Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros











Intervalo de año de publicación
1.
Sci Signal ; 17(827): eade3643, 2024 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-38470955

RESUMEN

Activation of the endoplasmic reticulum (ER)-resident adaptor protein STING, a component of a cytosolic DNA-sensing pathway, induces the transcription of genes encoding type I interferons (IFNs) and other proinflammatory factors. Because STING is activated at the Golgi apparatus, control of the localization and activation of STING is important in stimulating antiviral and antitumor immune responses. Through a genome-wide CRISPR interference screen, we found that STING activation required the Golgi-resident protein ACBD3, which promotes the generation of phosphatidylinositol 4-phosphate (PI4P) at the trans-Golgi network, as well as other PI4P-associated proteins. Appropriate localization and activation of STING at the Golgi apparatus required ACBD3 and the PI4P-generating kinase PI4KB. In contrast, STING activation was enhanced when the lipid-shuttling protein OSBP, which removes PI4P from the Golgi apparatus, was inhibited by the US Food and Drug Administration-approved antifungal itraconazole. The increase in the abundance of STING-activating phospholipids at the trans-Golgi network resulted in the increased production of IFN-ß and other cytokines in THP-1 cells. Furthermore, a mutant STING that could not bind to PI4P failed to traffic from the ER to the Golgi apparatus in response to a STING agonist, whereas forced relocalization of STING to PI4P-enriched areas elicited STING activation in the absence of stimulation with a STING agonist. Thus, PI4P is critical for STING activation, and manipulating PI4P abundance may therapeutically modulate STING-dependent immune responses.


Asunto(s)
Aparato de Golgi , Fosfolípidos , Fosfolípidos/metabolismo , Aparato de Golgi/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo
2.
JCI Insight ; 8(18)2023 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-37561594

RESUMEN

Mutation of the ATP2A2 gene encoding sarco-endoplasmic reticulum calcium ATPase 2 (SERCA2) was linked to Darier disease more than 2 decades ago; however, there remain no targeted therapies for this disorder causing recurrent skin blistering and infections. Since Atp2a2-knockout mice do not phenocopy its pathology, we established a human tissue model of Darier disease to elucidate its pathogenesis and identify potential therapies. Leveraging CRISPR/Cas9, we generated human keratinocytes lacking SERCA2, which replicated features of Darier disease, including weakened intercellular adhesion and defective differentiation in organotypic epidermis. To identify pathogenic drivers downstream of SERCA2 depletion, we performed RNA sequencing and proteomics analysis. SERCA2-deficient keratinocytes lacked desmosomal and cytoskeletal proteins required for epidermal integrity and exhibited excess MAPK signaling, which modulates keratinocyte adhesion and differentiation. Immunostaining patient biopsies substantiated these findings, with lesions showing keratin deficiency, cadherin mislocalization, and ERK hyperphosphorylation. Dampening ERK activity with MEK inhibitors rescued adhesive protein expression and restored keratinocyte sheet integrity despite SERCA2 depletion or chemical inhibition. In sum, coupling multiomic analysis with human organotypic epidermis as a preclinical model, we found that SERCA2 haploinsufficiency disrupts critical adhesive components in keratinocytes via ERK signaling and identified MEK inhibition as a treatment strategy for Darier disease.


Asunto(s)
Enfermedad de Darier , Ratones , Animales , Humanos , Enfermedad de Darier/tratamiento farmacológico , Enfermedad de Darier/genética , Enfermedad de Darier/metabolismo , Epidermis/metabolismo , Queratinocitos/metabolismo , Retículo Endoplásmico/metabolismo , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo
3.
JCI Insight ; 8(12)2023 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-37166992

RESUMEN

Cyclic GMP-AMP synthase (cGAS) is a DNA sensor and responsible for inducing an antitumor immune response. Recent studies reveal that cGAS is frequently inhibited in cancer, and therapeutic targets to promote antitumor cGAS function remain elusive. SRC is a proto-oncogene tyrosine kinase and is expressed at elevated levels in numerous cancers. Here, we demonstrate that SRC expression in primary and metastatic bladder cancer negatively correlates with innate immune gene expression and immune cell infiltration. We determine that SRC restricts cGAS signaling in human cell lines through SRC small molecule inhibitors, depletion, and overexpression. cGAS and SRC interact in cells and in vitro, while SRC directly inhibits cGAS enzymatic activity and DNA binding in a kinase-dependent manner. SRC phosphorylates cGAS, and inhibition of cGAS Y248 phosphorylation partially reduces SRC inhibition. Collectively, our study demonstrates that cGAS antitumor signaling is hindered by the proto-oncogene SRC and describes how cancer-associated proteins can regulate the innate immune system.


Asunto(s)
Neoplasias , Nucleotidiltransferasas , Humanos , Nucleotidiltransferasas/metabolismo , Inmunidad Innata , Neoplasias/genética , ADN/metabolismo , Proto-Oncogenes
4.
J Bacteriol ; 203(19): e0008021, 2021 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-34309402

RESUMEN

3'3'-Cyclic di-AMP (c-di-AMP) is an important nucleotide second messenger found throughout the bacterial domain of life. c-di-AMP is essential in many bacteria and regulates a diverse array of effector proteins controlling pathogenesis, cell wall homeostasis, osmoregulation, and central metabolism. Despite the ubiquity and importance of c-di-AMP, methods to detect this signaling molecule are limited, particularly at single-cell resolution. In this work, crystallization of the Listeria monocytogenes c-di-AMP effector protein Lmo0553 enabled structure-guided design of a Förster resonance energy transfer (FRET)-based biosensor, which we have named CDA5. CDA5 is a fully genetically encodable, specific, and reversible biosensor which allows the detection of c-di-AMP dynamics both in vitro and within live cells in a nondestructive manner. Our initial studies identified a distribution of c-di-AMP in Bacillus subtilis populations first grown in Luria broth and then resuspended in diluted Luria broth compatible with fluorescence analysis. Furthermore, we found that B. subtilis mutants lacking either a c-di-AMP phosphodiesterase and cyclase have higher and lower FRET responses, respectively. These findings provide novel insight into the c-di-AMP distribution within bacterial populations and establish CDA5 as a powerful platform for characterizing new aspects of c-di-AMP regulation. IMPORTANCE c-di-AMP is an important nucleotide second messenger for which detection methods are severely limited. In this work we engineered and implemented a c-di-AMP-specific FRET biosensor to remedy this dearth. We present this biosensor, CDA5, as a versatile tool to investigate previously intractable facets of c-di-AMP biology.


Asunto(s)
Técnicas Biosensibles , Fosfatos de Dinucleósidos/química , Transferencia Resonante de Energía de Fluorescencia , Nucleótidos/metabolismo , Bacillus subtilis/química , Bacillus subtilis/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Fosfatos de Dinucleósidos/metabolismo , Regulación Bacteriana de la Expresión Génica/fisiología , Listeria monocytogenes/metabolismo , Modelos Moleculares , Mutación , Conformación Proteica
5.
Elife ; 102021 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-33955352

RESUMEN

Pathogens encounter numerous antimicrobial responses during infection, including the reactive oxygen species (ROS) burst. ROS-mediated oxidation of host membrane poly-unsaturated fatty acids (PUFAs) generates the toxic alpha-beta carbonyl 4-hydroxy-2-nonenal (4-HNE). Although studied extensively in the context of sterile inflammation, research into 4-HNE's role during infection remains limited. Here, we found that 4-HNE is generated during bacterial infection, that it impacts growth and survival in a range of bacteria, and that the intracellular pathogen Listeria monocytogenes induces many genes in response to 4-HNE exposure. A component of the L. monocytogenes 4-HNE response is the expression of the genes lmo0103 and lmo0613, deemed rha1 and rha2 (reductase of host alkenals), respectively, which code for two NADPH-dependent oxidoreductases that convert 4-HNE to the product 4-hydroxynonanal (4-HNA). Loss of these genes had no impact on L. monocytogenes bacterial burdens during murine or tissue culture infection. However, heterologous expression of rha1/2 in Bacillus subtilis significantly increased bacterial resistance to 4-HNE in vitro and promoted bacterial survival following phagocytosis by murine macrophages in an ROS-dependent manner. Thus, Rha1 and Rha2 are not necessary for 4-HNE resistance in L. monocytogenes but are sufficient to confer resistance to an otherwise sensitive organism in vitro and in host cells. Our work demonstrates that 4-HNE is a previously unappreciated component of ROS-mediated toxicity encountered by bacteria within eukaryotic hosts.


Asunto(s)
Aldehídos/metabolismo , Interacciones Huésped-Patógeno , Listeria monocytogenes/genética , Listeria monocytogenes/patogenicidad , Animales , Bacillus subtilis/genética , Línea Celular , Femenino , Concentración de Iones de Hidrógeno , Macrófagos/microbiología , Ratones , Ratones Endogámicos C57BL , Fagocitosis
6.
Chembiochem ; 22(6): 1030-1041, 2021 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-33142009

RESUMEN

Cyclic dinucleotide signaling systems, which are found ubiquitously throughout nature, allow organisms to rapidly and dynamically sense and respond to alterations in their environments. In recent years, the second messenger, cyclic di-(3',5')-adenosine monophosphate (c-di-AMP), has been identified as an essential signaling molecule in a diverse array of bacterial genera. We and others have shown that defects in c-di-AMP homeostasis result in severe physiological defects and virulence attenuation in many bacterial species. Despite significant advancements in the field, there is still a major gap in the understanding of the environmental and cellular factors that influence c-di-AMP dynamics due to a lack of tools to sensitively and rapidly monitor changes in c-di-AMP levels. To address this limitation, we describe here the development of a luciferase-based coupled enzyme assay that leverages the cyclic nucleotide phosphodiesterase, CnpB, for the sensitive and high-throughput quantification of 3'3'-c-di-AMP. We also demonstrate the utility of this approach for the quantification of the cyclic oligonucleotide-based anti-phage signaling system (CBASS) effector, 3'3'-cGAMP. These findings establish CDA-Luc as a more affordable and sensitive alternative to conventional c-di-AMP detection tools with broad utility for the study of bacterial cyclic dinucleotide physiology.


Asunto(s)
3',5'-GMP Cíclico Fosfodiesterasas/metabolismo , Proteínas Bacterianas/metabolismo , Fosfatos de Dinucleósidos/análisis , Pruebas de Enzimas/métodos , Adenosina Monofosfato/metabolismo , Bacterias/metabolismo , Fosfatos de Dinucleósidos/química , Fosfatos de Dinucleósidos/metabolismo , Ensayos Analíticos de Alto Rendimiento , Hidrólisis , Luciferasas/metabolismo , Mediciones Luminiscentes/métodos , Mycobacterium tuberculosis/enzimología
7.
Nat Commun ; 11(1): 3533, 2020 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-32669552

RESUMEN

Cyclic dinucleotides (CDNs) are second messengers conserved across all three domains of life. Within eukaryotes they mediate protective roles in innate immunity against malignant, viral, and bacterial disease, and exert pathological effects in autoimmune disorders. Despite their ubiquitous role in diverse biological contexts, CDN detection methods are limited. Here, using structure guided design of the murine STING CDN binding domain, we engineer a Förster resonance energy transfer (FRET) based biosensor deemed BioSTING. Recombinant BioSTING affords real-time detection of CDN synthase activity and inhibition. Expression of BioSTING in live human cells allows quantification of localized bacterial and eukaryotic CDN levels in single cells with low nanomolar sensitivity. These findings establish BioSTING as a powerful kinetic in vitro platform amenable to high throughput screens and as a broadly applicable cellular tool to interrogate the temporal and spatial dynamics of CDN signaling in a variety of infectious, malignant, and autoimmune contexts.


Asunto(s)
Técnicas Biosensibles , Proteínas de la Membrana/química , Nucleótidos Cíclicos/análisis , Transducción de Señal , Animales , Bacillus subtilis/química , GMP Cíclico/metabolismo , Citometría de Flujo , Transferencia Resonante de Energía de Fluorescencia , Células HEK293 , Humanos , Inmunidad Innata , Ligandos , Ratones , Mutagénesis , Dominios Proteicos , Multimerización de Proteína , Proteínas Recombinantes
8.
Nature ; 573(7774): 434-438, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31511694

RESUMEN

The accumulation of DNA in the cytosol serves as a key immunostimulatory signal associated with infections, cancer and genomic damage1,2. Cytosolic DNA triggers immune responses by activating the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway3. The binding of DNA to cGAS activates its enzymatic activity, leading to the synthesis of a second messenger, cyclic guanosine monophosphate-adenosine monophosphate (2'3'-cGAMP)4-7. This cyclic dinucleotide (CDN) activates STING8, which in turn activates the transcription factors interferon regulatory factor 3 (IRF3) and nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB), promoting the transcription of genes encoding type I interferons and other cytokines and mediators that stimulate a broader immune response. Exogenous 2'3'-cGAMP produced by malignant cells9 and other CDNs, including those produced by bacteria10-12 and synthetic CDNs used in cancer immunotherapy13,14, must traverse the cell membrane to activate STING in target cells. How these charged CDNs pass through the lipid bilayer is unknown. Here we used a genome-wide CRISPR-interference screen to identify the reduced folate carrier SLC19A1, a folate-organic phosphate antiporter, as the major transporter of CDNs. Depleting SLC19A1 in human cells inhibits CDN uptake and functional responses, and overexpressing SLC19A1 increases both uptake and functional responses. In human cell lines and primary cells ex vivo, CDN uptake is inhibited by folates as well as two medications approved for treatment of inflammatory diseases, sulfasalazine and the antifolate methotrexate. The identification of SLC19A1 as the major transporter of CDNs into cells has implications for the immunotherapeutic treatment of cancer13, host responsiveness to CDN-producing pathogenic microorganisms11 and-potentially-for some inflammatory diseases.


Asunto(s)
ADN/metabolismo , Nucleótidos Cíclicos/metabolismo , Proteína Portadora de Folato Reducido/metabolismo , Animales , Citosol , ADN/inmunología , Humanos , Inflamación/genética , Inflamación/inmunología , Inflamación/metabolismo , Factor 3 Regulador del Interferón/metabolismo , Nucleótidos Cíclicos/inmunología , Nucleotidiltransferasas/metabolismo , Proteína Portadora de Folato Reducido/inmunología
9.
Methods Mol Biol ; 1280: 247-68, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25736753

RESUMEN

Nuclear factor-κB (NF-κB) signaling is classified into the canonical and noncanonical pathways. We describe in this chapter the methods used to study the noncanonical pathway, including derivation of primary cells, pathway stimulation, and immunoblotting.


Asunto(s)
FN-kappa B/metabolismo , Transducción de Señal , Péptidos y Proteínas Asociados a Receptores de Factores de Necrosis Tumoral/metabolismo , Animales , Linfocitos B/metabolismo , Western Blotting , Técnicas de Cultivo de Célula , Activación Enzimática , Fibroblastos/metabolismo , Separación Inmunomagnética/métodos , Macrófagos/metabolismo , Ratones , FN-kappa B/química , Cultivo Primario de Células , Unión Proteica , Transporte de Proteínas
10.
Nat Immunol ; 13(12): 1155-61, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23142775

RESUMEN

The induction of type I interferons by the bacterial secondary messengers cyclic di-GMP (c-di-GMP) or cyclic di-AMP (c-di-AMP) is dependent on a signaling axis that involves the adaptor STING, the kinase TBK1 and the transcription factor IRF3. Here we identified the heliase DDX41 as a pattern-recognition receptor (PRR) that sensed both c-di-GMP and c-di-AMP. DDX41 specifically and directly interacted with c-di-GMP. Knockdown of DDX41 via short hairpin RNA in mouse or human cells inhibited the induction of genes encoding molecules involved in the innate immune response and resulted in defective activation of STING, TBK1 and IRF3 in response to c-di-GMP or c-di-AMP. Our results suggest a mechanism whereby c-di-GMP and c-di-AMP are detected by DDX41, which forms a complex with STING to signal to TBK1-IRF3 and activate the interferon response.


Asunto(s)
GMP Cíclico/análogos & derivados , ARN Helicasas DEAD-box/metabolismo , Fosfatos de Dinucleósidos/metabolismo , Interferón Tipo I/inmunología , Listeria monocytogenes/inmunología , Listeria monocytogenes/metabolismo , Receptores de Reconocimiento de Patrones/metabolismo , Animales , Línea Celular , GMP Cíclico/metabolismo , ARN Helicasas DEAD-box/genética , Humanos , Inmunidad Innata , Factor 3 Regulador del Interferón/metabolismo , Macrófagos/inmunología , Proteínas de la Membrana/metabolismo , Ratones , Proteínas Serina-Treonina Quinasas/metabolismo , Interferencia de ARN , ARN Interferente Pequeño , Receptores de Reconocimiento de Patrones/genética , Sistemas de Mensajero Secundario , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA