Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Blood ; 134(20): 1730-1744, 2019 11 14.
Artículo en Inglés | MEDLINE | ID: mdl-31434702

RESUMEN

Heterozygous deletions within chromosome 20q, or del(20q), are frequent cytogenetic abnormalities detected in hematologic malignancies. To date, identification of genes in the del(20q) common deleted region that contribute to disease development have remained elusive. Through assessment of patient gene expression, we have identified STK4 (encoding Hippo kinase MST1) as a 20q gene that is downregulated below haploinsufficient amounts in myelodysplastic syndrome (MDS) and myeloproliferative neoplasm (MPN). Hematopoietic-specific gene inactivation in mice revealed Hippo kinase loss to induce splenomegaly, thrombocytopenia, megakaryocytic dysplasia, and a propensity for chronic granulocytosis; phenotypes that closely resemble those observed in patients harboring del(20q). In a JAK2-V617F model, heterozygous Hippo kinase inactivation led to accelerated development of lethal myelofibrosis, recapitulating adverse MPN disease progression and revealing a novel genetic interaction between these 2 molecular events. Quantitative serum protein profiling showed that myelofibrotic transformation in mice was associated with cooperative effects of JAK2-V617F and Hippo kinase inactivation on innate immune-associated proinflammatory cytokine production, including IL-1ß and IL-6. Mechanistically, MST1 interacted with IRAK1, and shRNA-mediated knockdown was sufficient to increase IRAK1-dependent innate immune activation of NF-κB in human myeloid cells. Consistent with this, treatment with a small molecule IRAK1/4 inhibitor rescued the aberrantly elevated IL-1ß production in the JAK2-V617F MPN model. This study identified Hippo kinase MST1 (STK4) as having a central role in the biology of del(20q)-associated hematologic malignancies and revealed a novel molecular basis of adverse MPN progression that may be therapeutically exploitable via IRAK1 inhibition.


Asunto(s)
Neoplasias Hematológicas/genética , Síndromes Mielodisplásicos/genética , Trastornos Mieloproliferativos/genética , Proteínas Serina-Treonina Quinasas/genética , Animales , Deleción Cromosómica , Cromosomas Humanos Par 20/genética , Regulación hacia Abajo , Regulación Neoplásica de la Expresión Génica , Neoplasias Hematológicas/inmunología , Humanos , Inmunidad Innata , Péptidos y Proteínas de Señalización Intracelular , Ratones , Síndromes Mielodisplásicos/inmunología , Trastornos Mieloproliferativos/inmunología , Proteínas Serina-Treonina Quinasas/inmunología
2.
J Immunol ; 196(9): 3887-95, 2016 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-27016605

RESUMEN

A balance between bone formation and bone resorption is critical for the maintenance of bone mass. In many pathological conditions, including chronic inflammation, uncontrolled activation of osteoclast differentiation often causes excessive bone resorption that results in osteoporosis. In this study, we identified the osteopenia phenotype of mice lacking Usp18 (also called Ubp43), which is a deISGylating enzyme and is known as a negative regulator of type I IFN signaling. The expression of Usp18 was induced in preosteoclasts upon receptor activator of NF-κB ligand (RANKL) treatment. In an in vitro osteoclast-differentiation assay, bone marrow macrophages from Usp18-deficient mice exhibited an enhanced differentiation to multinucleated cells, elevated activation of NFATc1, and an increased expression of osteoclast marker genes upon RANKL treatment. Furthermore, in vitro quantification of bone resorption revealed a great increase in osteoclastic activities in Usp18-deficient cells. Interestingly, proinflammatory cytokine genes, such as IP-10 (CXCL10), were highly expressed in Usp18-deficient bone marrow macrophages upon RANKL treatment compared with wild-type cells. In addition, serum cytokine levels, especially IP-10, were significantly high in Usp18-knockout mice. In sum, we suggest that, although type I IFN is known to restrict osteoclast differentiation, the exaggerated activation of the type I IFN response in Usp18-knockout mice causes an osteopenia phenotype in mice.


Asunto(s)
Macrófagos/fisiología , Osteoclastos/fisiología , Osteogénesis , Osteoporosis/inmunología , Ubiquitina Tiolesterasa/metabolismo , Animales , Diferenciación Celular , Células Cultivadas , Quimiocina CXCL10/metabolismo , Interferón Tipo I/metabolismo , Ratones , Ratones de la Cepa 129 , Ratones Noqueados , Factores de Transcripción NFATC/metabolismo , Osteogénesis/genética , Osteogénesis/inmunología , Ligando RANK/metabolismo , Ubiquitina Tiolesterasa/genética
3.
Proc Natl Acad Sci U S A ; 112(46): 14313-8, 2015 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-26515094

RESUMEN

Type I IFNs have broad activity in tissue inflammation and malignant progression that depends on the expression of IFN-stimulated genes (ISGs). ISG15, one such ISG, can form covalent conjugates to many cellular proteins, a process termed "protein ISGylation." Although type I IFNs are involved in multiple inflammatory disorders, the role of protein ISGylation during inflammation has not been evaluated. Here we report that protein ISGylation exacerbates intestinal inflammation and colitis-associated colon cancer in mice. Mechanistically, we demonstrate that protein ISGylation negatively regulates the ubiquitin-proteasome system, leading to increased production of IFN-induced reactive oxygen species (ROS). The increased cellular ROS then enhances LPS-induced activation of p38 MAP kinase and the expression of inflammation-related cytokines in macrophages. Thus our studies reveal a regulatory role for protein ISGylation in colonic inflammation and its related malignant progression, indicating that targeting ubiquitin-activating enzyme E1 homolog has therapeutic potential in treating inflammatory diseases.


Asunto(s)
Colitis/metabolismo , Colon/metabolismo , Interferón Tipo I/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Enzimas Activadoras de Ubiquitina/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Colitis/inducido químicamente , Colitis/patología , Colon/patología , Lipopolisacáridos/toxicidad , Ratones
4.
Biochem Biophys Res Commun ; 448(1): 95-100, 2014 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-24769207

RESUMEN

ISGylation, an ubiquitin-like post-translational modification by ISG15, has been reported to participate in the interferon (IFN)-mediated antiviral response. In this study, we analyzed the functional role of ISGylation in dengue virus 2 (DENV-2) replication. Overexpression of ISG15 was found to significantly suppress the amount of extracellular infectious virus released, while intracellular viral RNA was unaffected. This effect was not observed with a conjugation-defective ISG15 mutant. In addition, extracellular virus infectivity was decreased by ISG15 overexpression. To further clarify the role of ISGylation in the anti-DENV-2 response, we depleted endogenous ISG15 by RNA interference and analyzed the virus production in the absence or presence of type-I IFN. Results showed a significant reduction in extracellular DENV-2 RNA levels for cells treated with IFN, and that these DENV-2 RNA levels could be partially restored by the ISG15 knockdown. Among various DENV-2 proteins, NS3 and NS5 were subjected to the ISGylation. These results demonstrate that IFN-inducible ISGylation suppresses DENV-2 particle release, and that ISG15 is one of the mediators of IFN-induced inhibition of DENV-2 replication. ISG15 therefore functions as a host antiviral factor against DENV-2 infection.


Asunto(s)
Citocinas/metabolismo , Virus del Dengue/fisiología , Interferón Tipo I/farmacología , Ubiquitinas/metabolismo , Replicación Viral/efectos de los fármacos , Animales , Línea Celular , Cricetinae , Virus del Dengue/efectos de los fármacos , Células HEK293 , Células HeLa , Humanos , Interferencia de ARN , Proteínas no Estructurales Virales/metabolismo
5.
Brain Res ; 1840: 149082, 2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-38866307

RESUMEN

Ubiquitin specific protease 18 (USP18) serves as a potent inhibitor of Type I interferon (IFN) signaling. Previous studies have shown that Usp18 deficient (homozygous Usp18 gene knockout) mice exhibit hydrocephalus; however, the precise molecular mechanism underlying hydrocephalus development remains elusive. In this study, we demonstrate that mice lacking both type I IFN receptor subunit 1 (Ifnar1) and Usp18 (Ifnar1/Usp18 double knockout mice) are viable and do not display a hydrocephalus phenotype. Moreover, we observed that suppression of USP18 in ependymal cells treated with IFN significantly increased cell death, including pyroptosis, and decreased proliferation. These findings suggest that heightened sensitivity to type I IFN during brain development contributes to the onset of hydrocephalus. Furthermore, they imply that inhibition of IFN signaling may hold promise as a therapeutic strategy for hydrocephalus.


Asunto(s)
Hidrocefalia , Interferón Tipo I , Ratones Noqueados , Receptor de Interferón alfa y beta , Ubiquitina Tiolesterasa , Animales , Hidrocefalia/genética , Hidrocefalia/patología , Interferón Tipo I/metabolismo , Ratones , Ubiquitina Tiolesterasa/genética , Ubiquitina Tiolesterasa/metabolismo , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/metabolismo , Ratones Endogámicos C57BL , Transducción de Señal/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Encéfalo/efectos de los fármacos , Epéndimo/metabolismo , Proliferación Celular/efectos de los fármacos , Piroptosis/efectos de los fármacos , Piroptosis/fisiología
6.
STAR Protoc ; 5(3): 103139, 2024 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-38878286

RESUMEN

Flow cytometry, single-cell RNA sequencing, and other analyses enable us to capture immune profiles of the tumor microenvironment. Here, we present a protocol to characterize the immune profile of tumor-bearing mice. We describe steps for establishing mouse models and preparing single-cell suspensions from tumor tissue and other immune-related organs, which can be further analyzed by flow cytometry and other omics assays. We then detail procedures for staining, flow cytometry analysis, and phenotyping of the immune cell populations. For complete details on the use and execution of this protocol, please refer to Miyauchi et al.1.

7.
Front Immunol ; 15: 1390263, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38799433

RESUMEN

Cancer immunotherapy, such as immune checkpoint blockade (ICB), has emerged as a groundbreaking approach for effective cancer treatment. Despite its considerable potential, clinical studies have indicated that the current response rate to cancer immunotherapy is suboptimal, primarily attributed to low immunogenicity in certain types of malignant tumors. Immunogenic cell death (ICD) represents a form of regulated cell death (RCD) capable of enhancing tumor immunogenicity and activating tumor-specific innate and adaptive immune responses in immunocompetent hosts. Therefore, gaining a deeper understanding of ICD and its evolution is crucial for developing more effective cancer therapeutic strategies. This review focuses exclusively on both historical and recent discoveries related to ICD modes and their mechanistic insights, particularly within the context of cancer immunotherapy. Our recent findings are also highlighted, revealing a mode of ICD induction facilitated by atypical interferon (IFN)-stimulated genes (ISGs), including polo-like kinase 2 (PLK2), during hyperactive type I IFN signaling. The review concludes by discussing the therapeutic potential of ICD, with special attention to its relevance in both preclinical and clinical settings within the field of cancer immunotherapy.


Asunto(s)
Muerte Celular Inmunogénica , Inmunoterapia , Neoplasias , Humanos , Neoplasias/terapia , Neoplasias/inmunología , Inmunoterapia/métodos , Muerte Celular Inmunogénica/efectos de los fármacos , Animales , Transducción de Señal , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inhibidores de Puntos de Control Inmunológico/farmacología
8.
Blood Adv ; 8(10): 2410-2423, 2024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38513139

RESUMEN

ABSTRACT: The transcription factor RUNX1 is a master regulator of hematopoiesis and is frequently mutated in myeloid malignancies. Mutations in its runt homology domain (RHD) frequently disrupt DNA binding and result in loss of RUNX1 function. However, it is not clearly understood how other RUNX1 mutations contribute to disease development. Here, we characterized RUNX1 mutations outside of the RHD. Our analysis of the patient data sets revealed that mutations within the C-terminus frequently occur in hematopoietic disorders. Remarkably, most of these mutations were nonsense or frameshift mutations and were predicted to be exempt from nonsense-mediated messenger RNA decay. Therefore, this class of mutation is projected to produce DNA-binding proteins that contribute to the pathogenesis in a distinct manner. To model this, we introduced the RUNX1R320∗ mutation into the endogenous gene locus and demonstrated the production of RUNX1R320∗ protein. Expression of RUNX1R320∗ resulted in the disruption of RUNX1 regulated processes such as megakaryocytic differentiation, through a transcriptional signature different from RUNX1 depletion. To understand the underlying mechanisms, we used Global RNA Interactions with DNA by deep sequencing (GRID-seq) to examine enhancer-promoter connections. We identified widespread alterations in the enhancer-promoter networks within RUNX1 mutant cells. Additionally, we uncovered enrichment of RUNX1R320∗ and FOXK2 binding at the MYC super enhancer locus, significantly upregulating MYC transcription and signaling pathways. Together, our study demonstrated that most RUNX1 mutations outside the DNA-binding domain are not subject to nonsense-mediated decay, producing protein products that act in concert with additional cofactors to dysregulate hematopoiesis through mechanisms distinct from those induced by RUNX1 depletion.


Asunto(s)
Diferenciación Celular , Subunidad alfa 2 del Factor de Unión al Sitio Principal , Mutación , Regiones Promotoras Genéticas , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Humanos , Diferenciación Celular/genética , Elementos de Facilitación Genéticos , Células Sanguíneas/metabolismo , Redes Reguladoras de Genes , Regulación de la Expresión Génica
9.
Proc Natl Acad Sci U S A ; 107(36): 15856-61, 2010 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-20724660

RESUMEN

The rapid induction of type I IFN is a central event of the innate defense against viral infections and is tightly regulated by a number of cellular molecules. Viral components induce strong type I IFN responses through the activation of toll-like receptors (TLRs) and intracellular cytoplasmic receptors such as an RNA helicase RIG-I and/or MDA5. According to recent studies, the NF-kappaB essential modulator (NEMO, also called IKKgamma) is crucial for this virus-induced antiviral response. However, the precise roles of signal activation by NEMO adaptor have not been elucidated. Here, we show that virus-induced IRF3 and NF-kappaB activation depends on the K(lys)-27-linked polyubiquitination to NEMO by the novel ubiquitin E3 ligase triparite motif protein 23 (TRIM23). Virus-induced IRF3 and NF-kappaB activation, as well as K27-linked NEMO polyubiquitination, were abrogated in TRIM23 knockdown cells, whereas TRIM23 knockdown had no effect on TNFalpha-mediated NF-kappaB activation. Furthermore, in NEMO-deficient mouse embryo fibroblast cells, IFN-stimulated response element-driven reporter activity was restored by ectopic expression of WT NEMO, as expected, but only partial recovery by NEMO K165/309/325/326/344R multipoints mutant on which TRIM23-mediated ubiquitin conjugation was substantially reduced. Thus, we conclude that TRIM23-mediated ubiquitin conjugation to NEMO is essential for TLR3- and RIG-I/MDA5-mediated antiviral innate and inflammatory responses.


Asunto(s)
Proteínas de Unión al GTP/metabolismo , Quinasa I-kappa B/metabolismo , Poliubiquitina/metabolismo , Línea Celular , Técnicas de Silenciamiento del Gen , Humanos
10.
Cell Rep ; 42(12): 113560, 2023 12 26.
Artículo en Inglés | MEDLINE | ID: mdl-38100351

RESUMEN

Tumor-associated myeloid cells modulate the tumor microenvironment and affect tumor progression. Type I interferon (IFN-I) has multiple effects on tumors and immune response, and ubiquitin-specific peptidase 18 (USP18) functions as a negative regulator of IFN-I signal transduction. This study aims to examine the function of IFN-I in myeloid cells during tumor progression. Here, we show that deletion of USP18 in myeloid cells suppresses tumor progression. Enhanced IFN-I signaling and blocked USP18 expression prompt downregulation of colony stimulating factor 1 receptor (CSF1R) and polarization of tumor-associated macrophages toward pro-inflammatory phenotypes. Further in vitro experiments reveal that downregulation of CSF1R is mediated by ubiquitin-proteasome degradation via E3 ligase neural precursor cell-expressed, developmentaly downregulated 4 (NEDD4) and the IFN-induced increase in ubiquitin E2 ubiquitin-conjugating enzyme H5. USP18 impairs ubiquitination and subsequent degradation of CSF1R by interrupting NEDD4 binding to CSF1R. These results reveal a previously unappreciated role of IFN-I in macrophage polarization by regulating CSF1R via USP18 and suggest targeting USP18 in myeloid-lineage cells as an effective strategy for IFN-based therapies.


Asunto(s)
Transducción de Señal , Macrófagos Asociados a Tumores , Proteínas Tirosina Quinasas Receptoras , Ubiquitina , Ubiquitinación
11.
Nat Commun ; 14(1): 251, 2023 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-36646704

RESUMEN

While immunotherapy has emerged as a breakthrough cancer therapy, it is only effective in some patients, indicating the need of alternative therapeutic strategies. Induction of cancer immunogenic cell death (ICD) is one promising way to elicit potent adaptive immune responses against tumor-associated antigens. Type I interferon (IFN) is well known to play important roles in different aspects of immune responses, including modulating ICD in anti-tumor action. However, how to expand IFN effect in promoting ICD responses has not been addressed. Here we show that depletion of ubiquitin specific protease 18 (USP18), a negative regulator of IFN signaling, selectively induces cancer cell ICD. Lower USP18 expression correlates with better survival across human selected cancer types and delays cancer progression in mouse models. Mechanistically, nuclear USP18 controls the enhancer landscape of cancer cells and diminishes STAT2-mediated transcription complex binding to IFN-responsive elements. Consequently, USP18 suppression not only enhances expression of canonical IFN-stimulated genes (ISGs), but also activates the expression of a set of atypical ISGs and NF-κB target genes, including genes such as Polo like kinase 2 (PLK2), that induce cancer pyroptosis. These findings may support the use of targeting USP18 as a potential cancer immunotherapy.


Asunto(s)
Interferón Tipo I , Neoplasias , Ratones , Animales , Humanos , Piroptosis , Pool de Genes , Transducción de Señal , FN-kappa B/metabolismo , Interferón Tipo I/genética , Ubiquitina Tiolesterasa/metabolismo , Neoplasias/genética
12.
Elife ; 92020 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-32945770

RESUMEN

Cells use molecular circuits to interpret and respond to extracellular cues, such as hormones and cytokines, which are often released in a temporally varying fashion. In this study, we combine microfluidics, time-lapse microscopy, and computational modeling to investigate how the type I interferon (IFN)-responsive regulatory network operates in single human cells to process repetitive IFN stimulation. We found that IFN-α pretreatments lead to opposite effects, priming versus desensitization, depending on input durations. These effects are governed by a regulatory network composed of a fast-acting positive feedback loop and a delayed negative feedback loop, mediated by upregulation of ubiquitin-specific peptidase 18 (USP18). We further revealed that USP18 upregulation can only be initiated at the G1/early S phases of cell cycle upon the treatment onset, resulting in heterogeneous and delayed induction kinetics in single cells. This cell cycle gating provides a temporal compartmentalization of feedback loops, enabling duration-dependent desensitization to repetitive stimulations.


Asunto(s)
Ciclo Celular/fisiología , Retroalimentación Fisiológica/fisiología , Interferón-alfa/metabolismo , Ciclo Celular/efectos de los fármacos , Retroalimentación Fisiológica/efectos de los fármacos , Células HEK293 , Células HeLa , Humanos , Interferón-alfa/farmacología , Cinética , Análisis de la Célula Individual , Ubiquitina Tiolesterasa/genética , Ubiquitina Tiolesterasa/metabolismo , Regulación hacia Arriba
13.
Blood Cancer J ; 10(2): 16, 2020 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-32029705

RESUMEN

Large-scale chromosomal translocations are frequent oncogenic drivers in acute myeloid leukemia (AML). These translocations often occur in critical transcriptional/epigenetic regulators and contribute to malignant cell growth through alteration of normal gene expression. Despite this knowledge, the specific gene expression alterations that contribute to the development of leukemia remain incompletely understood. Here, through characterization of transcriptional regulation by the RUNX1-ETO fusion protein, we have identified Ras-association domain family member 2 (RASSF2) as a critical gene that is aberrantly transcriptionally repressed in t(8;21)-associated AML. Re-expression of RASSF2 specifically inhibits t(8;21) AML development in multiple models. Through biochemical and functional studies, we demonstrate RASSF2-mediated functions to be dependent on interaction with Hippo kinases, MST1 and MST2, but independent of canonical Hippo pathway signaling. Using proximity-based biotin labeling we define the RASSF2-proximal proteome in leukemia cells and reveal association with Rac GTPase-related proteins, including an interaction with the guanine nucleotide exchange factor, DOCK2. Importantly, RASSF2 knockdown impairs Rac GTPase activation, and RASSF2 expression is broadly correlated with Rac-mediated signal transduction in AML patients. Together, these data reveal a previously unappreciated mechanistic link between RASSF2, Hippo kinases, and Rac activity with potentially broad functional consequences in leukemia.


Asunto(s)
Cromosomas Humanos Par 21/genética , Cromosomas Humanos Par 8/genética , Regulación Neoplásica de la Expresión Génica , Leucemia Mieloide Aguda/prevención & control , Proteínas de Fusión Oncogénica/metabolismo , Translocación Genética , Proteínas Supresoras de Tumor/metabolismo , Proteínas de Unión al GTP rac/metabolismo , Animales , Biomarcadores de Tumor/genética , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Ratones , Ratones Endogámicos C57BL , Proteínas de Fusión Oncogénica/genética , ARN Largo no Codificante , Células Tumorales Cultivadas , Proteínas Supresoras de Tumor/genética , Ensayos Antitumor por Modelo de Xenoinjerto , Proteínas de Unión al GTP rac/genética
14.
Cancer Discov ; 10(3): 382-393, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31974171

RESUMEN

Type I interferons (IFN), which activate many IFN-stimulated genes (ISG), are known to regulate tumorigenesis. However, little is known regarding how various ISGs coordinate with one another in developing antitumor effects. Here, we report that the ISG UBA7 is a tumor suppressor in breast cancer. UBA7 encodes an enzyme that catalyzes the covalent conjugation of the ubiquitin-like protein product of another ISG (ISG15) to cellular proteins in a process known as "ISGylation." ISGylation of other ISGs, including STAT1 and STAT2, synergistically facilitates production of chemokine-receptor ligands to attract cytotoxic T cells. These gene-activation events are further linked to clustering and nuclear relocalization of STAT1/2 within IFN-induced promyelocytic leukemia (PML) bodies. Importantly, this coordinated ISG-ISGylation network plays a central role in suppressing murine breast cancer growth and metastasis, which parallels improved survival in patients with breast cancer. These findings reveal a cooperative IFN-inducible gene network in orchestrating a tumor-suppressive microenvironment. SIGNIFICANCE: We report a highly cooperative ISG network, in which UBA7-mediated ISGylation facilitates clustering of transcription factors and activates an antitumor gene-expression program. These findings provide mechanistic insights into immune evasion in breast cancer associated with UBA7 loss, emphasizing the importance of a functional ISG-ISGylation network in tumor suppression.This article is highlighted in the In This Issue feature, p. 327.


Asunto(s)
Neoplasias de la Mama/genética , Interferón Tipo I/genética , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT2/genética , Enzimas Activadoras de Ubiquitina/genética , Animales , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/patología , Proliferación Celular/genética , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Redes Reguladoras de Genes/inmunología , Humanos , Ratones , Linfocitos T/inmunología , Factores de Transcripción/genética , Ubiquitinas/genética , Ubiquitinas/inmunología
15.
Mol Immunol ; 45(4): 1078-84, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17719635

RESUMEN

The RNA helicase retinoic inducible gene I (RIG-I) recognizes viral double-stranded RNA and initiates signaling cascades that lead to activation of the protein kinases IKKalphabeta, TBK1 and IKKepsilon, and to subsequent activation of the transcription factors NF-kappaB and IRF3. We recently reported that RIG-I was ubiquitinated by RNF125, an ubiquitin E3 ligase, leading to proteasomal degradation. RIG-I is also reported to be ISGylated by an unidentified ISG15 (IFN-stimulated gene, 15kDa) E3 ligase. UbcH8, an ubiquitin E2 conjugating enzyme, was shown to be involved in RIG-I ISGylation. Here, we found that UbcH8 suppressed RIG-I ubiquitination by RNF125, and this suppression was relieved by ectopic expression of ISG15. Alternately, ISG15 conjugation to RIG-I was suppressed by RNF125. By analyzing this regulatory circuit, we found that UbcH8 and ISG15 are functional regulators of RNF125 E3 ligase activity, which regulates the level of ubiquitin and ISG15 conjugation of RIG-I.


Asunto(s)
ARN Helicasas DEAD-box/metabolismo , Enzimas Ubiquitina-Conjugadoras/metabolismo , Ubiquitina/metabolismo , Línea Celular , Clonación Molecular , ARN Helicasas DEAD-box/genética , Humanos , Unión Proteica , Ubiquitina/genética , Enzimas Ubiquitina-Conjugadoras/genética
16.
Uirusu ; 58(1): 47-54, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19122388

RESUMEN

As a defense mechanism against infection, host cells have evolved sensor molecules which detect pathogen components directly and induce protective responses against the infection. TLRs, well known receptors, recognize a pathogen on the surface of cells or endosome/lysosome. Many pathogens penetrate into cytoplasm, in where non-TLR sensors recognize pathogen components including double-stranded RNA (dsRNA). On the downstream of each sensor, a variety of functional signaling molecules are activated to produce various cytokines upon the microbial invasion to induce host defense responses. Because that cytokines produced to regulate the host defense responses are known to affect cell proliferation also, the level of these molecules are needed to be controlled tightly, which means requisites of negative regulation of the signaling activated by pathogen after the completion of proper immune responses. Recent studies suggest important roles of some ubiquitin systems in this regulation. Here we focus, in particular, ubiquitin conjugation to signaling molecules by virus activation and like to show how ubiquitin signaling plays roles in this regulation by introducing some recent works.


Asunto(s)
Transducción de Señal/fisiología , Receptores Toll-Like/inmunología , Ubiquitina/fisiología , Ubiquitinación/fisiología , Virus/inmunología , Animales , Citocinas/fisiología , Humanos , Señales de Clasificación de Proteína/fisiología , ARN Bicatenario/inmunología , Transducción de Señal/inmunología , Ubiquitina/metabolismo , Activación Viral
17.
J Leukoc Biol ; 2018 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-29357192

RESUMEN

Type I IFNs (α, ß, and others) are a family of cytokines that are produced in physiological conditions as well as in response to the activation of pattern recognition receptors. They are critically important in controlling the host innate and adaptive immune response to viral and some bacterial infections, cancer, and other inflammatory stimuli. However, dysregulation of type I IFN production or response can contribute to immune pathologies termed "interferonopathies", pointing to the importance of balanced activating signals with tightly regulated mechanisms of tuning this signaling. Here, we summarize the recent advances of how type I IFN production and response are controlled at multiple levels of the type I IFN signaling cascade.

18.
Sci Immunol ; 3(27)2018 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-30266866

RESUMEN

Type I interferon (IFN-I) signaling paradoxically impairs host immune responses during many primary and secondary bacterial infections. Lack of IFN-I receptor reduces bacterial replication and/or bacterial persistence during infection with several bacteria. However, the mechanisms that mediate the adverse IFN-I effect are incompletely understood. Here, we show that Usp18, an interferon-stimulated gene that negatively regulates IFN-I signaling, is primarily responsible for the deleterious effect of IFN-I signaling during infection of mice with Listeria monocytogenes or Staphylococcus aureus Mechanistically, USP18 promoted bacterial replication by inhibiting antibacterial tumor necrosis factor-α (TNF-α) signaling. Deleting IFNAR1 or USP18 in CD11c-Cre+ cells similarly reduced bacterial titers in multiple organs and enhanced survival. Our results demonstrate that inhibiting USP18 function can promote control of primary and secondary bacterial infection by enhancing the antibacterial effect of TNF-α, which correlates with induction of reactive oxygen species (ROS). These findings suggest that USP18 could be targeted therapeutically in patients to ameliorate disease caused by serious bacterial infections.


Asunto(s)
Interferón Tipo I/inmunología , Listeriosis/inmunología , Infecciones Estafilocócicas/inmunología , Ubiquitina Tiolesterasa/inmunología , Animales , Femenino , Listeria monocytogenes , Masculino , Ratones Transgénicos , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/inmunología , Transducción de Señal , Staphylococcus aureus , Factor de Necrosis Tumoral alfa/inmunología , Ubiquitina Tiolesterasa/genética
19.
SLAS Discov ; 23(9): 960-973, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29751735

RESUMEN

Vaccines are reliant on adjuvants to enhance the immune stimulus, and type I interferons (IFNs) have been shown to be beneficial in augmenting this response. We were interested in identifying compounds that would sustain activation of an endogenous type I IFN response as a co-adjuvant. We began with generation of a human monocytic THP-1 cell line with an IFN-stimulated response element (ISRE)-ß-lactamase reporter construct for high-throughput screening. Pilot studies were performed to optimize the parameters and conditions for this cell-based Förster resonance energy transfer (FRET) reporter assay for sustaining an IFN-α-induced ISRE activation signal. These conditions were confirmed in an initial pilot screen, followed by the main screen for evaluating prolongation of an IFN-α-induced ISRE activation signal at 16 h. Hit compounds were identified using a structure enrichment strategy based on chemoinformatic clustering and a naïve "Top X" approach. A select list of confirmed hits was then evaluated for toxicity and the ability to sustain IFN activity by gene and protein expression. Finally, for proof of concept, a panel of compounds was used to immunize mice as co-adjuvant with a model antigen and an IFN-inducing Toll-like receptor 4 agonist, lipopolysaccharide, as an adjuvant. Selected compounds significantly augmented antigen-specific immunoglobulin responses.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Descubrimiento de Drogas , Interferón Tipo I/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Línea Celular , Supervivencia Celular , Relación Dosis-Respuesta a Droga , Descubrimiento de Drogas/métodos , Genes Reporteros , Ensayos Analíticos de Alto Rendimiento , Humanos , Ratones , Flujo de Trabajo
20.
Nat Struct Mol Biol ; 24(3): 279-289, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28165510

RESUMEN

Type I interferons (IFNs) are multifunctional cytokines that regulate immune responses and cellular functions but also can have detrimental effects on human health. A tight regulatory network therefore controls IFN signaling, which in turn may interfere with medical interventions. The JAK-STAT signaling pathway transmits the IFN extracellular signal to the nucleus, thus resulting in alterations in gene expression. STAT2 is a well-known essential and specific positive effector of type I IFN signaling. Here, we report that STAT2 is also a previously unrecognized, crucial component of the USP18-mediated negative-feedback control in both human and mouse cells. We found that STAT2 recruits USP18 to the type I IFN receptor subunit IFNAR2 via its constitutive membrane-distal STAT2-binding site. This mechanistic coupling of effector and negative-feedback functions of STAT2 may provide novel strategies for treatment of IFN-signaling-related human diseases.


Asunto(s)
Endopeptidasas/metabolismo , Interferón Tipo I/metabolismo , Factor de Transcripción STAT2/metabolismo , Transducción de Señal , Animales , Línea Celular Tumoral , Retroalimentación Fisiológica , Humanos , Immunoblotting , Ratones , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Unión Proteica , Dominios Proteicos , Receptor de Interferón alfa y beta/metabolismo , Factor de Transcripción STAT2/química , Técnicas del Sistema de Dos Híbridos , Ubiquitina Tiolesterasa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA