Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Nat Immunol ; 21(8): 880-891, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32541830

RESUMEN

Bacterial lipopolysaccharide triggers human caspase-4 (murine caspase-11) to cleave gasdermin-D and induce pyroptotic cell death. How lipopolysaccharide sequestered in the membranes of cytosol-invading bacteria activates caspases remains unknown. Here we show that in interferon-γ-stimulated cells guanylate-binding proteins (GBPs) assemble on the surface of Gram-negative bacteria into polyvalent signaling platforms required for activation of caspase-4. Caspase-4 activation is hierarchically controlled by GBPs; GBP1 initiates platform assembly, GBP2 and GBP4 control caspase-4 recruitment, and GBP3 governs caspase-4 activation. In response to cytosol-invading bacteria, activation of caspase-4 through the GBP platform is essential to induce gasdermin-D-dependent pyroptosis and processing of interleukin-18, thereby destroying the replicative niche for intracellular bacteria and alerting neighboring cells, respectively. Caspase-11 and GBPs epistatically protect mice against lethal bacterial challenge. Multiple antagonists of the pathway encoded by Shigella flexneri, a cytosol-adapted bacterium, provide compelling evolutionary evidence for the importance of the GBP-caspase-4 pathway in antibacterial defense.


Asunto(s)
Caspasas Iniciadoras/inmunología , Proteínas de Unión al GTP/inmunología , Infecciones por Bacterias Gramnegativas/inmunología , Inflamasomas/inmunología , Transducción de Señal/inmunología , Animales , Bacterias Gramnegativas/inmunología , Células HeLa , Humanos , Lipopolisacáridos/inmunología , Ratones , Piroptosis/inmunología
2.
Nature ; 619(7971): 819-827, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37438530

RESUMEN

Understanding protective immunity to COVID-19 facilitates preparedness for future pandemics and combats new SARS-CoV-2 variants emerging in the human population. Neutralizing antibodies have been widely studied; however, on the basis of large-scale exome sequencing of protected versus severely ill patients with COVID-19, local cell-autonomous defence is also crucial1-4. Here we identify phospholipid scramblase 1 (PLSCR1) as a potent cell-autonomous restriction factor against live SARS-CoV-2 infection in parallel genome-wide CRISPR-Cas9 screens of human lung epithelia and hepatocytes before and after stimulation with interferon-γ (IFNγ). IFNγ-induced PLSCR1 not only restricted SARS-CoV-2 USA-WA1/2020, but was also effective against the Delta B.1.617.2 and Omicron BA.1 lineages. Its robust activity extended to other highly pathogenic coronaviruses, was functionally conserved in bats and mice, and interfered with the uptake of SARS-CoV-2 in both the endocytic and the TMPRSS2-dependent fusion routes. Whole-cell 4Pi single-molecule switching nanoscopy together with bipartite nano-reporter assays found that PLSCR1 directly targeted SARS-CoV-2-containing vesicles to prevent spike-mediated fusion and viral escape. A PLSCR1 C-terminal ß-barrel domain-but not lipid scramblase activity-was essential for this fusogenic blockade. Our mechanistic studies, together with reports that COVID-associated PLSCR1 mutations are found in some susceptible people3,4, identify an anti-coronavirus protein that interferes at a late entry step before viral RNA is released into the host-cell cytosol.


Asunto(s)
COVID-19 , Proteínas de Transferencia de Fosfolípidos , SARS-CoV-2 , Animales , Humanos , Ratones , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Quirópteros , COVID-19/inmunología , COVID-19/metabolismo , COVID-19/prevención & control , COVID-19/virología , Secuenciación del Exoma , Hepatocitos/inmunología , Hepatocitos/metabolismo , Interferón gamma/inmunología , Pulmón/inmunología , Pulmón/metabolismo , Fusión de Membrana , Proteínas de Transferencia de Fosfolípidos/química , Proteínas de Transferencia de Fosfolípidos/genética , Proteínas de Transferencia de Fosfolípidos/inmunología , Proteínas de Transferencia de Fosfolípidos/metabolismo , SARS-CoV-2/clasificación , SARS-CoV-2/inmunología , SARS-CoV-2/metabolismo , SARS-CoV-2/patogenicidad , Internalización del Virus
3.
Nat Immunol ; 17(5): 481-9, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-27092805

RESUMEN

Traditional views of the inflammasome highlight the assembly of pre-existing core components shortly after infection or tissue damage. Emerging work, however, suggests that the inflammasome machinery is also subject to 'tunable' or inducible signals that might accelerate its autocatalytic properties and dictate where inflammasome assembly takes place in the cell. Many of these signals operate downstream of interferon receptors to elicit inflammasome regulators, including a new family of interferon-induced GTPases called 'guanylate-binding proteins' (GBPs). Here we investigate the critical roles of interferon-induced GBPs in directing inflammasome subtype-specific responses and their consequences for cell-autonomous immunity to a wide variety of microbial pathogens. We discuss emerging mechanisms of action and the potential effect of these GBPs on predisposition to sepsis and other infectious or inflammatory diseases.


Asunto(s)
Proteínas de Unión al GTP/inmunología , Inflamasomas/inmunología , Interferones/inmunología , Transducción de Señal/inmunología , Animales , Resistencia a la Enfermedad/genética , Resistencia a la Enfermedad/inmunología , Proteínas de Unión al GTP/clasificación , Proteínas de Unión al GTP/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Infecciones/inmunología , Infecciones/microbiología , Infecciones/parasitología , Inflamasomas/genética , Inflamasomas/metabolismo , Interferones/metabolismo , Listeria monocytogenes/inmunología , Listeria monocytogenes/fisiología , Ratones , Modelos Inmunológicos , Filogenia , Transducción de Señal/genética , Toxoplasma/inmunología , Toxoplasma/fisiología
4.
Nature ; 607(7918): 339-344, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35768511

RESUMEN

Extreme weather conditions associated with climate change affect many aspects of plant and animal life, including the response to infectious diseases. Production of salicylic acid (SA), a central plant defence hormone1-3, is particularly vulnerable to suppression by short periods of hot weather above the normal plant growth temperature range via an unknown mechanism4-7. Here we show that suppression of SA production in Arabidopsis thaliana at 28 °C is independent of PHYTOCHROME B8,9 (phyB) and EARLY FLOWERING 310 (ELF3), which regulate thermo-responsive plant growth and development. Instead, we found that formation of GUANYLATE BINDING PROTEIN-LIKE 3 (GBPL3) defence-activated biomolecular condensates11 (GDACs) was reduced at the higher growth temperature. The altered GDAC formation in vivo is linked to impaired recruitment of GBPL3 and SA-associated Mediator subunits to the promoters of CBP60g and SARD1, which encode master immune transcription factors. Unlike many other SA signalling components, including the SA receptor and biosynthetic genes, optimized CBP60g expression was sufficient to broadly restore SA production, basal immunity and effector-triggered immunity at the elevated growth temperature without significant growth trade-offs. CBP60g family transcription factors are widely conserved in plants12. These results have implications for safeguarding the plant immune system as well as understanding the concept of the plant-pathogen-environment disease triangle and the emergence of new disease epidemics in a warming climate.


Asunto(s)
Aclimatación , Proteínas de Arabidopsis , Arabidopsis , Ambiente , Calentamiento Global , Inmunidad de la Planta , Temperatura , Arabidopsis/crecimiento & desarrollo , Arabidopsis/inmunología , Arabidopsis/metabolismo , Proteínas de Arabidopsis/genética , Proteínas de Arabidopsis/metabolismo , Proteínas de Unión a Calmodulina/genética , Regulación de la Expresión Génica de las Plantas , Calentamiento Global/estadística & datos numéricos , Interacciones Huésped-Patógeno , Fitocromo B , Enfermedades de las Plantas/genética , Inmunidad de la Planta/genética , Ácido Salicílico/metabolismo , Factores de Transcripción
5.
Nature ; 594(7863): 424-429, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34040255

RESUMEN

Liquid-liquid phase separation (LLPS) has emerged as a central paradigm for understanding how membraneless organelles compartmentalize diverse cellular activities in eukaryotes1-3. Here we identify a superfamily of plant guanylate-binding protein (GBP)-like GTPases (GBPLs) that assemble LLPS-driven condensates within the nucleus to protect against infection and autoimmunity. In Arabidopsis thaliana, two members of this family-GBPL1 and GBPL3-undergo phase-transition behaviour to control transcriptional responses as part of an allosteric switch that is triggered by exposure to biotic stress. GBPL1, a pseudo-GTPase, sequesters catalytically active GBPL3 under basal conditions but is displaced by GBPL3 LLPS when it enters the nucleus following immune cues to drive the formation of unique membraneless organelles termed GBPL defence-activated condensates (GDACs) that we visualized by in situ cryo-electron tomography. Within these mesoscale GDAC structures, native GBPL3 directly bound defence-gene promoters and recruited specific transcriptional coactivators of the Mediator complex and RNA polymerase II machinery to massively reprogram host gene expression for disease resistance. Together, our study identifies a GBPL circuit that reinforces the biological importance of phase-separated condensates, in this case, as indispensable players in plant defence.


Asunto(s)
Arabidopsis/inmunología , Núcleo Celular/química , Núcleo Celular/metabolismo , Proteínas de Unión al GTP/metabolismo , Proteínas Intrínsecamente Desordenadas/metabolismo , Transición de Fase , Inmunidad de la Planta , Arabidopsis/genética , Arabidopsis/metabolismo , Arabidopsis/ultraestructura , Núcleo Celular/genética , Núcleo Celular/ultraestructura , Cromatina/genética , Microscopía por Crioelectrón , Proteínas de Unión al GTP/química , Proteínas de Unión al GTP/ultraestructura , Regulación de la Expresión Génica de las Plantas/genética , Proteínas Intrínsecamente Desordenadas/química , Proteínas Intrínsecamente Desordenadas/ultraestructura , Complejo Mediador , Familia de Multigenes/genética , Orgánulos/química , Orgánulos/inmunología , Orgánulos/metabolismo , Orgánulos/ultraestructura , Células Vegetales/química , Células Vegetales/inmunología , Células Vegetales/metabolismo , Células Vegetales/ultraestructura , Enfermedades de las Plantas/inmunología , Inmunidad de la Planta/genética , Regiones Promotoras Genéticas/genética , ARN Polimerasa II/metabolismo , Transcripción Genética
6.
Nat Immunol ; 10(8): 907-17, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19620982

RESUMEN

Vertebrate immunity to infection enlists a newly identified family of 47-kilodalton immunity-related GTPases (IRGs). One IRG in particular, Irgm1, is essential for macrophage host defense against phagosomal pathogens, including Mycobacterium tuberculosis (Mtb). Here we show that Irgm1 targets the mycobacterial phagosome through lipid-mediated interactions with phosphatidylinositol-3,4-bisphosphate (PtdIns(3,4)P(2)) and PtdIns(3,4,5)P(3). An isolated Irgm1 amphipathic helix conferred lipid binding in vitro and in vivo. Substitutions in this region blocked phagosome recruitment and failed to complement the antimicrobial defect in Irgm1(-/-) macrophages. Removal of PtdIns(3,4,5)P(3) or inhibition of class I phosphatidylinositol-3-OH kinase (PI(3)K) mimicked this effect in wild-type cells. Cooperation between Irgm1 and PI(3)K further facilitated the engagement of Irgm1 with its fusogenic effectors at the site of infection, thereby ensuring pathogen-directed responses during innate immunity.


Asunto(s)
Proteínas de Unión al GTP/metabolismo , Mycobacterium tuberculosis/fisiología , Fagosomas/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Animales , Células Cultivadas , Proteínas de Unión al GTP/genética , Inmunidad Innata , Interferón gamma/fisiología , Membranas Intracelulares/metabolismo , Lisosomas/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/microbiología , Ratones , Mycobacterium tuberculosis/inmunología , Fosfatidilinositol 3-Quinasas/metabolismo , Unión Proteica , Estructura Secundaria de Proteína , Transporte de Proteínas/fisiología , Proteínas SNARE/metabolismo , Transducción de Señal
7.
Immunity ; 31(2): 232-44, 2009 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-19699172

RESUMEN

Dendritic cells (DCs) have the striking ability to cross-present exogenous antigens in association with major histocompatibility complex (MHC) class I to CD8(+) T cells. However, the intracellular pathways underlying cross-presentation remain ill defined. Current models involve cytosolic proteolysis of antigens by the proteasome and peptide import into endoplasmic reticulum (ER) or phagosomal lumen by the transporters associated with antigen processing (TAP1 and TAP2). Here, we show that DCs expressed an ER-resident 47 kDa immune-related GTPase, Igtp (Irgm3). Igtp resides on ER and lipid body (LB) membranes where it binds the LB coat component ADFP. Inactivation of genes encoding for either Igtp or ADFP led to defects in LB formation in DCs and severely impaired cross-presentation of phagocytosed antigens to CD8(+) T cells but not antigen presentation to CD4(+) T cells. We thus define a new role for LB organelles in regulating cross-presentation of exogenous antigens to CD8(+) T lymphocytes in DCs.


Asunto(s)
Presentación de Antígeno/inmunología , Reactividad Cruzada , Células Dendríticas/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Lípidos/inmunología , Fagocitosis , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Retículo Endoplásmico/inmunología , GTP Fosfohidrolasas/genética , GTP Fosfohidrolasas/inmunología , GTP Fosfohidrolasas/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Ratones Transgénicos , Perilipina-2
8.
Nature ; 551(7680): 303-305, 2017 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-29072295
9.
J Biol Chem ; 291(3): 1123-36, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26555265

RESUMEN

Many immunostimulants act as vaccine adjuvants via activation of the innate immune system, although in many cases it is unclear which specific molecules contribute to the stimulatory activity. QS-21 is a defined, highly purified, and soluble saponin adjuvant currently used in licensed and exploratory vaccines, including vaccines against malaria, cancer, and HIV-1. However, little is known about the mechanisms of cellular activation induced by QS-21. We observed QS-21 to elicit caspase-1-dependent IL-1ß and IL-18 release in antigen-presenting cells such as macrophages and dendritic cells when co-stimulated with the TLR4-agonist adjuvant monophosphoryl lipid A. Furthermore, our data suggest that the ASC-NLRP3 inflammasome is responsible for QS-21-induced IL-1ß/IL-18 release. At higher concentrations, QS-21 induced macrophage and dendritic cell death in a caspase-1-, ASC-, and NLRP3-independent manner, whereas the presence of cholesterol rescued cell viability. A nanoparticulate adjuvant that contains QS-21 as part of a heterogeneous mixture of saponins also induced IL-1ß in an NLRP3-dependent manner. Interestingly, despite the role NLRP3 plays for cellular activation in vitro, NLRP3-deficient mice immunized with HIV-1 gp120 and QS-21 showed significantly higher levels of Th1 and Th2 antigen-specific T cell responses and increased IgG1 and IgG2c compared with wild type controls. Thus, we have identified QS-21 as a nonparticulate single molecular saponin that activates the NLRP3 inflammasome, but this signaling pathway may contribute to decreased antigen-specific responses in vivo.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Proteínas Portadoras/metabolismo , Células Dendríticas/efectos de los fármacos , Inmunidad Innata/efectos de los fármacos , Inflamasomas/efectos de los fármacos , Macrófagos/efectos de los fármacos , Saponinas/farmacología , Vacunas contra el SIDA/agonistas , Vacunas contra el SIDA/inmunología , Adyuvantes Inmunológicos/análisis , Adyuvantes Inmunológicos/química , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/inmunología , Proteínas Portadoras/genética , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Células Dendríticas/citología , Células Dendríticas/inmunología , Proteínas de Unión al GTP/genética , Proteínas de Unión al GTP/metabolismo , Proteína gp120 de Envoltorio del VIH/agonistas , Proteína gp120 de Envoltorio del VIH/inmunología , Inmunoglobulina G/análisis , Inmunoglobulina G/biosíntesis , Inflamasomas/inmunología , Inflamasomas/metabolismo , Lípido A/agonistas , Lípido A/análogos & derivados , Lípido A/farmacología , Macrófagos/citología , Macrófagos/inmunología , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR , Saponinas/análisis , Saponinas/química , Solubilidad , Células TH1/efectos de los fármacos , Células TH1/inmunología , Células TH1/metabolismo , Células Th2/efectos de los fármacos , Células Th2/inmunología , Células Th2/metabolismo
10.
Proc Natl Acad Sci U S A ; 110(32): E2997-3006, 2013 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-23882081

RESUMEN

Macrophage migration inhibitory factor (MIF), an innate cytokine encoded in a functionally polymorphic genetic locus, contributes to detrimental inflammation but may be crucial for controlling infection. We explored the role of variant MIF alleles in tuberculosis. In a Ugandan cohort, genetic low expressers of MIF were 2.4-times more frequently identified among patients with Mycobacterium tuberculosis (TB) bacteremia than those without. We also found mycobacteria-stimulated transcription of MIF and serum MIF levels to be correlated with MIF genotype in human macrophages and in a separate cohort of US TB patients, respectively. To determine mechanisms for MIF's protective role, we studied both aerosolized and i.v. models of mycobacterial infection and observed MIF-deficient mice to succumb more quickly with higher organism burden, increased lung pathology, and decreased innate cytokine production (TNF-α, IL-12, IL-10). MIF-deficient animals showed increased pulmonary neutrophil accumulation but preserved adaptive immune response. MIF-deficient macrophages demonstrated decreased cytokine and reactive oxygen production and impaired mycobacterial killing. Transcriptional investigation of MIF-deficient macrophages revealed reduced expression of the pattern recognition receptor dectin-1; restoration of dectin-1 expression recovered innate cytokine production and mycobacterial killing. Our data place MIF in a crucial upstream position in the innate immune response to mycobacteria and suggest that commonly occurring low expression MIF alleles confer an increased risk of TB disease in some populations.


Asunto(s)
Inmunidad Innata/inmunología , Factores Inhibidores de la Migración de Macrófagos/inmunología , Mycobacterium tuberculosis/inmunología , Tuberculosis/inmunología , Adulto , Animales , Línea Celular , Citocinas/inmunología , Citocinas/metabolismo , Femenino , Expresión Génica/inmunología , Genotipo , Humanos , Inmunidad Innata/genética , Lectinas Tipo C/genética , Lectinas Tipo C/inmunología , Lectinas Tipo C/metabolismo , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/microbiología , Factores Inhibidores de la Migración de Macrófagos/sangre , Factores Inhibidores de la Migración de Macrófagos/genética , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/microbiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neutrófilos/inmunología , Neutrófilos/metabolismo , Polimorfismo Genético , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tasa de Supervivencia , Tuberculosis/genética , Tuberculosis/mortalidad , Uganda , Adulto Joven
11.
PLoS Pathog ; 9(7): e1003491, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23853600

RESUMEN

Interferon Regulatory Factor 8 (IRF8) is required for development, maturation and expression of anti-microbial defenses of myeloid cells. BXH2 mice harbor a severely hypomorphic allele at Irf8 (Irf8(R294C)) that causes susceptibility to infection with intracellular pathogens including Mycobacterium tuberculosis. We report that BXH2 are completely resistant to the development of cerebral malaria (ECM) following Plasmodium berghei ANKA infection. Comparative transcriptional profiling of brain RNA as well as chromatin immunoprecipitation and high-throughput sequencing (ChIP-seq) was used to identify IRF8-regulated genes whose expression is associated with pathological acute neuroinflammation. Genes increased by infection were strongly enriched for IRF8 binding sites, suggesting that IRF8 acts as a transcriptional activator in inflammatory programs. These lists were enriched for myeloid-specific pathways, including interferon responses, antigen presentation and Th1 polarizing cytokines. We show that inactivation of several of these downstream target genes (including the Irf8 transcription partner Irf1) confers protection against ECM. ECM-resistance in Irf8 and Irf1 mutants is associated with impaired myeloid and lymphoid cells function, including production of IL12p40 and IFNγ. We note strong overlap between genes bound and regulated by IRF8 during ECM and genes regulated in the lungs of M. tuberculosis infected mice. This IRF8-dependent network contains several genes recently identified as risk factors in acute and chronic human inflammatory conditions. We report a common core of IRF8-bound genes forming a critical inflammatory host-response network.


Asunto(s)
Encéfalo/inmunología , Regulación de la Expresión Génica , Inmunidad Innata , Factores Reguladores del Interferón/metabolismo , Malaria Cerebral/inmunología , Proteínas del Tejido Nervioso/metabolismo , Plasmodium berghei/inmunología , Sustitución de Aminoácidos , Animales , Sitios de Unión , Encéfalo/metabolismo , Encéfalo/parasitología , Células Cultivadas , Citocinas/biosíntesis , Citocinas/sangre , Perfilación de la Expresión Génica , Factor 1 Regulador del Interferón/genética , Factor 1 Regulador del Interferón/metabolismo , Factores Reguladores del Interferón/química , Factores Reguladores del Interferón/genética , Malaria Cerebral/sangre , Malaria Cerebral/metabolismo , Malaria Cerebral/parasitología , Ratones , Ratones Noqueados , Ratones Mutantes , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/genética , Neuronas/inmunología , Neuronas/metabolismo , Neuronas/parasitología , Organismos Libres de Patógenos Específicos , Bazo/inmunología , Bazo/metabolismo , Bazo/patología , Células TH1/inmunología , Células TH1/metabolismo , Células TH1/parasitología
12.
PLoS Pathog ; 9(4): e1003320, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23633952

RESUMEN

IFN-γ activates cells to restrict intracellular pathogens by upregulating cellular effectors including the p65 family of guanylate-binding proteins (GBPs). Here we test the role of Gbp1 in the IFN-γ-dependent control of T. gondii in the mouse model. Virulent strains of T. gondii avoided recruitment of Gbp1 to the parasitophorous vacuole in a strain-dependent manner that was mediated by the parasite virulence factors ROP18, an active serine/threonine kinase, and the pseudokinase ROP5. Increased recruitment of Gbp1 to Δrop18 or Δrop5 parasites was associated with clearance in IFN-γ-activated macrophages in vitro, a process dependent on the autophagy protein Atg5. The increased susceptibility of Δrop18 mutants in IFN-γ-activated macrophages was reverted in Gbp1(-/-) cells, and decreased virulence of this mutant was compensated in Gbp1(-/-) mice, which were also more susceptible to challenge with type II strain parasites of intermediate virulence. These findings demonstrate that Gbp1 plays an important role in the IFN-γ-dependent, cell-autonomous control of toxoplasmosis and predict a broader role for this protein in host defense.


Asunto(s)
Proteínas de Unión al GTP/metabolismo , Interferón gamma/metabolismo , Macrófagos/inmunología , Toxoplasma/inmunología , Toxoplasmosis/inmunología , Animales , Proteína 5 Relacionada con la Autofagia , Células de la Médula Ósea/citología , Células Cultivadas , Proteínas de Unión al GTP/genética , Inmunidad Celular , Activación de Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Protozoarias , Toxoplasma/patogenicidad , Toxoplasmosis/parasitología
13.
J Immunol ; 189(2): 813-8, 2012 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-22675202

RESUMEN

Autophagy is a major innate immune defense pathway in both plants and animals. In mammals, this cascade can be elicited by cytokines (IFN-γ) or pattern recognition receptors (TLRs and nucleotide-binding oligomerization domain-like receptors). Many signaling components in TLR- and nucleotide-binding oligomerization domain-like receptor-induced autophagy are now known; however, those involved in activating autophagy via IFN-γ remain to be elucidated. In this study, we engineered macrophages encoding a tandem fluorescently tagged LC3b (tfLC3) autophagosome reporter along with stably integrated short hairpin RNAs to demonstrate IFN-γ-induced autophagy required JAK 1/2, PI3K, and p38 MAPK but not STAT1. Moreover, the autophagy-related guanosine triphosphatase Irgm1 proved dispensable in both stable tfLC3-expressing RAW 264.7 and tfLC3-transduced Irgm1(-/-) primary macrophages, revealing a novel p38 MAPK-dependent, STAT1-independent autophagy pathway that bypasses Irgm1. These unexpected findings have implications for understanding how IFN-γ-induced autophagy is mobilized within macrophages for inflammation and host defense.


Asunto(s)
Autofagia/inmunología , Interferón gamma/fisiología , Sistema de Señalización de MAP Quinasas/inmunología , Macrófagos/citología , Macrófagos/inmunología , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología , Animales , Autofagia/genética , Células de la Médula Ósea/citología , Células de la Médula Ósea/enzimología , Células de la Médula Ósea/inmunología , Línea Celular , Proteínas de Unión al GTP/deficiencia , Proteínas de Unión al GTP/fisiología , Genes Reporteros/inmunología , Macrófagos/enzimología , Ratones , Ratones Noqueados , Fagosomas/enzimología , Fagosomas/inmunología , Fagosomas/metabolismo , Factor de Transcripción STAT1/deficiencia , Factor de Transcripción STAT1/fisiología
14.
PLoS Genet ; 7(6): e1002097, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21731497

RESUMEN

IRF8 (Interferon Regulatory Factor 8) plays an important role in defenses against intracellular pathogens, including several aspects of myeloid cells function. It is required for ontogeny and maturation of macrophages and dendritic cells, for activation of anti-microbial defenses, and for production of the Th1-polarizing cytokine interleukin-12 (IL-12) in response to interferon gamma (IFNγ) and protection against infection with Mycobacterium tuberculosis. The transcriptional programs and cellular pathways that are regulated by IRF8 in response to IFNγ and that are important for defenses against M. tuberculosis are poorly understood. These were investigated by transcript profiling and chromatin immunoprecipitation on microarrays (ChIP-chip). Studies in primary macrophages identified 368 genes that are regulated by IRF8 in response to IFNγ/CpG and that behave as stably segregating expression signatures (eQTLs) in F2 mice fixed for a wild-type or mutant allele at IRF8. A total of 319 IRF8 binding sites were identified on promoters genome-wide (ChIP-chip) in macrophages treated with IFNγ/CpG, defining a functional G/AGAAnTGAAA motif. An analysis of the genes bearing a functional IRF8 binding site, and showing regulation by IFNγ/CpG in macrophages and/or in M. tuberculosis-infected lungs, revealed a striking enrichment for the pathways of antigen processing and presentation, including multiple structural and enzymatic components of the Class I and Class II MHC (major histocompatibility complex) antigen presentation machinery. Also significantly enriched as IRF8 targets are the group of endomembrane- and phagosome-associated small GTPases of the IRG (immunity-related GTPases) and GBP (guanylate binding proteins) families. These results identify IRF8 as a key regulator of early response pathways in myeloid cells, including phagosome maturation, antigen processing, and antigen presentation by myeloid cells.


Asunto(s)
Presentación de Antígeno , Factores Reguladores del Interferón/inmunología , Pulmón/microbiología , Células Mieloides/inmunología , Tuberculosis Pulmonar/inmunología , Alelos , Animales , Sitios de Unión , Western Blotting , Línea Celular , Inmunoprecipitación de Cromatina , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Genotipo , Factores Reguladores del Interferón/genética , Factores Reguladores del Interferón/metabolismo , Interferón gamma/inmunología , Pulmón/inmunología , Pulmón/metabolismo , Complejo Mayor de Histocompatibilidad , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Mycobacterium tuberculosis/inmunología , Células Mieloides/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Regiones Promotoras Genéticas , Tuberculosis Pulmonar/genética , Tuberculosis Pulmonar/microbiología
15.
Science ; 384(6693): eadl2016, 2024 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-38635718

RESUMEN

Infectious diseases continue to claim many lives. Prevention of morbidity and mortality from these diseases would benefit not just from new medicines and vaccines but also from a better understanding of what constitutes protective immunity. Among the major immune signals that mobilize host defense against infection is interferon-γ (IFN-γ), a protein secreted by lymphocytes. Forty years ago, IFN-γ was identified as a macrophage-activating factor, and, in recent years, there has been a resurgent interest in IFN-γ biology and its role in human defense. Here we assess the current understanding of IFN-γ, revisit its designation as an "interferon," and weigh its prospects as a therapeutic against globally pervasive microbial pathogens.


Asunto(s)
Enfermedades Transmisibles , Interferón gamma , Humanos , Enfermedades Transmisibles/inmunología , Enfermedades Transmisibles/terapia , Interferón gamma/genética , Interferón gamma/metabolismo , Interferón gamma/uso terapéutico , Inmunidad/genética
16.
Science ; 383(6686): eabm9903, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38422126

RESUMEN

All living organisms deploy cell-autonomous defenses to combat infection. In plants and animals, large supramolecular complexes often activate immune proteins for protection. In this work, we resolved the native structure of a massive host-defense complex that polymerizes 30,000 guanylate-binding proteins (GBPs) over the surface of gram-negative bacteria inside human cells. Construction of this giant nanomachine took several minutes and remained stable for hours, required guanosine triphosphate hydrolysis, and recruited four GBPs plus caspase-4 and Gasdermin D as a cytokine and cell death immune signaling platform. Cryo-electron tomography suggests that GBP1 can adopt an extended conformation for bacterial membrane insertion to establish this platform, triggering lipopolysaccharide release that activated coassembled caspase-4. Our "open conformer" model provides a dynamic view into how the human GBP1 defense complex mobilizes innate immunity to infection.


Asunto(s)
Bacterias , Infecciones Bacterianas , Membrana Celular , Proteínas de Unión al GTP , Reconocimiento de Inmunidad Innata , Humanos , Citocinas/química , Tomografía con Microscopio Electrónico , Proteínas de Unión al GTP/química , Guanosina Trifosfato/química , Hidrólisis , Inmunidad Celular , Microscopía por Crioelectrón , Gasderminas/química , Proteínas de Unión a Fosfato/química , Conformación Proteica , Membrana Celular/química , Membrana Celular/inmunología , Caspasas Iniciadoras/química , Infecciones Bacterianas/inmunología , Bacterias/inmunología
17.
Sci Signal ; 15(764): eadf0778, 2022 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-36512642

RESUMEN

Interferons (IFNs) activate cell-autonomous immunity to combat infection and control inflammation. In this issue of Science Signaling, Boccuni et al. reveal how macrophages incorporate stress signals through the p38 MAPK pathway to enhance IFN-induced responses against intracellular pathogens.


Asunto(s)
Interferones , Transducción de Señal , Proteínas Quinasas p38 Activadas por Mitógenos , Macrófagos , Lógica
18.
Science ; 373(6552)2021 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-34437126

RESUMEN

Activation of cell-autonomous defense by the immune cytokine interferon-γ (IFN-γ) is critical to the control of life-threatening infections in humans. IFN-γ induces the expression of hundreds of host proteins in all nucleated cells and tissues, yet many of these proteins remain uncharacterized. We screened 19,050 human genes by CRISPR-Cas9 mutagenesis and identified IFN-γ-induced apolipoprotein L3 (APOL3) as a potent bactericidal agent protecting multiple non-immune barrier cell types against infection. Canonical apolipoproteins typically solubilize mammalian lipids for extracellular transport; APOL3 instead targeted cytosol-invasive bacteria to dissolve their anionic membranes into human-bacterial lipoprotein nanodiscs detected by native mass spectrometry and visualized by single-particle cryo-electron microscopy. Thus, humans have harnessed the detergent-like properties of extracellular apolipoproteins to fashion an intracellular lysin, thereby endowing resident nonimmune cells with a mechanism to achieve sterilizing immunity.


Asunto(s)
Apolipoproteínas L/metabolismo , Membrana Celular/metabolismo , Citosol/microbiología , Bacterias Gramnegativas/fisiología , Interferón gamma/inmunología , Apolipoproteínas L/química , Apolipoproteínas L/genética , Membrana Externa Bacteriana/metabolismo , Bacteriólisis , Sistemas CRISPR-Cas , Membrana Celular/química , Membrana Celular/ultraestructura , Permeabilidad de la Membrana Celular , Células Cultivadas , Detergentes/metabolismo , Proteínas de Unión al GTP/metabolismo , Edición Génica , Bacterias Gramnegativas/inmunología , Bacterias Gramnegativas/patogenicidad , Bacterias Gramnegativas/ultraestructura , Humanos , Inmunidad Innata , Lipoproteínas/química , Viabilidad Microbiana , Antígenos O/metabolismo , Dominios Proteicos , Salmonella typhimurium/inmunología , Salmonella typhimurium/patogenicidad , Salmonella typhimurium/fisiología , Salmonella typhimurium/ultraestructura , Solubilidad
19.
J Exp Med ; 216(3): 482-500, 2019 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-30755454

RESUMEN

Guanylate-binding proteins (GBPs) have recently emerged as central orchestrators of immunity to infection, inflammation, and neoplastic diseases. Within numerous host cell types, these IFN-induced GTPases assemble into large nanomachines that execute distinct host defense activities against a wide variety of microbial pathogens. In addition, GBPs customize inflammasome responses to bacterial infection and sepsis, where they act as critical rheostats to amplify innate immunity and regulate tissue damage. Similar functions are becoming evident for metabolic inflammatory syndromes and cancer, further underscoring the importance of GBPs within infectious as well as altered homeostatic settings. A better understanding of the basic biology of these IFN-induced GTPases could thus benefit clinical approaches to a wide spectrum of important human diseases.


Asunto(s)
Proteínas de Unión al GTP/química , Proteínas de Unión al GTP/metabolismo , Interacciones Huésped-Parásitos/inmunología , Interacciones Huésped-Patógeno/inmunología , Interferones/metabolismo , Animales , Colitis/inmunología , Colitis/metabolismo , Proteínas de Unión al GTP/inmunología , Humanos , Inflamasomas/fisiología , Inflamación/inmunología , Inflamación/metabolismo , Vertebrados
20.
Curr Opin Immunol ; 60: 71-80, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31176142

RESUMEN

Inside host cells, guanylate binding proteins (GBPs) rapidly assemble into large antimicrobial defense complexes that combat a wide variety of bacterial pathogens. These massive nanomachines often completely coat targeted microbes where they act as recruitment platforms for downstream effectors capable of direct bactericidal activity. GBP-containing platforms also serve as sensory hubs to activate inflammasome-driven responses in the mammalian cytosol while in plants like Arabidopsis, GBP orthologues may facilitate intranuclear signaling for immunity against invasive phytopathogens. Together, this group of immune GTPases serve as a major defensive repertoire to protect the host cell interior from bacterial colonization across plant and animal kingdoms.


Asunto(s)
Células Eucariotas/inmunología , Células Eucariotas/metabolismo , Proteínas de Unión al GTP/metabolismo , Interacciones Huésped-Patógeno/inmunología , Inmunidad , Interferones/metabolismo , Animales , Infecciones Bacterianas/genética , Infecciones Bacterianas/inmunología , Infecciones Bacterianas/metabolismo , Infecciones Bacterianas/microbiología , Evolución Biológica , Células Eucariotas/microbiología , Proteínas de Unión al GTP/química , Proteínas de Unión al GTP/genética , Regulación de la Expresión Génica , Regulación Enzimológica de la Expresión Génica , Humanos , Enfermedades de las Plantas , Fenómenos Fisiológicos de las Plantas , Plantas/genética , Plantas/inmunología , Plantas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA