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34251050

RESUMEN

In this issue, we introduce the second part of our review series focusing on lesser-known enigmatic inflammasomes. This part of the collection introduces one more under-studied NLR, NLRP7, and not only its role as a regulator of inflammation in response to bacterial infections but also its non-inflammasome role in early pregnancy. In addition, the enigmatic function of extracellular ASC specks is also introduced, where extracellular ASC specks are presented as 'danger signals' to propagate inflammation. The series is concluded with an article that reviews the immunometabolic regulation of all of these lesser-known NLRs, demonstrating that metabolic regulation of inflammasome activation is central for the whole NLR family. These three reviews, together with the four articles that were published in the first part of the series in December 2020, offer new insights into the complex functions of NLRs, well beyond the well-known NLRP3. The review series as a whole provides a thought-provoking platform with some of the latest findings in the NLR field and sparks our imagination into what might be discovered in this space in the future.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Infecciones Bacterianas/inmunología , Inflamasomas/metabolismo , Inflamación/inmunología , Embarazo/inmunología , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Femenino , Humanos , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Transducción de Señal
5.
PLoS Pathog ; 17(4): e1009417, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33861800

RESUMEN

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.


Asunto(s)
Infecciones por VIH/virología , Inflamasomas/inmunología , Interleucina-18/metabolismo , Interleucina-1beta/metabolismo , Macrófagos/virología , Fragmentos de Péptidos/metabolismo , Comunicación Celular/genética , Comunicación Celular/inmunología , Expresión Génica/genética , Expresión Génica/inmunología , Infecciones por VIH/metabolismo , Humanos , Inmunidad Innata/genética , Inmunidad Innata/inmunología , Inflamasomas/metabolismo , Macrófagos/inmunología , Proteína Inhibidora de la Apoptosis Neuronal/genética , Transducción de Señal/inmunología
6.
Immunology ; 162(3): 249-251, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33590488

RESUMEN

Inflammasomes are generally thought of as large protein complexes that assemble in the cytosol in response to danger such as tissue damage or infection; once activated, they trigger production of inflammatory cytokines and drive cells towards a pro-inflammatory death - termed pyroptosis. Inflammasome activation is a two-step process; priming or Signal 1 (typically via Toll or other receptors that activate NF-kB) induces transcription of pro-forms of IL-1ß and IL-18, while activation or Signal 2 (by many effectors, including a number of bacterial toxins that form pores in cell membranes) comprises activation of caspase-1 in the inflammasome that in turn cleaves pro-IL-1ß and pro-IL-18 and induces release of the active pro-inflammatory cytokines. The most studied inflammasome is the NLRP3 inflammasome, but in addition to NLRP3, there are several lesser-known or enigmatic inflammasomes whose functions seem to range from non-canonical inflammasome activation, pathogen/damage, suppression or modulation of inflammation and even embryonic development. In this review series, which will be presented in two parts, we will focus on lesser-known inflammasomes, such as NLRP6 (non-canonical inflammasome activation), NLRP9 (restricting rotavirus infection in intestinal epithelial cells), NLRX1 (negative regulators of inflammation), NLRC5 (regulating antigen presentation) and NLRP7 (sensing of bacterial lipoproteins). Although the function of NLRP3 is understood, the functions of these lesser-studied inflammasomes are largely unstudied. Given that after a decade of research, new inflammasome and new inflammasome activators are still being discovered indicates that there is a lot more that we need to find out in the NLR field. Only by understanding all of the members of the NLR family, will we be able to target them therapeutically in the future.


Asunto(s)
Inflamasomas/metabolismo , Mediadores de Inflamación/metabolismo , Inflamación/metabolismo , Animales , Humanos , Inflamación/inmunología , Inflamación/patología , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Piroptosis , Transducción de Señal
7.
J Invest Dermatol ; 139(1): 157-166, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30048652

RESUMEN

Human T-cell leukemia virus type 1 (HTLV-1) propagates within and between individuals via cell-to-cell transmission, and primary infection typically occurs across juxtaposed mucosal surfaces during breastfeeding or sexual intercourse. It is therefore likely that dendritic cells (DCs) are among the first potential targets for HTLV-1. However, it remains unclear how DCs contribute to virus transmission and dissemination in the early stages of infection. We show that an HTLV-1-infected cell line (MT-2) and naturally infected CD4+ T cells transfer p19+ viral particles to the surface of allogeneic DCs via cell-to-cell contacts. Similarly organized cell-to-cell contacts also facilitate DC-mediated transfer of HTLV-1 to autologous CD4+ T cells. These findings shed light on the cellular structures involved in anterograde and retrograde transmission and suggest a key role for DCs in the natural history and pathogenesis of HTLV-1 infection.


Asunto(s)
Linfocitos T CD4-Positivos/virología , Células Dendríticas/virología , Virus Linfotrópico T Tipo 1 Humano/fisiología , Leucemia de Células T/patología , Replicación Viral , Linfocitos T CD4-Positivos/inmunología , Células Dendríticas/metabolismo , Células Dendríticas/ultraestructura , Humanos , Leucemia de Células T/metabolismo , Leucemia de Células T/virología , Microscopía Electrónica de Rastreo , Células Tumorales Cultivadas
8.
J Biol Chem ; 293(15): 5509-5521, 2018 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-29463677

RESUMEN

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.


Asunto(s)
Colesterol/biosíntesis , Acido Graso Sintasa Tipo I/metabolismo , Activación de Macrófagos , Macrófagos/enzimología , Transducción de Señal , Acilcoenzima A/metabolismo , Animales , Inflamación/enzimología , Ratones , Ácido Palmítico/metabolismo , Receptor Toll-Like 4/metabolismo
9.
Trends Mol Med ; 23(2): 165-180, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28109721

RESUMEN

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.


Asunto(s)
Inflamasomas/inmunología , Inflamación/inmunología , Enfermedad de Alzheimer/inmunología , Animales , Aterosclerosis/inmunología , Enfermedades Autoinflamatorias Hereditarias/inmunología , Humanos , Inmunidad Innata , Interleucina-1beta/inmunología , Enfermedades Metabólicas/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Neuroinmunomodulación , Obesidad/inmunología , Receptores Toll-Like/inmunología
10.
Immunology ; 147(2): 152-64, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26572245

RESUMEN

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.


Asunto(s)
Activación de Complemento , Proteínas del Sistema Complemento/inmunología , Inmunidad Innata , Inflamasomas/inmunología , Inflamación/inmunología , Animales , Antiinflamatorios/uso terapéutico , Activación de Complemento/efectos de los fármacos , Proteínas del Sistema Complemento/metabolismo , Humanos , Inmunidad Innata/efectos de los fármacos , Inflamasomas/efectos de los fármacos , Inflamasomas/metabolismo , Inflamación/metabolismo , Inflamación/prevención & control , Mediadores de Inflamación/inmunología , Mediadores de Inflamación/metabolismo , Ligandos , Receptores Inmunológicos/inmunología , Receptores Inmunológicos/metabolismo , Transducción de Señal
11.
Trends Microbiol ; 22(10): 580-8, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24986075

RESUMEN

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.


Asunto(s)
Inmunidad Innata/inmunología , Inflamasomas/inmunología , Hierro/metabolismo , Pulmón/inmunología , Humanos , Inflamación/inmunología , Activación Viral
12.
Cell Host Microbe ; 15(6): 768-78, 2014 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-24922578

RESUMEN

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.


Asunto(s)
Disbiosis/microbiología , Periodontitis/microbiología , Porphyromonas gingivalis/patogenicidad , Receptor de Anafilatoxina C5a/metabolismo , Receptor Toll-Like 2/metabolismo , Animales , Infecciones por Bacteroidaceae/inmunología , Proteínas del Sistema Complemento/inmunología , Disbiosis/inmunología , Interacciones Huésped-Patógeno/inmunología , Ratones , Ratones Mutantes , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/metabolismo , Neutrófilos/inmunología , Neutrófilos/microbiología , Periodontitis/inmunología , Fagocitosis , Fosfatidilinositol 3-Quinasas/metabolismo , Porphyromonas gingivalis/inmunología , Receptor de Anafilatoxina C5a/genética , Receptor de Anafilatoxina C5a/inmunología , Transducción de Señal , Receptor Toll-Like 2/genética , Receptor Toll-Like 2/inmunología
13.
J Biol Chem ; 289(22): 15309-18, 2014 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-24737331

RESUMEN

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.


Asunto(s)
Inflamación/inmunología , Macrófagos/inmunología , Receptores de IgG/inmunología , Receptor Toll-Like 4/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Sitios de Unión , Células CHO , Línea Celular , Cricetulus , Dimerización , Femenino , Humanos , Inflamación/metabolismo , Macrófagos/citología , Microdominios de Membrana/inmunología , Microdominios de Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Receptores de IgG/metabolismo , Receptor Toll-Like 4/química , Receptor Toll-Like 4/metabolismo , Células U937
14.
Am J Obstet Gynecol ; 210(2): 122.e1-122.e10, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24080302

RESUMEN

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.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Herpes Genital/virología , Herpesvirus Humano 2/fisiología , Proteínas Nucleares/fisiología , Fosfoproteínas/fisiología , Receptor Toll-Like 2/fisiología , Activación Viral/fisiología , Femenino , Herpes Genital/inmunología , Humanos , Inmunidad Innata , Proteínas de Unión al ARN , Receptor Toll-Like 9/fisiología , Vagina/inmunología , Vagina/virología
15.
Am J Respir Cell Mol Biol ; 49(6): 923-34, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23815151

RESUMEN

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.


Asunto(s)
Bronquios/metabolismo , Bronquios/virología , Señalización del Calcio/fisiología , Proteínas Portadoras/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Rhinovirus/patogenicidad , Asma/etiología , Asma/metabolismo , Asma/virología , Proteínas Portadoras/antagonistas & inhibidores , Proteínas Portadoras/genética , Células Cultivadas , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/virología , Técnicas de Silenciamiento del Gen , Aparato de Golgi/metabolismo , Aparato de Golgi/virología , Humanos , Inflamasomas/metabolismo , Interleucina-1beta/biosíntesis , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Péptidos y Proteínas de Señalización Intracelular/genética , Proteína con Dominio Pirina 3 de la Familia NLR , Infecciones por Picornaviridae/complicaciones , Infecciones por Picornaviridae/metabolismo , Infecciones por Picornaviridae/virología , Enfermedad Pulmonar Obstructiva Crónica/etiología , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/virología , Rhinovirus/genética , Rhinovirus/metabolismo , Proteínas Virales/metabolismo
16.
PLoS One ; 8(4): e61199, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23593431

RESUMEN

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.


Asunto(s)
Amnios/patología , Células Epiteliales/metabolismo , Células Epiteliales/microbiología , Receptores Toll-Like/metabolismo , Infecciones por Ureaplasma/metabolismo , Ureaplasma urealyticum/fisiología , Ureaplasma/fisiología , Membrana Celular/metabolismo , Citocinas/metabolismo , Endocitosis , Endosomas/metabolismo , Endosomas/microbiología , Transferencia Resonante de Energía de Fluorescencia , Gangliósido G(M1)/metabolismo , Silenciador del Gen , Células HEK293 , Humanos , Espacio Intracelular/microbiología , Microdominios de Membrana/metabolismo , Factor 88 de Diferenciación Mieloide/metabolismo , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 6/metabolismo , Receptor Toll-Like 9/metabolismo , Infecciones por Ureaplasma/microbiología
17.
J Cell Sci ; 126(Pt 13): 2903-13, 2013 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-23613465

RESUMEN

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.


Asunto(s)
Calcio/metabolismo , Proteínas Portadoras/genética , Membrana Celular/efectos de los fármacos , Complejo de Ataque a Membrana del Sistema Complemento/farmacología , Células Epiteliales/efectos de los fármacos , Inflamasomas/efectos de los fármacos , Mucosa Respiratoria/efectos de los fármacos , Calcio/agonistas , Canales de Calcio/genética , Canales de Calcio/metabolismo , Proteínas Portadoras/inmunología , Membrana Celular/inmunología , Retículo Endoplásmico/metabolismo , Células Epiteliales/citología , Células Epiteliales/inmunología , Humanos , Inflamasomas/metabolismo , Receptores de Inositol 1,4,5-Trifosfato/genética , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Interleucina-1beta/inmunología , Interleucina-1beta/metabolismo , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Potencial de la Membrana Mitocondrial/inmunología , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR , Cultivo Primario de Células , Mucosa Respiratoria/citología , Mucosa Respiratoria/inmunología , Canal Liberador de Calcio Receptor de Rianodina/genética , Canal Liberador de Calcio Receptor de Rianodina/metabolismo
19.
Thorax ; 68(1): 66-75, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23229815

RESUMEN

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.


Asunto(s)
Proteínas Portadoras/metabolismo , Inflamasomas/metabolismo , Virus Sincitial Respiratorio Humano/genética , Adulto , Bronquiolitis Viral/genética , Bronquiolitis Viral/fisiopatología , Proteínas Portadoras/genética , Células Cultivadas , Células Epiteliales/metabolismo , Células Epiteliales/fisiología , Humanos , Inflamasomas/genética , Pulmón/citología , Masculino , Microscopía Confocal , Proteína con Dominio Pirina 3 de la Familia NLR , Neumonía Viral/metabolismo , Neumonía Viral/fisiopatología , ARN Viral/genética , ARN Viral/metabolismo , Infecciones por Virus Sincitial Respiratorio/genética , Infecciones por Virus Sincitial Respiratorio/fisiopatología , Virus Sincitial Respiratorio Humano/fisiología , Sensibilidad y Especificidad , Transducción de Señal/genética
20.
J Cell Sci ; 125(Pt 20): 4761-9, 2012 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-22797917

RESUMEN

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.


Asunto(s)
ARN Helicasas DEAD-box , Virus ARN , ARN Bicatenario , Adulto , Proteína 58 DEAD Box , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/inmunología , ARN Helicasas DEAD-box/metabolismo , Enterovirus/genética , Enterovirus/metabolismo , Femenino , Células HEK293 , Humanos , Inmunidad Innata/genética , Infecciones/genética , Infecciones/inmunología , Helicasa Inducida por Interferón IFIH1 , Melanoma/genética , Melanoma/inmunología , Células Musculares/citología , Células Musculares/metabolismo , Virus ARN/genética , Virus ARN/inmunología , Virus ARN/metabolismo , ARN Bicatenario/genética , ARN Bicatenario/inmunología , ARN Bicatenario/metabolismo , Receptores Inmunológicos , Transducción de Señal/genética , Transducción de Señal/inmunología , Replicación Viral/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...