Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cancer Res Commun ; 2(9): 937-950, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-36922936

RESUMEN

CLDN18.2 (Claudin18.2)-targeting therapeutic antibodies have shown promising clinical efficacy in approximately 30% of gastric cancers expressing high levels of CLDN18.2 and less pronounced activity in low expressing malignancies. Here, we report that ZL-1211 is a mAb targeting CLDN18.2 engineered to promote enhanced antibody-dependent cellular cytotoxicity (ADCC) with the goal of achieving more potent activity in a wider spectrum of high- and low-CLDN18.2 expressing tumors. ZL-1211 demonstrated more robust in vitro ADCC activity than clinical benchmark not only in CLDN18.2-high but also CLDN18.2-low expressing gastric tumor cell lines. Greater antitumor efficacy was also observed in mouse xenograft models. Natural killer (NK) cell played critical roles in ZL-1211 efficacy and NK-cell depletion abrogated ZL-1211-mediated ADCC activity in vitro. ZL-1211 efficacy in vivo was also dependent on the presence of an NK compartment. Strikingly, NK cells strongly induced an inflammatory response in response to ZL-1211 treatment, including increased IFNγ, TNFα, and IL6 production, and were recruited into tumor microenvironment in patient-derived gastric tumors expressing CLDN18.2 upon ZL-1211 treatment to lyse the tumor cells. Taken together, our data suggest that ZL-1211 more effectively targets CLDN18.2-high gastric cancers as well as -low expressing malignancies that may not be eligible for treatment with the leading clinical benchmark by inducing enhanced ADCC response and activating NK cells with robust inflammation to enhance antitumor efficacy. Clinical activity of ZL-1211 is currently under evaluation in a phase I clinical trial (NCT05065710). Significance: ZL-1211, anti-CLDN18.2 therapeutic antibody can target CLDN18.2-high as well as -low gastric cancers that may not be eligible for treatment with clinical benchmark. ZL-1211 treatment induces NK-cell activation with robust inflammation to further activate antitumor immunity in tumor microenvironment.


Asunto(s)
Neoplasias Gástricas , Ratones , Animales , Humanos , Neoplasias Gástricas/tratamiento farmacológico , Citotoxicidad Celular Dependiente de Anticuerpos , Células Asesinas Naturales , Línea Celular Tumoral , Inflamación/tratamiento farmacológico , Microambiente Tumoral
2.
Mol Cancer Res ; 17(4): 974-986, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30587523

RESUMEN

Ovarian cancer is the sixth most prevalent cancer in women and the most lethal of the gynecologic malignancies. Treatments have comprised the use of immunotherapeutic agents as well as oncolytic viruses, with varying results for reasons that remain to be clarified. To better understand the mechanisms that may help predict treatment outcome, we have evaluated innate immune signaling in select ovarian cancer cell lines, governed by the Stimulator of Interferon Genes (STING), which controls self or viral DNA-triggered cytokine production. Our results indicate that STING-dependent signaling is habitually defective in majority of ovarian cancer cells examined, frequently through the suppression of STING and/or the cyclic dinucleotide (CDN) enzyme Cyclic GMP-AMP synthase (cGAS) expression, by epigenetic processes. However, STING-independent, dsRNA-activated innate immune cytokine production, which require RIG-I/MDA5, were largely unaffected. Such defects enabled ovarian cancer cells to avoid DNA damage-mediated cytokine production, which would alert the immunosurveillance system. Loss of STING signaling also rendered ovarian cancer cells highly susceptible to viral oncolytic γ34.5 deleted-HSV1 (Herpes simplex virus) infection in vitro and in vivo. IMPLICATIONS: STING signaling evaluation in tumors may help predict disease outcome and possibly dictate the efficacy of oncoviral and other types of cancer therapies.


Asunto(s)
Proteínas de la Membrana/metabolismo , Viroterapia Oncolítica/métodos , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/terapia , Animales , Línea Celular Tumoral , Femenino , Humanos , Proteínas de la Membrana/biosíntesis , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Virus Oncolíticos/fisiología , Neoplasias Ováricas/genética , Neoplasias Ováricas/virología , Transducción de Señal , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Nature ; 564(7735): 268-272, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30479382

RESUMEN

T cells are key elements of cancer immunotherapy1 but certain fundamental properties, such as the development and migration of T cells within tumours, remain unknown. The enormous T cell receptor (TCR) repertoire, which is required for the recognition of foreign and self-antigens2, could serve as lineage tags to track these T cells in tumours3. Here we obtained transcriptomes of 11,138 single T cells from 12 patients with colorectal cancer, and developed single T cell analysis by RNA sequencing and TCR tracking (STARTRAC) indices to quantitatively analyse the dynamic relationships among 20 identified T cell subsets with distinct functions and clonalities. Although both CD8+ effector and 'exhausted' T cells exhibited high clonal expansion, they were independently connected with tumour-resident CD8+ effector memory cells, implicating a TCR-based fate decision. Of the CD4+ T cells, most tumour-infiltrating T regulatory (Treg) cells showed clonal exclusivity, whereas certain Treg cell clones were developmentally linked to several T helper (TH) cell clones. Notably, we identified two IFNG+ TH1-like cell clusters in tumours that were associated with distinct IFNγ-regulating transcription factors -the GZMK+ effector memory T cells, which were associated with EOMES and RUNX3, and CXCL13+BHLHE40+ TH1-like cell clusters, which were associated with BHLHE40. Only CXCL13+BHLHE40+ TH1-like cells were preferentially enriched in patients with microsatellite-instable tumours, and this might explain their favourable responses to immune-checkpoint blockade. Furthermore, IGFLR1 was highly expressed in both CXCL13+BHLHE40+ TH1-like cells and CD8+ exhausted T cells and possessed co-stimulatory functions. Our integrated STARTRAC analyses provide a powerful approach to dissect the T cell properties in colorectal cancer comprehensively, and could provide insights into the dynamic relationships of T cells in other cancers.


Asunto(s)
Linfocitos T CD4-Positivos/citología , Linfocitos T CD8-positivos/citología , Linaje de la Célula , Movimiento Celular , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/patología , Proteínas Adaptadoras Transductoras de Señales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Proteínas Portadoras/metabolismo , Rastreo Celular , Células Cultivadas , Células Clonales/citología , Células Clonales/inmunología , Humanos , Células TH1/citología , Células TH1/inmunología
4.
Front Microbiol ; 9: 2347, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30319597

RESUMEN

Cyclic dimeric adenosine 3',5'-monophosphate (c-di-AMP), a recently identified secondary messenger in bacteria, plays a role in several bacterial processes, including biofilm formation. It is enzymatically produced by diadenylate cyclase and cleaved by c-di-AMP phosphodiesterase. c-di-AMP is believed to be essential for the viability of bacterial cells that produce it. In the current study, the biochemical and biological roles of GdpP (SMU_2140c) and DhhP (SMU_1297), two distinct Streptococcus mutans phosphodiesterases involved in the pathway producing AMP from c-di-AMP, were investigated. Liquid chromatography-tandem mass spectrometry revealed that c-di-AMP was degraded to phosphoadenylyl adenosine (pApA) by truncated recombinant GdpP, and pApA was cleaved by recombinant DhhP to yield AMP. In-frame deletion mutants lacking the dhhP gene (ΔdhhP) and both the gdpP and dhhP genes (ΔgdpPΔdhhP) displayed significantly more biofilm formation than the wild-type and a mutant strain lacking the gdpP gene (ΔgdpP; p < 0.01). Furthermore, biofilm formation was restored to the level of the wild type strain upon complementation with dhhP. Optical and electron microscopy observations revealed that ΔdhhP and ΔgdpPΔdhhP mutants self-aggregated into large cell clumps, correlated with increased biofilm formation, but cell clumps were not observed in cultures of wild-type, ΔgdpP, or strains complemented with gdpP and dhhP. Thus, deletion of dhhP presumably leads to the formation of bacterial cell aggregates and a subsequent increase in biofilm production.

5.
Cell Rep ; 23(4): 1112-1123, 2018 04 24.
Artículo en Inglés | MEDLINE | ID: mdl-29694889

RESUMEN

The cellular sensor stimulator of interferon genes (STING) initiates type I interferon (IFN) and cytokine production following association with cyclic dinucleotides (CDNs) generated from intracellular bacteria or via a cellular synthase, cGAS, after binding microbial or self-DNA. Although essential for protecting the host against infection, unscheduled STING signaling is now known to be responsible for a variety of autoinflammatory disorders. Here, we report a gain-of-function mutation in STING (R284S), isolated from a patient who did not require CDNs to augment activity and who manifested a constitutively active phenotype. Control of the Unc-51-like autophagy activating kinase 1 (ULK1) pathway, which has previously been shown to influence STING function, was potently able to suppress STING (R284S) activity to alleviate cytokine production. Our findings add to the growing list of inflammatory syndromes associated with spontaneous STING signaling and provide a therapeutic strategy for the treatment of STING-induced inflammatory disease.


Asunto(s)
Autofagia/inmunología , Mutación con Ganancia de Función , Proteínas de la Membrana/inmunología , Animales , Autofagia/genética , Homólogo de la Proteína 1 Relacionada con la Autofagia/genética , Homólogo de la Proteína 1 Relacionada con la Autofagia/inmunología , Células HEK293 , Humanos , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/inmunología , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados
6.
Oncogene ; 37(15): 2037-2051, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29367762

RESUMEN

The production of cytokines in response to DNA-damage events may be an important host defense response to help prevent the escape of pre-cancerous cells. The innate immune pathways involved in these events are known to be regulated by cellular molecules such as stimulator of interferon genes (STING), which controls type I interferon and pro-inflammatory cytokine production in response to the presence of microbial DNA or cytosolic DNA that has escaped from the nucleus. STING signaling has been shown to be defective in a variety of cancers, such as colon cancer and melanoma, actions that may enable damaged cells to escape the immunosurveillance system. Here, we report through examination of databases that STING signaling may be commonly suppressed in a greater variety of tumors due to loss-of-function mutation or epigenetic silencing of the STING/cGAS promoter regions. In comparison, RNA activated innate immune pathways controlled by RIG-I/MDA5 were significantly less affected. Examination of reported missense STING variants confirmed that many exhibited a loss-of-function phenotype and could not activate cytokine production following exposure to cytosolic DNA or DNA-damage events. Our data imply that the STING signaling pathway may be recurrently suppressed by a number of mechanisms in a considerable variety of malignant disease and be a requirement for cellular transformation.


Asunto(s)
Citocinas/toxicidad , Citoprotección/genética , Daño del ADN/genética , Silenciador del Gen/fisiología , Proteínas de la Membrana/genética , Mutación Missense/fisiología , Animales , Células Cultivadas , Chlorocebus aethiops , Citocinas/metabolismo , Epigénesis Genética/fisiología , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Mediadores de Inflamación/metabolismo , Mediadores de Inflamación/toxicidad , Ratones , Ratones Noqueados , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/metabolismo , Nucleotidiltransferasas/genética , Transducción de Señal/genética , Transducción de Señal/inmunología , Células Vero
7.
Sci Immunol ; 2(11)2017 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-28763789

RESUMEN

Cytosolic DNA species derived from invading microbes or leaked from the nuclear or mitochondrial compartments of the cell can trigger the induction of host defense genes by activating the endoplasmic reticulum-associated protein STING (stimulator of interferon genes). Using a mass spectrometry-based approach, we show that after association with cyclic dinucleotides, delivery of Tank-binding kinase 1 to interferon regulatory factors (IRFs), such as IRF3, relies on K63-linked ubiquitination of K224 on STING. Blocking K224 ubiquitination specifically prevented IRF3 but not nuclear factor κB activation, additionally indicating that STING trafficking is not required to stimulate the latter signaling pathway. By carrying out a limited small interfering RNA screen, we have identified MUL1 (mitochondrial E3 ubiquitin protein ligase 1) as an E3 ligase that catalyzes the ubiquitination of STING on K224. These data demonstrate the critical role of K224 ubiquitination in STING function and provide molecular insight into the mechanisms governing host defense responses.

8.
Cancer Res ; 76(22): 6747-6759, 2016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-27680683

RESUMEN

The innate immunoregulator STING stimulates cytokine production in response to the presence of cytosolic DNA, which can arise following DNA damage. Extrinsic STING signaling is also needed for antigen-presenting cells to stimulate antitumor T-cell immunity. Here, we show that STING signaling is recurrently suppressed in melanoma cells, where this event may enable immune escape after DNA damage. Mechanistically, STING signaling was suppressed most frequently by epigenetic silencing of either STING or the cyclic GMP-AMP synthase, which generates STING-activating cyclic dinucleotides after binding cytosolic DNA species. Loss of STING function rendered melanoma cells unable to produce type I IFN and other immune cytokines after exposure to cytosolic DNA species. Consequently, such cells were highly susceptible to infection with DNA viruses including HSV1, a variant of which is being developed presently as a therapeutic oncolytic virus [talimogene laherparepvec (T-VEC)]. Our findings provide insight into the basis for susceptibility to viral oncolysis by agents such as HSV1. Cancer Res; 76(22); 6747-59. ©2016 AACR.


Asunto(s)
Melanoma/inmunología , Virus Oncolíticos/inmunología , Animales , Humanos , Ratones , Transducción de Señal
9.
Nat Commun ; 7: 11932, 2016 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-27324217

RESUMEN

Stimulator of interferon genes (STING) is essential for the type I interferon response against DNA pathogens. In response to the presence of DNA and/or cyclic dinucleotides, STING translocates from the endoplasmic reticulum to perinuclear compartments. However, the role of this subcellular translocation remains poorly defined. Here we show that palmitoylation of STING at the Golgi is essential for activation of STING. Treatment with palmitoylation inhibitor 2-bromopalmitate (2-BP) suppresses palmitoylation of STING and abolishes the type I interferon response. Mutation of two membrane-proximal Cys residues (Cys88/91) suppresses palmitoylation, and this STING mutant cannot induce STING-dependent host defense genes. STING variants that constitutively induce the type I interferon response were found in patients with autoimmune diseases. The response elicited by these STING variants is effectively inhibited by 2-BP or an introduction of Cys88/91Ser mutation. Our results may lead to new treatments for cytosolic DNA-triggered autoinflammatory diseases.


Asunto(s)
Fibroblastos/inmunología , Aparato de Golgi/metabolismo , Inmunidad Innata , Interferón Tipo I/genética , Proteínas de la Membrana/genética , Animales , Células COS , Chlorocebus aethiops , Embrión de Mamíferos , Retículo Endoplásmico/inmunología , Retículo Endoplásmico/metabolismo , Fibroblastos/virología , Regulación de la Expresión Génica , Aparato de Golgi/inmunología , Células HEK293 , Herpesvirus Humano 1/crecimiento & desarrollo , Herpesvirus Humano 1/inmunología , Humanos , Interferón Tipo I/inmunología , Lipoilación , Proteínas de la Membrana/agonistas , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/inmunología , Ratones , Mutación , Palmitatos/farmacología , Cultivo Primario de Células , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/inmunología , Transporte de Proteínas , Xantonas/farmacología
10.
Cell Rep ; 14(2): 282-97, 2016 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-26748708

RESUMEN

Stimulator of interferon genes (STING) has been shown to be critical for controlling antiviral responses as well as anti-tumor adaptive immunity, but little is known regarding its regulation in human tumors. Here, we report that STING signaling is recurrently suppressed in a wide variety of cancers, including colorectal carcinoma. Loss of STING signaling impeded DNA damage responses accountable for generating key cytokines that facilitate tissue repair and anti-tumor T cell priming, such as type I interferons (IFNs). Correspondingly, defective STING function was also highly predictive of effectual DNA-virus-mediated oncolytic activity. Thus, impaired STING responses may enable damaged cells to evade host immunosurveillance processes, although they provide a critical prognostic measurement that could help predict the outcome of effective oncoviral therapy.


Asunto(s)
Carcinogénesis/genética , Neoplasias Colorrectales/genética , Daño del ADN/genética , Animales , Humanos , Ratones , Transducción de Señal , Transfección
11.
Blood ; 127(5): 605-15, 2016 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-26603838

RESUMEN

GLI1 oncogene has been implicated in the pathobiology of several neoplasms including diffuse large B-cell lymphoma (DLBCL). However, mechanisms underlying GLI1-increased activity in DLBCL are poorly characterized. Herein, we demonstrate that IKKß phosphorylates GLI1 in DLBCL. IKKß activation increased GLI1 protein levels and transcriptional activity, whereas IKKß silencing decreased GLI1 levels and transcriptional activity. Tumor necrosis factor-α (TNFα) mediated IKKß activation-impaired GLI1 binding with the E3 ubiquitin ligase-ITCH, leading to decreased K48-linked ubiquitination/degradation of GLI1. We found 8 IKKß-dependent phosphorylation sites that mediate GLI1 stability. Mutating or deleting these residues facilitated GLI1-ITCH interaction and decreased the protective effect of TNFα on GLI1 stability. IKKß-GLI1 crosstalk is significant because combined inhibition of both molecules resulted in synergistic suppression of DLBCL viability in vivo and in vitro. By linking IKKß-mediated nuclear factor-κB activity with GLI1, we identified a crosstalk between these 2 pathways that can inform the design of novel therapeutic strategies in DLBCL.


Asunto(s)
Quinasa I-kappa B/metabolismo , Linfoma de Células B Grandes Difuso/metabolismo , Factores de Transcripción/metabolismo , Línea Celular Tumoral , Supervivencia Celular , Humanos , Linfoma de Células B Grandes Difuso/genética , FN-kappa B/metabolismo , Fosforilación , Estabilidad Proteica , Proteínas Represoras/metabolismo , Factores de Transcripción/química , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , Proteína con Dedos de Zinc GLI1
12.
J Exp Med ; 212(11): 1967-85, 2015 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-26458771

RESUMEN

TRAF-interacting protein with forkhead-associated domain B (TIFAB) is a haploinsufficient gene in del(5q) myelodysplastic syndrome (MDS). Deletion of Tifab results in progressive bone marrow (BM) and blood defects, including skewed hematopoietic stem/progenitor cell (HSPC) proportions and altered myeloid differentiation. A subset of mice transplanted with Tifab knockout (KO) HSPCs develop a BM failure with neutrophil dysplasia and cytopenia. In competitive transplants, Tifab KO HSPCs are out-competed by wild-type (WT) cells, suggesting a cell-intrinsic defect. Gene expression analysis of Tifab KO HSPCs identified dysregulation of immune-related signatures, and hypersensitivity to TLR4 stimulation. TIFAB forms a complex with TRAF6, a mediator of immune signaling, and reduces TRAF6 protein stability by a lysosome-dependent mechanism. In contrast, TIFAB loss increases TRAF6 protein and the dynamic range of TLR4 signaling, contributing to ineffective hematopoiesis. Moreover, combined deletion of TIFAB and miR-146a, two genes associated with del(5q) MDS/AML, results in a cooperative increase in TRAF6 expression and hematopoietic dysfunction. Re-expression of TIFAB in del(5q) MDS/AML cells results in attenuated TLR4 signaling and reduced viability. These findings underscore the importance of efficient regulation of innate immune/TRAF6 signaling within HSPCs by TIFAB, and its cooperation with miR-146a as it relates to the pathogenesis of hematopoietic malignancies, such as del(5q) MDS/AML.


Asunto(s)
Hematopoyesis , Proteínas/fisiología , Transducción de Señal/fisiología , Factor 6 Asociado a Receptor de TNF/fisiología , Receptores Toll-Like/fisiología , Animales , Apoptosis , Trasplante de Médula Ósea , Diferenciación Celular , Cromosomas Humanos Par 5 , Humanos , Péptidos y Proteínas de Señalización Intracelular , Masculino , Ratones , Ratones Endogámicos C57BL , MicroARNs/fisiología , FN-kappa B/antagonistas & inhibidores , FN-kappa B/fisiología , Proteínas/genética
13.
Sci Rep ; 5: 10758, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-26029823

RESUMEN

Nuclear factor (NF)-κB-inducing kinase (NIK) is a serine/threonine kinase that activates NF-κB pathways, thereby regulating a wide variety of immune systems. Aberrant NIK activation causes tumor malignancy, suggesting a requirement for precise regulation of NIK activity. To explore novel interacting proteins of NIK, we performed in vitro virus screening and identified the catalytic subunit Aα isoform of serine/threonine phosphatase calcineurin (CnAα) as a novel NIK-interacting protein. The interaction of NIK with CnAα in living cells was confirmed by co-immunoprecipitation. Calcineurin catalytic subunit Aß isoform (CnAß) also bound to NIK. Experiments using domain deletion mutants suggested that CnAα and CnAß interact with both the kinase domain and C-terminal region of NIK. Moreover, the phosphatase domain of CnAα is responsible for the interaction with NIK. Intriguingly, we found that TRAF3, a critical regulator of NIK activity, also binds to CnAα and CnAß. Depletion of CnAα and CnAß significantly enhanced lymphotoxin-ß receptor (LtßR)-mediated expression of the NIK-dependent gene Spi-B and activation of RelA and RelB, suggesting that CnAα and CnAß attenuate NF-κB activation mediated by LtßR-NIK signaling. Overall, these findings suggest a possible role of CnAα and CnAß in modifying NIK functions.


Asunto(s)
Calcineurina/metabolismo , Regulación de la Expresión Génica , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Dominio Catalítico , Línea Celular/metabolismo , Citocina TWEAK , Humanos , Isoenzimas , Receptor beta de Linfotoxina/metabolismo , Ratones , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Transporte de Proteínas , Proteínas Proto-Oncogénicas c-ets/genética , Transducción de Señal , Factor 3 Asociado a Receptor de TNF/metabolismo , Factor de Transcripción ReIA/metabolismo , Factor de Transcripción ReIB/metabolismo , Factores de Transcripción/genética , Factores de Necrosis Tumoral/metabolismo , Quinasa de Factor Nuclear kappa B
14.
Microbes Infect ; 16(12): 998-1001, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25449752

RESUMEN

The innate immune system is critically important for the primary sensing of invading pathogens. Over the past decade, the cellular sensors important for recognizing microbial entry into the host cell have been largely elucidated. These sensors, some of which are evolutionarily conserved, include the Toll-like receptor (TLR) and RIG-I-like helicase family (RLH) pathway that can recognize bacterial and viral non-self nucleic acid. In addition, a cellular sensor referred to as STING (for stimulator of interferon genes) has been shown to be critical for triggering host defense countermeasures, including stimulation of the adaptive immune response, following the detection of cytosolic DNA species. The STING pathway has now been shown to be critical for activating innate immune gene transcription in response to infection by DNA pathogens such as herpes simplex virus 1 (HSV1) as well as retroviruses. In addition, it is clear that chronic STING activation can also cause autoinflammatory disease manifested by self-DNA. Here we review recent developments in our understanding of STING function, including importance in the control of microbial disease.


Asunto(s)
Enfermedades Transmisibles/inmunología , Enfermedades Transmisibles/metabolismo , ADN/inmunología , Interacciones Huésped-Patógeno , Inmunidad Innata/fisiología , Proteínas de la Membrana/metabolismo , Transducción de Señal , Animales , Enfermedades Transmisibles/genética , Citosol/metabolismo , Humanos , Proteínas de la Membrana/genética
15.
Nat Commun ; 5: 5166, 2014 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-25300616

RESUMEN

Chronic stimulation of innate immune pathways by microbial agents or damaged tissue is known to promote inflammation-driven tumorigenesis by mechanisms that are not well understood. Here we demonstrate that mutagenic 7,12-dimethylbenz(a)anthracene (DMBA), cisplatin and etoposide induce nuclear DNA leakage into the cytosol that intrinsically activates stimulator of interferon genes (STING)-dependent cytokine production. Inflammatory cytokine levels are subsequently augmented in a STING-dependent extrinsic manner by infiltrating phagocytes purging dying cells. Consequently, STING(-/-) mice, or wild-type mice adoptively transferred with STING(-/-) bone marrow, are almost completely resistant to DMBA-induced skin carcinogenesis compared with their wild-type counterparts. Our data establish a role for STING in the control of cancer, shed significant insight into the causes of inflammation-driven carcinogenesis and may provide a basis for therapeutic strategies to help prevent malignant disease.


Asunto(s)
Proteínas de la Membrana/inmunología , Neoplasias Cutáneas/inmunología , 9,10-Dimetil-1,2-benzantraceno/toxicidad , Animales , Carcinogénesis/genética , Carcinogénesis/inmunología , Carcinogénesis/patología , Citocinas/genética , Citocinas/inmunología , Exodesoxirribonucleasas/genética , Exodesoxirribonucleasas/inmunología , Femenino , Humanos , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Fosfoproteínas/genética , Fosfoproteínas/inmunología , Neoplasias Cutáneas/inducido químicamente , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología
16.
PLoS One ; 9(5): e95992, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24788966

RESUMEN

Virus-derived double-stranded RNAs (dsRNAs) are sensed in the cytosol by retinoic acid-inducible gene (RIG)-I-like receptors (RLRs). These induce the expression of type I IFN and proinflammatory cytokines through signaling pathways mediated by the mitochondrial antiviral signaling (MAVS) protein. TNF receptor-associated factor (TRAF) family proteins are reported to facilitate the RLR-dependent expression of type I IFN by interacting with MAVS. However, the precise regulatory mechanisms remain unclear. Here, we show the role of FK506-binding protein 51 (FKBP51) in regulating the dsRNA-dependent expression of type I IFN. The binding of FKBP51 to TRAF6 was first identified by "in vitro virus" selection and was subsequently confirmed with a coimmunoprecipitation assay in HEK293T cells. The TRAF-C domain of TRAF6 is required for its interaction, although FKBP51 does not contain the consensus motif for interaction with the TRAF-C domain. Besides TRAF6, we found that FKBP51 also interacts with TRAF3. The depletion of FKBP51 reduced the expression of type I IFN induced by dsRNA transfection or Newcastle disease virus infection in murine fibroblasts. Consistent with this, the FKBP51 depletion attenuated dsRNA-mediated phosphorylations of IRF3 and JNK and nuclear translocation of RelA. Interestingly, dsRNA stimulation promoted the accumulation of FKBP51 in the mitochondria. Moreover, the overexpression of FKBP51 inhibited RLR-dependent transcriptional activation, suggesting a scaffolding function for FKBP51 in the MAVS-mediated signaling pathway. Overall, we have demonstrated that FKBP51 interacts with TRAF proteins and facilitates the expression of type I IFN induced by cytosolic dsRNA. These findings suggest a novel role for FKBP51 in the innate immune response to viral infection.


Asunto(s)
Núcleo Celular/metabolismo , Regulación de la Expresión Génica , Interferón Tipo I/genética , Mitocondrias/metabolismo , Proteínas de Unión a Tacrolimus/metabolismo , Péptidos y Proteínas Asociados a Receptores de Factores de Necrosis Tumoral/metabolismo , Animales , Línea Celular , Humanos , Inmunidad Innata , Interferón Tipo I/metabolismo , Ratones , Virus de la Enfermedad de Newcastle/genética , Virus de la Enfermedad de Newcastle/inmunología , Virus de la Enfermedad de Newcastle/metabolismo , Unión Proteica , Mapeo de Interacción de Proteínas , Mapas de Interacción de Proteínas , Transporte de Proteínas , ARN Bicatenario/metabolismo , Transducción de Señal , Factor 3 Asociado a Receptor de TNF/metabolismo , Factor 6 Asociado a Receptor de TNF/metabolismo , Proteínas de Unión a Tacrolimus/genética
17.
APMIS ; 122(10): 951-60, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24628454

RESUMEN

Streptococcus sanguinis is an early colonizer of tooth surfaces and forms biofilms with other species of microorganisms. In vitro, S. sanguinis produces water-soluble glucans from sucrose and releases them into the culture supernatant; however, the role played by these glucans in biofilm formation is unclear. The present study examined both the effect of glucans on biofilm formation by S. sanguinis and the proportion of this bacterial species within the biofilms. Inactivation of the gtfP gene, annotated as glucosyltransferase in the S. sanguinis genome database, caused a marked reduction in the amount of water-soluble glucans in the culture supernatant, but not in the amount of water-insoluble glucans expressed on the bacterial cell surface. Scanning electron microscopy revealed that wild-type S. sanguinis, but not the gtfP-deficient mutant, produced large amounts of sticky material in the presence of 1% (w/v) sucrose. In addition, biofilm production by wild-type bacteria was greater than that by the mutant strain. By contrast, co-culture of mutant bacteria with Streptococcus mutans, S. sobrinus, S. oralis, S. gordonii, S. anginosus, or S. salivarius showed that inactivating the gtfP gene had little effect on the amount of biofilm produced. Furthermore, inactivating the gtfP gene did not greatly alter the proportion of S. sanguinis in the biofilms formed by the co-cultures. Thus, despite the role of S. sanguinis glucosyltransferase in formation of water-soluble glucans and biofilms in monoculture, the functional gene contributed little to biofilms in co-culture experiments.


Asunto(s)
Proteínas Bacterianas/metabolismo , Biopelículas/crecimiento & desarrollo , Glucosiltransferasas/metabolismo , Streptococcus/metabolismo , Adhesión Bacteriana/genética , Adhesión Bacteriana/fisiología , Proteínas Bacterianas/genética , Técnicas de Cocultivo , Genes Bacterianos/genética , Glucanos/genética , Glucanos/metabolismo , Glucosiltransferasas/genética , Streptococcus/genética
18.
Cell ; 155(3): 688-98, 2013 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-24119841

RESUMEN

Activation of the stimulator of interferon genes (STING) pathway by microbial or self-DNA, as well as cyclic dinucleotides (CDNs), results in the induction of numerous genes that suppress pathogen replication and facilitate adaptive immunity. However, sustained gene transcription is rigidly prevented to avoid lethal STING-dependent proinflammatory disease by mechanisms that remain unknown. We demonstrate here that, after autophagy-dependent STING delivery of TANK-binding kinase 1 (TBK1) to endosomal/lysosomal compartments and activation of transcription factors interferon regulatory factor 3 (IRF3) and NF-κB, STING is subsequently phosphorylated by serine/threonine UNC-51-like kinase (ULK1/ATG1), and IRF3 function is suppressed. ULK1 activation occurred following disassociation from its repressor AMP activated protein kinase (AMPK) and was elicited by CDNs generated by the cGAMP synthase, cGAS. Thus, although CDNs may initially facilitate STING function, they subsequently trigger negative-feedback control of STING activity, thus preventing the persistent transcription of innate immune genes.


Asunto(s)
Inmunidad Innata , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Secuencia de Aminoácidos , Homólogo de la Proteína 1 Relacionada con la Autofagia , Humanos , Proteínas de la Membrana/química , Datos de Secuencia Molecular , Nucleótidos Cíclicos/metabolismo , Fosforilación , Alineación de Secuencia
19.
Mol Cell ; 50(1): 5-15, 2013 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-23478444

RESUMEN

How the cell recognizes cytosolic DNA including DNA-based microbes to trigger host-defense-related gene activation remains to be fully resolved. Here, we demonstrate that STING (stimulator of interferon genes), an endoplasmic reticulum translocon-associated transmembrane protein, acts to detect cytoplasmic DNA species. STING homodimers were able to complex with self- (apoptotic, necrotic) or pathogen-related ssDNA and dsDNA and were indispensible for HSV-1-mediated transcriptional activation of a wide array of innate immune and proinflammatory genes in addition to type I IFN. Our data indicate that STING instigates cytoplasmic DNA-mediated cellular defense gene transcription and facilitates adoptive responses that are required for protection of the host. In contrast, chronic STING activation may manifest inflammatory responses and possibly autoimmune disease triggered by self-DNA.


Asunto(s)
Citoplasma/metabolismo , ADN de Cadena Simple/metabolismo , Proteínas de la Membrana/metabolismo , Animales , Apoptosis , Sitios de Unión , Citoplasma/inmunología , ADN de Cadena Simple/inmunología , Genes Reporteros , Células HEK293 , Herpesvirus Humano 1/inmunología , Humanos , Inmunidad Innata , Mediadores de Inflamación/metabolismo , Interferón Tipo I/genética , Interferón Tipo I/metabolismo , Luciferasas de Luciérnaga/biosíntesis , Luciferasas de Luciérnaga/genética , Proteínas de la Membrana/genética , Ratones , Necrosis , Multimerización de Proteína , Interferencia de ARN , Telomerasa/genética , Telomerasa/metabolismo , Transcripción Genética , Transfección
20.
Biochem Biophys Res Commun ; 408(4): 530-6, 2011 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-21527253

RESUMEN

Medullary thymic epithelial cells (mTECs) are essential for thymic negative selection to prevent autoimmunity. Previous studies show that mTEC development is dependent on the signal transducers TRAF6 and NIK. However, the downstream target genes of signals controlled by these molecules remain unknown. We performed a microarray analysis on mRNAs down-regulated by deficiencies in TRAF6 or functional NIK in an in vitro organ culture of fetal thymic stromata (2DG-FTOC). An in silico analysis of transcription factor binding sites in plausible promoter regions of differentially expressed genes suggests that STAT1 is involved in TRAF6- and NIK-dependent gene expression. Indeed, the signal of RANK, a TNF receptor family member that activates TRAF6 and NIK, induces the activation of STAT1 in 2DG-FTOC. Moreover, RANK signaling induces the up-regulation of interferon (IFN)-stimulated gene (ISG) expression, suggesting that the RANKL-dependent activation of STAT1 up-regulates ISG expression. The RANKL-dependent expression levels of ISGs were reduced but not completely abolished in interferon α receptor 1-deficient (Ifnar1(-/-)) 2DG-FTOC. Our data suggest that RANK signaling induces ISG expression in both type I interferon-independent and interferon-dependent mechanisms.


Asunto(s)
Células Epiteliales/inmunología , Regulación de la Expresión Génica , Interferón Tipo I/inmunología , Receptor Activador del Factor Nuclear kappa-B/metabolismo , Autotolerancia/genética , Timo/inmunología , Animales , Feto , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Receptor Activador del Factor Nuclear kappa-B/genética , Receptor de Interferón alfa y beta/genética , Transducción de Señal , Células del Estroma/inmunología , Factor 6 Asociado a Receptor de TNF/genética , Factor 6 Asociado a Receptor de TNF/metabolismo , Quinasa de Factor Nuclear kappa B
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...