Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.400
Filtrar
Más filtros

Intervalo de año de publicación
1.
Nat Immunol ; 25(9): 1754-1763, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39191945

RESUMEN

T cell antigen receptor (TCR) recognition followed by clonal expansion is a fundamental feature of adaptive immune responses. Here, we present a mass cytometric (CyTOF) approach to track T cell responses by combining antibodies for specific TCR Vα and Vß chains with antibodies against T cell activation and differentiation proteins in mice. This strategy identifies expansions of CD8+ and CD4+ T cells expressing specific Vß and Vα chains with varying differentiation states in response to Listeria monocytogenes, tumors and respiratory influenza infection. Expanded T cell populations expressing Vß chains could be directly linked to the recognition of specific antigens from Listeria, tumor cells or influenza. In the setting of influenza infection, we found that common therapeutic approaches of intramuscular vaccination or convalescent serum transfer altered the TCR diversity and differentiation state of responding T cells. Thus, we present a method to monitor broad changes in TCR use paired with T cell phenotyping during adaptive immune responses.


Asunto(s)
Linfocitos T CD8-positivos , Diferenciación Celular , Citometría de Flujo , Listeria monocytogenes , Listeriosis , Animales , Diferenciación Celular/inmunología , Ratones , Listeria monocytogenes/inmunología , Linfocitos T CD8-positivos/inmunología , Listeriosis/inmunología , Citometría de Flujo/métodos , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Ratones Endogámicos C57BL , Infecciones por Orthomyxoviridae/inmunología , Activación de Linfocitos/inmunología , Linfocitos T CD4-Positivos/inmunología , Inmunidad Adaptativa , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores de Antígenos de Linfocitos T/inmunología
2.
Nat Immunol ; 22(11): 1382-1390, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34663978

RESUMEN

Intergenerational inheritance of immune traits linked to epigenetic modifications has been demonstrated in plants and invertebrates. Here we provide evidence for transmission of trained immunity across generations to murine progeny that survived a sublethal systemic infection with Candida albicans or a zymosan challenge. The progeny of trained mice exhibited cellular, developmental, transcriptional and epigenetic changes associated with the bone marrow-resident myeloid effector and progenitor cell compartment. Moreover, the progeny of trained mice showed enhanced responsiveness to endotoxin challenge, alongside improved protection against systemic heterologous Escherichia coli and Listeria monocytogenes infections. Sperm DNA of parental male mice intravenously infected with the fungus C. albicans showed DNA methylation differences linked to immune gene loci. These results provide evidence for inheritance of trained immunity in mammals, enhancing protection against infections.


Asunto(s)
Candida albicans/inmunología , Candidiasis/inmunología , Infecciones por Escherichia coli/inmunología , Escherichia coli/inmunología , Herencia , Inmunidad Innata/genética , Listeria monocytogenes/inmunología , Listeriosis/inmunología , Células Mieloides/inmunología , Animales , Candida albicans/patogenicidad , Candidiasis/genética , Candidiasis/metabolismo , Candidiasis/microbiología , Células Cultivadas , Metilación de ADN , Modelos Animales de Enfermedad , Epigénesis Genética , Escherichia coli/patogenicidad , Infecciones por Escherichia coli/genética , Infecciones por Escherichia coli/metabolismo , Infecciones por Escherichia coli/microbiología , Interacciones Huésped-Patógeno , Listeria monocytogenes/patogenicidad , Listeriosis/genética , Listeriosis/metabolismo , Listeriosis/microbiología , Masculino , Ratones Transgénicos , Células Mieloides/metabolismo , Células Mieloides/microbiología , Espermatozoides/inmunología , Espermatozoides/metabolismo , Transcripción Genética
3.
Immunity ; 57(8): 1812-1827.e7, 2024 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-38955184

RESUMEN

An important property of the host innate immune response during microbial infection is its ability to control the expression of antimicrobial effector proteins, but how this occurs post-transcriptionally is not well defined. Here, we describe a critical antibacterial role for the classic antiviral gene 2'-5'-oligoadenylate synthetase 1 (OAS1). Human OAS1 and its mouse ortholog, Oas1b, are induced by interferon-γ and protect against cytosolic bacterial pathogens such as Francisella novicida and Listeria monocytogenes in vitro and in vivo. Proteomic and transcriptomic analysis showed reduced IRF1 protein expression in OAS1-deficient cells. Mechanistically, OAS1 binds and localizes IRF1 mRNA to the rough endoplasmic reticulum (ER)-Golgi endomembranes, licensing effective translation of IRF1 mRNA without affecting its transcription or decay. OAS1-dependent translation of IRF1 leads to the enhanced expression of antibacterial effectors, such as GBPs, which restrict intracellular bacteria. These findings uncover a noncanonical function of OAS1 in antibacterial innate immunity.


Asunto(s)
2',5'-Oligoadenilato Sintetasa , Inmunidad Innata , Factor 1 Regulador del Interferón , 2',5'-Oligoadenilato Sintetasa/metabolismo , 2',5'-Oligoadenilato Sintetasa/genética , Factor 1 Regulador del Interferón/metabolismo , Factor 1 Regulador del Interferón/genética , Animales , Humanos , Ratones , Biosíntesis de Proteínas/inmunología , Listeria monocytogenes/inmunología , Ratones Noqueados , Ratones Endogámicos C57BL , Listeriosis/inmunología , Interferón gamma/metabolismo , Interferón gamma/inmunología
4.
Cell ; 175(6): 1651-1664.e14, 2018 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-30392956

RESUMEN

The activator and composition of the NLRP6 inflammasome remain poorly understood. We find that lipoteichoic acid (LTA), a molecule produced by Gram-positive bacteria, binds and activates NLRP6. In response to cytosolic LTA or infection with Listeria monocytogenes, NLRP6 recruited caspase-11 and caspase-1 via the adaptor ASC. NLRP6 activation by LTA induced processing of caspase-11, which promoted caspase-1 activation and interleukin-1ß (IL-1ß)/IL-18 maturation in macrophages. Nlrp6-/- and Casp11-/- mice were less susceptible to L. monocytogenes infection, which was associated with reduced pathogen loads and impaired IL-18 production. Administration of IL-18 to Nlrp6-/- or Casp11-/- mice restored the susceptibility of mutant mice to L. monocytogenes infection. These results reveal a previously unrecognized innate immunity pathway triggered by cytosolic LTA that is sensed by NLRP6 and exacerbates systemic Gram-positive pathogen infection via the production of IL-18.


Asunto(s)
Inmunidad Innata , Inflamasomas/inmunología , Lipopolisacáridos/inmunología , Listeria monocytogenes/inmunología , Listeriosis/inmunología , Receptores de Superficie Celular/inmunología , Ácidos Teicoicos/inmunología , Animales , Caspasa 1/genética , Caspasa 1/inmunología , Caspasas/genética , Caspasas/inmunología , Caspasas Iniciadoras , Inflamasomas/genética , Interleucina-18/genética , Interleucina-18/inmunología , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Listeriosis/genética , Listeriosis/patología , Ratones , Ratones Noqueados , Receptores de Superficie Celular/genética
5.
Cell ; 168(1-2): 150-158.e10, 2017 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-28041849

RESUMEN

Bacterial CRISPR-Cas systems utilize sequence-specific RNA-guided nucleases to defend against bacteriophage infection. As a countermeasure, numerous phages are known that produce proteins to block the function of class 1 CRISPR-Cas systems. However, currently no proteins are known to inhibit the widely used class 2 CRISPR-Cas9 system. To find these inhibitors, we searched cas9-containing bacterial genomes for the co-existence of a CRISPR spacer and its target, a potential indicator for CRISPR inhibition. This analysis led to the discovery of four unique type II-A CRISPR-Cas9 inhibitor proteins encoded by Listeria monocytogenes prophages. More than half of L. monocytogenes strains with cas9 contain at least one prophage-encoded inhibitor, suggesting widespread CRISPR-Cas9 inactivation. Two of these inhibitors also blocked the widely used Streptococcus pyogenes Cas9 when assayed in Escherichia coli and human cells. These natural Cas9-specific "anti-CRISPRs" present tools that can be used to regulate the genome engineering activities of CRISPR-Cas9.


Asunto(s)
Bacteriófagos/metabolismo , Sistemas CRISPR-Cas , Endonucleasas/antagonistas & inhibidores , Ingeniería Genética , Listeria monocytogenes/enzimología , Proteínas Bacterianas/antagonistas & inhibidores , Proteína 9 Asociada a CRISPR , Escherichia coli , Células HEK293 , Humanos , Listeria monocytogenes/inmunología , Listeria monocytogenes/virología , Profagos
6.
Immunity ; 54(4): 829-844.e5, 2021 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-33705706

RESUMEN

Memory T cells are thought to rely on oxidative phosphorylation and short-lived effector T cells on glycolysis. Here, we investigated how T cells arrive at these states during an immune response. To understand the metabolic state of rare, early-activated T cells, we adapted mass cytometry to quantify metabolic regulators at single-cell resolution in parallel with cell signaling, proliferation, and effector function. We interrogated CD8+ T cell activation in vitro and in response to Listeria monocytogenes infection in vivo. This approach revealed a distinct metabolic state in early-activated T cells characterized by maximal expression of glycolytic and oxidative metabolic proteins. Cells in this transient state were most abundant 5 days post-infection before rapidly decreasing metabolic protein expression. Analogous findings were observed in chimeric antigen receptor (CAR) T cells interrogated longitudinally in advanced lymphoma patients. Our study demonstrates the utility of single-cell metabolic analysis by mass cytometry to identify metabolic adaptations of immune cell populations in vivo and provides a resource for investigations of metabolic regulation of immune responses across a variety of applications.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Activación de Linfocitos/inmunología , Transducción de Señal/inmunología , Animales , Proliferación Celular/fisiología , Femenino , Glucólisis/inmunología , Memoria Inmunológica/inmunología , Listeria monocytogenes/inmunología , Listeriosis/inmunología , Listeriosis/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fosforilación Oxidativa , Receptores Quiméricos de Antígenos/inmunología , Análisis de la Célula Individual/métodos
7.
Nature ; 631(8021): 635-644, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38961291

RESUMEN

Innate immune pattern recognition receptors, such as the Toll-like receptors (TLRs), are key mediators of the immune response to infection and central to our understanding of health and disease1. After microbial detection, these receptors activate inflammatory signal transduction pathways that involve IκB kinases, mitogen-activated protein kinases, ubiquitin ligases and other adaptor proteins. The mechanisms that connect the proteins in the TLR pathways are poorly defined. To delineate TLR pathway activities, we engineered macrophages to enable microscopy and proteomic analysis of the endogenous myddosome constituent MyD88. We found that myddosomes form transient contacts with activated TLRs and that TLR-free myddosomes are dynamic in size, number and composition over the course of 24 h. Analysis using super-resolution microscopy revealed that, within most myddosomes, MyD88 forms barrel-like structures that function as scaffolds for effector protein recruitment. Proteomic analysis demonstrated that myddosomes contain proteins that act at all stages and regulate all effector responses of the TLR pathways, and genetic analysis defined the epistatic relationship between these effector modules. Myddosome assembly was evident in cells infected with Listeria monocytogenes, but these bacteria evaded myddosome assembly and TLR signalling during cell-to-cell spread. On the basis of these findings, we propose that the entire TLR signalling pathway is executed from within the myddosome.


Asunto(s)
Macrófagos , Transducción de Señal , Receptores Toll-Like , Animales , Humanos , Ratones , Listeria monocytogenes/inmunología , Listeriosis/inmunología , Listeriosis/microbiología , Listeriosis/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/microbiología , Factor 88 de Diferenciación Mieloide/metabolismo , Proteómica , Receptores Toll-Like/metabolismo , Microscopía , Inmunidad Innata
8.
Mol Cell ; 82(3): 527-541.e7, 2022 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-35016033

RESUMEN

Citrulline can be converted into argininosuccinate by argininosuccinate synthetase (ASS1) in the urea cycle and the citrulline-nitric oxide cycle. However, the regulation and biological function of citrulline metabolism remain obscure in the immune system. Unexpectedly, we found that macrophage citrulline declines rapidly after interferon gamma (IFN-γ) and/or lipopolysaccharide (LPS) stimulation, which is required for efficient proinflammatory signaling activation. Mechanistically, IFN-γ and/or LPS stimulation promotes signal transducers and activators of transcription 1 (STAT1)-mediated ASS1 transcription and Janus kinase2 (JAK2)-mediated phosphorylation of ASS1 at tyrosine 87, thereby leading to citrulline depletion. Reciprocally, increased citrulline directly binds to JAK2 and inhibits JAK2-STAT1 signaling. Blockage of ASS1-mediated citrulline depletion suppresses the host defense against bacterial infection in vivo. We therefore define a central role for ASS1 in controlling inflammatory macrophage activation and antibacterial defense through depletion of cellular citrulline and, further, identify citrulline as an innate immune-signaling metabolite that engages a metabolic checkpoint for proinflammatory responses.


Asunto(s)
Argininosuccinato Sintasa/metabolismo , Citrulina/metabolismo , Inmunidad Innata , Inflamación/enzimología , Listeriosis/enzimología , Activación de Macrófagos , Macrófagos/enzimología , Animales , Argininosuccinato Sintasa/genética , Modelos Animales de Enfermedad , Células HEK293 , Humanos , Inflamación/genética , Inflamación/inmunología , Mediadores de Inflamación/metabolismo , Janus Quinasa 2/genética , Janus Quinasa 2/metabolismo , Listeria monocytogenes/inmunología , Listeriosis/genética , Listeriosis/inmunología , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación , Células RAW 264.7 , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/metabolismo , Transducción de Señal
9.
Immunity ; 52(4): 591-605.e6, 2020 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-32294405

RESUMEN

Human toll-like receptor 8 (TLR8) activation induces a potent T helper-1 (Th1) cell response critical for defense against intracellular pathogens, including protozoa. The receptor harbors two distinct binding sites, uridine and di- and/or trinucleotides, but the RNases upstream of TLR8 remain poorly characterized. We identified two endolysosomal endoribonucleases, RNase T2 and RNase 2, that act synergistically to release uridine from oligoribonucleotides. RNase T2 cleaves preferentially before, and RNase 2 after, uridines. Live bacteria, P. falciparum-infected red blood cells, purified pathogen RNA, and synthetic oligoribonucleotides all required RNase 2 and T2 processing to activate TLR8. Uridine supplementation restored RNA recognition in RNASE2-/- or RNASET2-/- but not RNASE2-/-RNASET2-/- cells. Primary immune cells from RNase T2-hypomorphic patients lacked a response to bacterial RNA but responded robustly to small-molecule TLR8 ligands. Our data identify an essential function of RNase T2 and RNase 2 upstream of TLR8 and provide insight into TLR8 activation.


Asunto(s)
Endorribonucleasas/metabolismo , Monocitos/inmunología , Neutrófilos/inmunología , ARN Bacteriano/metabolismo , ARN Protozoario/metabolismo , Receptor Toll-Like 8/metabolismo , Sistemas CRISPR-Cas , Línea Celular , Endorribonucleasas/inmunología , Eritrocitos/inmunología , Eritrocitos/parasitología , Escherichia coli/química , Escherichia coli/inmunología , Edición Génica/métodos , Humanos , Listeria monocytogenes/química , Listeria monocytogenes/inmunología , Monocitos/microbiología , Monocitos/parasitología , Neutrófilos/microbiología , Neutrófilos/parasitología , Plasmodium falciparum/química , Plasmodium falciparum/inmunología , Cultivo Primario de Células , Estabilidad del ARN , ARN Bacteriano/inmunología , ARN Protozoario/inmunología , Serratia marcescens/química , Serratia marcescens/inmunología , Staphylococcus aureus/química , Staphylococcus aureus/inmunología , Streptococcus/química , Streptococcus/inmunología , Células THP-1 , Receptor Toll-Like 8/inmunología
10.
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
11.
Nat Immunol ; 17(10): 1167-75, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27548433

RESUMEN

CD8α(+) dendritic cells (DCs) are specialized at cross-presenting extracellular antigens on major histocompatibility complex (MHC) class I molecules to initiate cytotoxic T lymphocyte (CTL) responses; however, details of the mechanisms that regulate cross-presentation remain unknown. We found lower expression of the lectin family member Siglec-G in CD8α(+) DCs, and Siglec-G deficient (Siglecg(-/-)) mice generated more antigen-specific CTLs to inhibit intracellular bacterial infection and tumor growth. MHC class I-peptide complexes were more abundant on Siglecg(-/-) CD8α(+) DCs than on Siglecg(+/+) CD8α(+) DCs. Mechanistically, phagosome-expressed Siglec-G recruited the phosphatase SHP-1, which dephosphorylated the NADPH oxidase component p47(phox) and inhibited the activation of NOX2 on phagosomes. This resulted in excessive hydrolysis of exogenous antigens, which led to diminished formation of MHC class I-peptide complexes for cross-presentation. Therefore, Siglec-G inhibited DC cross-presentation by impairing such complex formation, and our results add insight into the regulation of cross-presentation in adaptive immunity.


Asunto(s)
Reactividad Cruzada , Células Dendríticas/inmunología , Lectinas/metabolismo , Listeria monocytogenes/inmunología , Listeriosis/inmunología , Neoplasias Experimentales/inmunología , Receptores de Antígenos de Linfocitos B/metabolismo , Linfocitos T Citotóxicos/inmunología , Animales , Antígenos/metabolismo , Antígenos CD8/metabolismo , Antígenos de Histocompatibilidad Clase I/metabolismo , Lectinas/genética , Activación de Linfocitos , Melanoma Experimental , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , NADPH Oxidasas/metabolismo , Fragmentos de Péptidos/metabolismo , Fagocitosis/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 6/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo , Receptores de Antígenos de Linfocitos B/genética , Lectinas Similares a la Inmunoglobulina de Unión a Ácido Siálico , Transducción de Señal , Carga Tumoral/genética
12.
Nature ; 606(7915): 769-775, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35676476

RESUMEN

Adaptive immune components are thought to exert non-overlapping roles in antimicrobial host defence, with antibodies targeting pathogens in the extracellular environment and T cells eliminating infection inside cells1,2. Reliance on antibodies for vertically transferred immunity from mothers to babies may explain neonatal susceptibility to intracellular infections3,4. Here we show that pregnancy-induced post-translational antibody modification enables protection against the prototypical intracellular pathogen Listeria monocytogenes. Infection susceptibility was reversed in neonatal mice born to preconceptually primed mothers possessing L. monocytogenes-specific IgG or after passive transfer of antibodies from primed pregnant, but not virgin, mice. Although maternal B cells were essential for producing IgGs that mediate vertically transferred protection, they were dispensable for antibody acquisition of protective function, which instead required sialic acid acetyl esterase5 to deacetylate terminal sialic acid residues on IgG variable-region N-linked glycans. Deacetylated L. monocytogenes-specific IgG protected neonates through the sialic acid receptor CD226,7, which suppressed IL-10 production by B cells leading to antibody-mediated protection. Consideration of the maternal-fetal dyad as a joined immunological unit reveals protective roles for antibodies against intracellular infection and fine-tuned adaptations to enhance host defence during pregnancy and early life.


Asunto(s)
Inmunidad Materno-Adquirida , Inmunoglobulina G , Espacio Intracelular , Listeria monocytogenes , Madres , Embarazo , Acetilesterasa , Animales , Animales Recién Nacidos , Linfocitos B , Femenino , Inmunidad Materno-Adquirida/inmunología , Inmunoglobulina G/inmunología , Interleucina-10/biosíntesis , Espacio Intracelular/inmunología , Espacio Intracelular/microbiología , Listeria monocytogenes/inmunología , Listeriosis/inmunología , Listeriosis/prevención & control , Ratones , Ácido N-Acetilneuramínico/metabolismo , Embarazo/inmunología , Lectina 2 Similar a Ig de Unión al Ácido Siálico , Linfocitos T
13.
Nature ; 606(7916): 960-967, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35705808

RESUMEN

Among the caspases that cause regulated cell death, a unique function for caspase-7 has remained elusive. Caspase-3 performs apoptosis, whereas caspase-7 is typically considered an inefficient back-up. Caspase-1 activates gasdermin D pores to lyse the cell; however, caspase-1 also activates caspase-7 for unknown reasons1. Caspases can also trigger cell-type-specific death responses; for example, caspase-1 causes the extrusion of intestinal epithelial cell (IECs) in response to infection with Salmonella enterica subsp. enterica serovar Typhimurium (S. Typhimurium)2,3. Here we show in both organoids and mice that caspase-7-deficient IECs do not complete extrusion. Mechanistically, caspase-7 counteracts gasdermin D pores and preserves cell integrity by cleaving and activating acid sphingomyelinase (ASM), which thereby generates copious amounts of ceramide to enable enhanced membrane repair. This provides time to complete the process of IEC extrusion. In parallel, we also show that caspase-7 and ASM cleavage are required to clear Chromobacterium violaceum and Listeria monocytogenes after perforin-pore-mediated attack by natural killer cells or cytotoxic T lymphocytes, which normally causes apoptosis in infected hepatocytes. Therefore, caspase-7 is not a conventional executioner but instead is a death facilitator that delays pore-driven lysis so that more-specialized processes, such as extrusion or apoptosis, can be completed before cell death. Cells must put their affairs in order before they die.


Asunto(s)
Caspasa 7 , Perforina , Proteínas de Unión a Fosfato , Proteínas Citotóxicas Formadoras de Poros , Esfingomielina Fosfodiesterasa , Animales , Apoptosis , Caspasa 7/metabolismo , Chromobacterium/inmunología , Células Epiteliales/citología , Intestinos/citología , Células Asesinas Naturales/inmunología , Listeria monocytogenes/inmunología , Ratones , Organoides , Perforina/metabolismo , Proteínas de Unión a Fosfato/metabolismo , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Esfingomielina Fosfodiesterasa/metabolismo , Linfocitos T Citotóxicos/inmunología
14.
Nat Immunol ; 16(4): 366-75, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25729924

RESUMEN

Neutrophils express Toll-like receptors (TLRs) for the recognition of conserved bacterial elements to initiate antimicrobial responses. However, whether other cytosolic DNA sensors are expressed by neutrophils remains elusive. Here we found constitutive expression of the transcription factor Sox2 in the cytoplasm of mouse and human neutrophils. Neutrophil-specific Sox2 deficiency exacerbated bacterial infection. Sox2 directly recognized microbial DNA through its high-mobility-group (HMG) domain. Upon challenge with bacterial DNA, Sox2 dimerization was needed to activate a complex of the kinase TAK1 and its binding partner TAB2, which led to activation of the transcription factors NF-κB and AP-1 in neutrophils. Deficiency in TAK1 or TAB2 impaired Sox2-mediated antibacterial immunity. Overall, we reveal a previously unrecognized role for Sox2 as a cytosolic sequence-specific DNA sensor in neutrophils, which might provide potential therapeutic strategies for the treatment of infectious diseases.


Asunto(s)
ADN Bacteriano/inmunología , Inmunidad Innata , Listeriosis/inmunología , Neutrófilos/inmunología , Factores de Transcripción SOXB1/inmunología , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/inmunología , Animales , Citoplasma/inmunología , Citoplasma/microbiología , Regulación de la Expresión Génica , Humanos , Listeria monocytogenes/inmunología , Listeriosis/genética , Listeriosis/microbiología , Listeriosis/mortalidad , Quinasas Quinasa Quinasa PAM/deficiencia , Quinasas Quinasa Quinasa PAM/genética , Quinasas Quinasa Quinasa PAM/inmunología , Ratones , FN-kappa B/genética , FN-kappa B/inmunología , Neutrófilos/microbiología , Multimerización de Proteína , Factores de Transcripción SOXB1/genética , Transducción de Señal , Análisis de Supervivencia , Receptores Toll-Like/genética , Receptores Toll-Like/inmunología , Factor de Transcripción AP-1/genética , Factor de Transcripción AP-1/inmunología
15.
Immunity ; 48(4): 716-729.e8, 2018 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-29625895

RESUMEN

Protective immunity against pathogens depends on the efficient generation of functionally diverse effector and memory T lymphocytes. However, whether plasticity during effector-to-memory CD8+ T cell differentiation affects memory lineage specification and functional versatility remains unclear. Using genetic fate mapping analysis of highly cytotoxic KLRG1+ effector CD8+ T cells, we demonstrated that KLRG1+ cells receiving intermediate amounts of activating and inflammatory signals downregulated KLRG1 during the contraction phase in a Bach2-dependent manner and differentiated into all memory T cell linages, including CX3CR1int peripheral memory cells and tissue-resident memory cells. "ExKLRG1" memory cells retained high cytotoxic and proliferative capacity distinct from other populations, which contributed to effective anti-influenza and anti-tumor immunity. Our work demonstrates that developmental plasticity of KLRG1+ effector CD8+ T cells is important in promoting functionally versatile memory cells and long-term protective immunity.


Asunto(s)
Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Memoria Inmunológica/inmunología , Activación de Linfocitos/inmunología , Receptores Inmunológicos/metabolismo , Animales , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Diferenciación Celular/inmunología , Línea Celular Tumoral , Linaje de la Célula/inmunología , Virus de la Influenza A/inmunología , Subunidad p35 de la Interleucina-12/inmunología , Lectinas Tipo C , Listeria monocytogenes/inmunología , Melanoma Experimental , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores Inmunológicos/genética , Virus de la Estomatitis Vesicular Indiana/inmunología
16.
Nat Immunol ; 15(6): 562-70, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24777531

RESUMEN

Deubiquitinases (DUBs) are a new class of drug targets, although the physiological function of only few DUBs has been characterized. Here we identified the DUB USP15 as a crucial negative regulator of T cell activation. USP15 stabilized the E3 ubiquitin ligase MDM2, which in turn negatively regulated T cell activation by targeting the degradation of the transcription factor NFATc2. USP15 deficiency promoted T cell activation in vitro and enhanced T cell responses to bacterial infection and tumor challenge in vivo. USP15 also stabilized MDM2 in cancer cells and regulated p53 function and cancer-cell survival. Our results suggest that inhibition of USP15 may both induce tumor cell apoptosis and boost antitumor T cell responses.


Asunto(s)
Factores de Transcripción NFATC/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/inmunología , Células TH1/inmunología , Proteasas Ubiquitina-Específicas/inmunología , Traslado Adoptivo , Animales , Apoptosis/inmunología , Diferenciación Celular/inmunología , Línea Celular Tumoral , Supervivencia Celular , Células HCT116 , Humanos , Leupeptinas/farmacología , Listeria monocytogenes/inmunología , Listeriosis/inmunología , Activación de Linfocitos/inmunología , Melanoma Experimental/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Proto-Oncogénicas c-mdm2/genética , Escape del Tumor , Proteína p53 Supresora de Tumor/inmunología , Proteasas Ubiquitina-Específicas/genética , Ubiquitinación/genética , Ubiquitinación/inmunología
17.
Nat Immunol ; 15(4): 365-372, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24584088

RESUMEN

T lymphocytes responding to microbial infection give rise to effector cells that mediate acute host defense and memory cells that provide long-lived immunity, but the fundamental question of when and how these cells arise remains unresolved. Here we combined single-cell gene-expression analyses with 'machine-learning' approaches to trace the transcriptional 'roadmap' of individual CD8(+) T lymphocytes throughout the course of an immune response in vivo. Gene-expression signatures predictive of eventual fates could be discerned as early as the first T lymphocyte division and may have been influenced by asymmetric partitioning of the receptor for interleukin 2 (IL-2Rα) during mitosis. Our findings emphasize the importance of single-cell analyses in understanding fate determination and provide new insights into the specification of divergent lymphocyte fates early during an immune response to microbial infection.


Asunto(s)
Inmunidad Adaptativa , Linfocitos T CD8-positivos/inmunología , Perfilación de la Expresión Génica/métodos , Infecciones/inmunología , Infecciones/microbiología , Receptores de Interleucina-2/metabolismo , Análisis de la Célula Individual/métodos , Subgrupos de Linfocitos T/inmunología , Traslado Adoptivo , Animales , Linfocitos T CD8-positivos/microbiología , Linfocitos T CD8-positivos/virología , Diferenciación Celular/genética , Linaje de la Célula/genética , Simulación por Computador , Listeria monocytogenes/genética , Listeria monocytogenes/inmunología , Activación de Linfocitos/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mitosis/genética , Mitosis/inmunología , Ovalbúmina/genética , Ovalbúmina/inmunología , Receptores de Interleucina-2/genética , Subgrupos de Linfocitos T/microbiología , Subgrupos de Linfocitos T/virología , Activación Transcripcional/inmunología
18.
Nat Immunol ; 15(11): 1046-54, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25263126

RESUMEN

Chaperone-mediated autophagy (CMA) targets soluble proteins for lysosomal degradation. Here we found that CMA was activated in T cells in response to engagement of the T cell antigen receptor (TCR), which induced expression of the CMA-related lysosomal receptor LAMP-2A. In activated T cells, CMA targeted the ubiquitin ligase Itch and the calcineurin inhibitor RCAN1 for degradation to maintain activation-induced responses. Consequently, deletion of the gene encoding LAMP-2A in T cells caused deficient in vivo responses to immunization or infection with Listeria monocytogenes. Impaired CMA activity also occurred in T cells with age, which negatively affected their function. Restoration of LAMP-2A in T cells from old mice resulted in enhancement of activation-induced responses. Our findings define a role for CMA in regulating T cell activation through the targeted degradation of negative regulators of T cell activation.


Asunto(s)
Autofagia/inmunología , Activación de Linfocitos/inmunología , Proteína 2 de la Membrana Asociada a los Lisosomas/inmunología , Chaperonas Moleculares/inmunología , Células TH1/inmunología , Envejecimiento/inmunología , Animales , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/biosíntesis , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/genética , Inhibidores de la Calcineurina/metabolismo , Proteínas de Unión al Calcio , Células Cultivadas , Oxidasas Duales , Femenino , Humanos , Inmunización , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Listeria monocytogenes/inmunología , Listeriosis/inmunología , Proteína 2 de la Membrana Asociada a los Lisosomas/biosíntesis , Proteína 2 de la Membrana Asociada a los Lisosomas/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Musculares/metabolismo , NADPH Oxidasas/genética , Estrés Oxidativo/inmunología , Interferencia de ARN , ARN Mensajero/biosíntesis , ARN Interferente Pequeño , Especies Reactivas de Oxígeno/metabolismo , Receptores de Antígenos de Linfocitos T/inmunología , Ubiquitina-Proteína Ligasas/metabolismo
19.
Immunity ; 47(2): 374-388.e6, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28813662

RESUMEN

The liver is positioned at the interface between two routes traversed by pathogens in disseminating infection. Whereas blood-borne pathogens are efficiently cleared in hepatic sinusoids by Kupffer cells (KCs), it is unknown how the liver prevents dissemination of peritoneal pathogens accessing its outer membrane. We report here that the hepatic capsule harbors a contiguous cellular network of liver-resident macrophages phenotypically distinct from KCs. These liver capsular macrophages (LCMs) were replenished in the steady state from blood monocytes, unlike KCs that are embryonically derived and self-renewing. LCM numbers increased after weaning in a microbiota-dependent process. LCMs sensed peritoneal bacteria and promoted neutrophil recruitment to the capsule, and their specific ablation resulted in decreased neutrophil recruitment and increased intrahepatic bacterial burden. Thus, the liver contains two separate and non-overlapping niches occupied by distinct resident macrophage populations mediating immunosurveillance at these two pathogen entry points to the liver.


Asunto(s)
Macrófagos del Hígado/fisiología , Listeria monocytogenes/inmunología , Listeriosis/inmunología , Hígado/inmunología , Macrófagos/inmunología , Neutrófilos/inmunología , Peritoneo/microbiología , Animales , Comunicación Celular , Autorrenovación de las Células , Interacciones Huésped-Patógeno , Humanos , Inmunidad Innata , Macrófagos del Hígado/microbiología , Hígado/microbiología , Hígado/patología , Macrófagos/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/inmunología , Infiltración Neutrófila , Peritoneo/patología
20.
J Immunol ; 213(5): 619-627, 2024 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-39037267

RESUMEN

Sepsis is a complex condition of inflammatory and immune dysregulation, triggered by severe infection. In survivors, chronic inflammation and immune dysregulation linger, facilitating the emergence of infections. CD8 dysfunction contributes to immunosuppression in sepsis survivors. We devised an animal model that enabled us to identify and analyze CD8-intrinsic defects induced by sepsis. We adoptively transferred CD45.1 CD8 OT-I T cells into CD45.2 congenic mice and subjected them to cecal ligature and puncture, to induce abdominal sepsis. One month later, we isolated the transferred CD8 cells. Surface marker expression confirmed they had not been activated through the TCR. CD8 OT-I T cells isolated from septic (or sham-operated) mice were transferred to second recipients, which were challenged with OVA-expressing Listeria monocytogenes. We compared effector capacities between OT-I cells exposed to sepsis and control cells. Naive mice that received OT-I cells exposed to sepsis had higher bacterial burden and a shorter survival when challenged with OVA-expressing L. monocytogenes. OT-I cells isolated from septic mice produced less IFN-γ but had conserved activation, expansion potential, and cytotoxic function. We observed lower transcript levels of IFN-γ and of the long noncoding RNA Ifng-as1, a local regulator of the epigenetic landscape, in cells exposed to sepsis. Accordingly, local abundance of a histone modification characteristic of active promoter regions was reduced in sepsis-exposed CD8 T cells. Our results identify a mechanism through which inflammation in the context of sepsis affects CD8 T cell function intrinsically.


Asunto(s)
Linfocitos T CD8-positivos , Cromatina , Interferón gamma , Listeria monocytogenes , Sepsis , Animales , Ratones , Traslado Adoptivo , Linfocitos T CD8-positivos/inmunología , Cromatina/inmunología , Cromatina/metabolismo , Modelos Animales de Enfermedad , Interferón gamma/inmunología , Listeria monocytogenes/inmunología , Listeriosis/inmunología , Activación de Linfocitos/inmunología , Ratones Endogámicos C57BL , Sepsis/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA