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










Base de dados
Intervalo de ano de publicação
1.
Discov Immunol ; 3(1): kyae003, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38567293

RESUMO

Basophils are the rarest leukocytes, but they have essential roles in protection against helminths, allergic disorders, autoimmune diseases, and some cancers. For years, the clinical significance of basophils has been neglected because of the lack of proper experimental tools to study them. The development of basophil-specific antibodies and animal models, along with genomic advances like single-cell transcriptomics, has greatly enhanced our understanding of basophil biology. Recent discoveries regarding basophils prompted us to write this review, emphasizing the basophil developmental pathway. In it, we chronologically examine the steps of basophil development in various species, which reveals the apparent advent of basophils predating IgE and basophil's IgE-independent regulatory role in primitive vertebrates. Then, we cover studies of basophil development in adult bone marrow, and compare those of murine and human basophils, introducing newly identified basophil progenitors and mature basophil subsets, as well as the transcription factors that regulate the transitions between them. Last, we discuss the heterogeneity of tissue-resident basophils, which may develop through extramedullary hematopoiesis. We expect that this review will contribute to a deeper understanding of basophil biology from the intricate aspects of basophil development and differentiation, offering valuable insights for both researchers and clinicians.

2.
J Allergy Clin Immunol ; 153(1): 132-145, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-37783432

RESUMO

BACKGROUND: Basophils are rare but important effector cells in many allergic disorders. Contrary to their early progenitors, the terminal developmental processes of basophils in which they gain their unique functional properties are unknown. OBJECTIVE: We sought to identify a novel late-stage basophil precursor and a transcription factor regulating the terminal maturation of basophils. METHODS: Using flow cytometry, transcriptome analysis, and functional assays, we investigated the identification and functionality of the basophil precursors as well as basophil development. We generated mice with basophil-specific deletion of nuclear factor IL-3 (NFIL3)/E4BP4 and analyzed the functional impairment of NFIL3/E4BP4-deficient basophils in vitro and in vivo using an oxazolone-induced murine model of allergic dermatitis. RESULTS: We report a new mitotic transitional basophil precursor population (referred to as transitional basophils) that expresses the FcεRIα chain at higher levels than mature basophils. Transitional basophils are less responsive to IgE-linked degranulation but produce more cytokines in response to IL-3, IL-33, or IgE cross-linking than mature basophils. In particular, we found that the expression of NFIL3/E4BP4 gradually rises as cells mature from the basophil progenitor stage. Basophil-specific deletion of NFIL3/E4BP4 reduces the expression of genes necessary for basophil function and impairs IgE receptor signaling, cytokine secretion, and degranulation in the context of murine atopic dermatitis. CONCLUSIONS: We discovered transitional basophils, a novel late-stage mitotic basophil precursor cell population that exists between basophil progenitors and postmitotic mature basophils. We demonstrated that NFIL3/E4BP4 augments the IgE-mediated functions of basophils, pointing to a potential therapeutic regulator for allergic diseases.


Assuntos
Fatores de Transcrição de Zíper de Leucina Básica , Basófilos , Animais , Camundongos , Basófilos/citologia , Basófilos/metabolismo , Dermatite Atópica/metabolismo , Hipersensibilidade/metabolismo , Imunoglobulina E/metabolismo , Interleucina-3/metabolismo , Fatores de Transcrição/metabolismo , Fatores de Transcrição de Zíper de Leucina Básica/metabolismo
3.
EMBO Rep ; 24(12): e57496, 2023 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-37916870

RESUMO

Stimulator of interferon genes (STING) is a core DNA sensing adaptor in innate immune signaling. STING activity is regulated by a variety of post-translational modifications (PTMs), including phosphorylation, ubiquitination, sumoylation, palmitoylation, and oxidation, as well as the balance between active and inactive polymer formation. It remains unclear, though, how different PTMs and higher order structures cooperate to regulate STING activity. Here, we report that the mitochondrial ubiquitin ligase MARCH5 (Membrane Associated Ring-CH-type Finger 5, also known as MITOL) ubiquitinates STING and enhances its activation. A long-term MARCH5 deficiency, in contrast, leads to the production of reactive oxygen species, which then facilitate the formation of inactive STING polymers by oxidizing mouse STING cysteine 205. We show that MARCH5-mediated ubiquitination of STING prevents the oxidation-induced STING polymer formation. Our findings highlight that MARCH5 balances STING ubiquitination and polymer formation and its control of STING activation is contingent on oxidative conditions.


Assuntos
Mitocôndrias , Ubiquitina-Proteína Ligases , Animais , Camundongos , Imunidade Inata , Mitocôndrias/metabolismo , Polímeros/metabolismo , Transdução de Sinais , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação
4.
Exp Mol Med ; 54(10): 1756-1765, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36229591

RESUMO

Clonal hematopoiesis of indeterminate potential (CHIP), a common aging-related process that predisposes individuals to various inflammatory responses, has been reported to be associated with COVID-19 severity. However, the immunological signature and the exact gene expression program by which the presence of CHIP exerts its clinical impact on COVID-19 remain to be elucidated. In this study, we generated a single-cell transcriptome landscape of severe COVID-19 according to the presence of CHIP using peripheral blood mononuclear cells. Patients with CHIP exhibited a potent IFN-γ response in exacerbating inflammation, particularly in classical monocytes, compared to patients without CHIP. To dissect the regulatory mechanism of CHIP (+)-specific IFN-γ response gene expression in severe COVID-19, we identified DNMT3A CHIP mutation-dependent differentially methylated regions (DMRs) and annotated their putative target genes based on long-range chromatin interactions. We revealed that CHIP mutant-driven hypo-DMRs at poised cis-regulatory elements appear to facilitate the CHIP (+)-specific IFN-γ-mediated inflammatory immune response. Our results highlight that the presence of CHIP may increase the susceptibility to hyperinflammation through the reorganization of chromatin architecture, establishing a novel subgroup of severe COVID-19 patients.


Assuntos
COVID-19 , Hematopoiese Clonal , Humanos , Transcriptoma , Hematopoese/genética , COVID-19/genética , Leucócitos Mononucleares , Mutação , Cromatina/genética , Perfilação da Expressão Gênica
5.
Small ; 18(17): e2106648, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35297560

RESUMO

Although the dendritic cell (DC)-based modulation of immune responses has emerged as a promising therapeutic strategy for tumors, infections, and autoimmune diseases, basic research and therapeutic applications of DCs are hampered by expensive growth factors and sophisticated culture procedures. Furthermore, the platform to drive the differentiation of a certain DC subset without any additional biochemical manipulations has not yet been developed. Here, five types of polymer films with different hydrophobicity via an initiated chemical vapor deposition (iCVD) process to modulate the interactions related to cell-substrate adhesion are introduced. Especially, poly(cyclohexyl methacrylate) (pCHMA) substantially enhances the expansion and differentiation of conventional type 1 DCs (cDC1s), the prime DC subset for antigen cross-presentation, and CD8+ T cell activation, by 4.8-fold compared to the conventional protocol. The cDC1s generated from the pCHMA-coated plates retain the bona fide DC functions including the expression of co-stimulatory molecules, cytokine secretion, antigen uptake and processing, T cell activation, and induction of antitumor immune responses. To the authors' knowledge, this is the first report highlighting that the modulation of surface hydrophobicity of the culture plate can be an incisive approach to construct an advanced DC culture platform with high efficiency, which potentially facilitates basic research and the development of immunotherapy employing DCs.


Assuntos
Células Dendríticas , Polímeros , Apresentação de Antígeno , Técnicas de Cultura de Células/métodos , Células Dendríticas/metabolismo , Ativação Linfocitária , Polímeros/metabolismo
6.
ACS Biomater Sci Eng ; 7(9): 4446-4453, 2021 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-34435775

RESUMO

Multiple sclerosis (MS) treatment via cytokine-mediated immunomodulation has been hampered by the difficulty with which cytokines can be stably and noninvasively delivered to the central nervous system. Here, we show that interleukin (IL)-13 packaged in extra-large-pore mesoporous silica nanoparticles (XL-MSNs) is protected from degradation and directs the alternative activation of macrophages both in vitro and in vivo. Furthermore, the noninvasive intranasal delivery of IL-13-loaded XL-MSNs ameliorated the symptoms of experimental autoimmune encephalomyelitis, a murine model of MS, accompanied by the induction of chemokines orchestrating immune cell infiltration. These results demonstrate the therapeutic potential of IL-13-loaded XL-MSNs for MS patients.


Assuntos
Encefalomielite Autoimune Experimental , Nanopartículas , Animais , Encefalomielite Autoimune Experimental/tratamento farmacológico , Humanos , Interleucina-13 , Macrófagos , Camundongos , Dióxido de Silício
7.
J Immunother Cancer ; 9(6)2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-34083422

RESUMO

BACKGROUND: Multiple types of immune cells producing IL-17 are found in the tumor microenvironment. However, their roles in tumor progression and exhaustion of CD8+ tumor-infiltrating lymphocytes (TILs) remain unclear. METHODS: To determine the role of type 17 immunity in tumor, we investigated the growth of B16F10 melanoma and the exhaustion of CD8+ TILs in Il17a-/- mice, Il17aCreR26DTA mice, RORγt inhibitor-treated mice, or their respective control mice. Adoptive transfer of tumor-specific IL-17-producing T cells was performed in B16F10-bearing congenic mice. Anti-CD4 or anti-Ly6G antibodies were used to deplete CD4+ T cells or CD11b+Gr-1hi myeloid cells in vivo, respectively. Correlation between type 17 immunity and T cell exhaustion in human cancer was evaluated by interrogating TCGA dataset. RESULTS: Depletion of CD4+ T cells promotes the exhaustion of CD8+ T cells with a concomitant increase in IL-17-producing CD8+ T (Tc17) cells in the tumor. Unlike IFN-γ-producing CD8+ T (Tc1) cells, tumor-infiltrating Tc17 cells exhibit CD103+KLRG1-IL-7Rαhi tissue resident memory-like phenotypes and are poorly cytolytic. Adoptive transfer of IL-17-producing tumor-specific T cells increases, while depletion of IL-17-producing cells decreases, the frequency of PD-1hiTim3+TOX+ terminally exhausted CD8+ T cells in the tumor. Blockade of IL-17 or RORγt pathway inhibits exhaustion of CD8+ T cells and also delays tumor growth in vivo. Consistent with these results, human TCGA analyses reveal a strong positive correlation between type 17 and CD8+ T cell exhaustion signature gene sets in multiple cancers. CONCLUSION: IL-17-producing cells promote terminal exhaustion of CD8+ T cells and tumor progression in vivo, which can be reversed by blockade of IL-17 or RORγt pathway. These findings unveil a novel role for IL-17-producing cells as tumor-promoting cells facilitating CD8+ T cell exhaustion, and propose type 17 immunity as a promising target for cancer immunotherapy.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Deleção de Genes , Interleucina-17/genética , Melanoma Experimental/terapia , Transferência Adotiva , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/transplante , Linhagem Celular Tumoral , Feminino , Humanos , Masculino , Melanoma Experimental/genética , Melanoma Experimental/imunologia , Camundongos , Microambiente Tumoral
8.
Exp Mol Med ; 52(4): 643-657, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32284536

RESUMO

Chromosomal instability (CIN) in cancer cells has been reported to activate the cGAS-STING innate immunity pathway via micronuclei formation, thus affecting tumor immunity and tumor progression. However, adverse effects of the cGAS/STING pathway as they relate to CIN have not yet been investigated. We addressed this issue using knockdown and add-back approaches to analyze each component of the cGAS/STING/TBK1/IRF3 pathway, and we monitored the extent of CIN by measuring micronuclei formation after release from nocodazole-induced mitotic arrest. Interestingly, knockdown of cGAS (cyclic GMP-AMP synthase) along with induction of mitotic arrest in HeLa and U2OS cancer cells clearly resulted in increased micronuclei formation and chromosome missegregation. Knockdown of STING (stimulator of interferon genes), TBK1 (TANK-binding kinase-1), or IRF3 (interferon regulatory factor-3) also resulted in increased micronuclei formation. Moreover, transfection with cGAMP, the product of cGAS enzymatic activity, as well as add-back of cGAS WT (but not catalytic-dead mutant cGAS), or WT or constitutively active STING (but not an inactive STING mutant) rescued the micronuclei phenotype, demonstrating that all components of the cGAS/STING/TBK1/IRF3 pathway play a role in preventing CIN. Moreover, p21 levels were decreased in cGAS-, STING-, TBK1-, and IRF3-knockdown cells, which was accompanied by the precocious G2/M transition of cells and the enhanced micronuclei phenotype. Overexpression of p21 or inhibition of CDK1 in cGAS-depleted cells reduced micronuclei formation and abrogated the precocious G2/M transition, indicating that the decrease in p21 and the subsequent precocious G2/M transition is the main mechanism underlying the induction of CIN through disruption of cGAS/STING signaling.


Assuntos
Instabilidade Cromossômica , Imunidade Inata , Fator Regulador 3 de Interferon/metabolismo , Proteínas de Membrana/metabolismo , Nucleotidiltransferases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais , Linhagem Celular Tumoral , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Ativação Enzimática , Expressão Gênica , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Proteínas de Membrana/genética , Micronúcleos com Defeito Cromossômico , Nucleotidiltransferases/genética
9.
Sci Rep ; 10(1): 718, 2020 01 20.
Artigo em Inglês | MEDLINE | ID: mdl-31959808

RESUMO

Quantitative and reliable measurement of cellular invasion is important to understand a range of biological processes such as cancer metastasis and angiogenesis. Spheroid invasion assays are an attractive in vitro platform because they effectively mimic the tumor cell invasion of solid tissues. Here, we developed an image analysis-based method to quantify the invasiveness of HT1080 human fibrosarcoma tumor cell spheroids. We segmented a cell-covered area into three subareas using objectively set threshold pixel intensities and calculated invasion indices using these subareas. Comparison with conventional parameters for spheroid invasion assays, such as area, length, and detached cells, showed that our indices present the invasion event at an early time and without being convoluted by proliferation. As an application, we then examined paracrine interactions between LLC1 mouse lung carcinoma cells and Raw264.7 mouse macrophage cells with our developed analysis method. We found that the invasion of tumor spheroids was increased by a macrophage-conditioned medium, concomitantly with a decrease in tumor cell proliferation. Importantly, invasion was further enhanced by a conditioned medium from activated macrophages by co-culture with tumor cells. Thus, our indices reveal that tumor cell invasion is facilitated in a feed-forward manner by communication between tumor cells and macrophages in the tumor microenvironment.


Assuntos
Fibrossarcoma/patologia , Macrófagos/patologia , Invasividade Neoplásica/patologia , Microambiente Tumoral , Animais , Linhagem Celular Tumoral , Proliferação de Células , Técnicas de Cocultura , Humanos , Camundongos , Células RAW 264.7 , Esferoides Celulares/patologia
10.
J Microbiol ; 58(1): 46-53, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31768937

RESUMO

The Chikungunya virus (CHIKV) belongs to the Alphavirus genus of Togaviridae family and contains a positive-sense single stranded RNA genome. Infection by this virus mainly causes sudden high fever, rashes, headache, and severe joint pain that can last for several months or years. CHIKV, a mosquito-borne arbovirus, is considered a re-emerging pathogen that has become one of the most pressing global health concerns due to a rapid increase in epidemics. Because handling of CHIKV is restricted to Biosafety Level 3 (BSL-3) facilities, the evaluation of prophylactic vaccines or antivirals has been substantially hampered. In this study, we first iden-tified the whole structural polyprotein sequence of a CHIKV strain isolated in South Korea (KNIH/2009/77). Phylogenetic analysis showed that this sequence clustered within the East/ Central/South African CHIKV genotype. Using this sequence information, we constructed a CHIKV-pseudotyped lenti-virus expressing the structural polyprotein of the Korean CHIKV isolate (CHIKVpseudo) and dual reporter genes of green fluorescence protein and luciferase. We then developed a pseudovirus-based neutralization assay (PBNA) using CHIKVpseudo. Results from this assay compared to those from the conventional plaque reduction neutralization test showed that our PBNA was a reliable and rapid method to evaluate the efficacy of neutralizing antibodies. More importantly, the neutralizing activities of human sera from CHIKV-infected individuals were quantitated by PBNA using CHIKVpseudo. Taken together, these results suggest that our PBNA for CHIKV may serve as a useful and safe method for testing the neutralizing activity of antibodies against CHIKV in BSL-2 facilities.


Assuntos
Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Febre de Chikungunya , Vírus Chikungunya/imunologia , Testes de Neutralização/métodos , Febre de Chikungunya/imunologia , Febre de Chikungunya/virologia , Vírus Chikungunya/isolamento & purificação , Humanos , República da Coreia
11.
RNA Biol ; 17(3): 325-334, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31852354

RESUMO

Retinoic acid-inducible gene I (RIG-I) is responsible for innate immunity via the recognition of short double-stranded RNAs in the cytosol. With the clue that G-U wobble base pairs in the influenza A virus's RNA promoter region are responsible for RIG-I activation, we determined the complex structure of RIG-I ΔCARD and a short hairpin RNA with G-U wobble base pairs by X-ray crystallography. Interestingly, the overall helical backbone trace was not affected by the presence of the wobble base pairs; however, the base pair inclination and helical axis angle changed upon RIG-I binding. NMR spectroscopy revealed that RIG-I binding renders the flexible base pair of the influenza A virus's RNA promoter region between the two G-U wobble base pairs even more flexible. Binding to RNA with wobble base pairs resulted in a more flexible RIG-I complex. This flexible complex formation correlates with the entropy-favoured binding of RIG-I and RNA, which results in tighter binding affinity and RIG-I activation. This study suggests that the structure and dynamics of RIG-I are tailored to the binding of specific RNA sequences with different flexibility.


Assuntos
Proteína DEAD-box 58/química , Proteína DEAD-box 58/metabolismo , RNA de Cadeia Dupla/química , RNA de Cadeia Dupla/metabolismo , Receptores Imunológicos/química , Receptores Imunológicos/metabolismo , Pareamento de Bases , Cristalografia por Raios X , Entropia , Células HEK293 , Humanos , Hidrogênio/química , Interferon gama/metabolismo , Espectroscopia de Ressonância Magnética , Modelos Moleculares , Prótons
12.
Mol Cells ; 41(11): 953-963, 2018 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-30396239

RESUMO

The stepwise development of T cells from a multipotent precursor is guided by diverse mechanisms, including interactions among lineage-specific transcription factors (TFs) and epigenetic changes, such as DNA methylation and hydroxymethylation, which play crucial roles in mammalian development and lineage commitment. To elucidate the transcriptional networks and epigenetic mechanisms underlying T-cell lineage commitment, we investigated genome-wide changes in gene expression, DNA methylation and hydroxymethylation among populations representing five successive stages of T-cell development (DN3, DN4, DP, CD4+, and CD8+) by performing RNA-seq, MBD-seq and hMeDIP-seq, respectively. The most significant changes in the transcriptomes and epigenomes occurred during the DN4 to DP transition. During the DP stage, many genes involved in chromatin modification were up-regulated and exhibited dramatic changes in DNA hydroxymethylation. We also observed 436 alternative splicing events, and approximately 57% (252) of these events occurred during the DP stage. Many stage-specific, differentially methylated regions were observed near the stage-specific, differentially expressed genes. The dynamic changes in DNA methylation and hydroxymethylation were associated with the recruitment of stage-specific TFs. We elucidated interactive networks comprising TFs, chromatin modifiers, and DNA methylation and hope that this study provides a framework for the understanding of the molecular networks underlying T-cell lineage commitment.


Assuntos
DNA/genética , Epigenômica , Redes Reguladoras de Genes , Linfócitos T/fisiologia , Transcriptoma , Processamento Alternativo , Animais , Diferenciação Celular , Linhagem da Célula , Metilação de DNA , Regulação da Expressão Gênica , Hematopoese , Humanos , Fatores de Transcrição/metabolismo
13.
Biochim Biophys Acta Mol Basis Dis ; 1864(10): 3145-3153, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30293565

RESUMO

BACKGROUND: Human ANKRD9 (ankyrin repeat domain 9) expression is altered in some cancers. METHODS: We tested genetic association of ANKRD9 with gastric cancer susceptibility and examined functional association of ANKRD9 with altered proliferation of MKN45 gastric cancer cells. We then identified ANKRD9-binding partners in HEK 293 embryonic kidney cells using quantitative proteomics, western blotting and complex reconstitution assays. We finally demonstrated ANKRD9's role of recognizing substrates for ubiquitination using in vitro ubiquitylation assay. RESULTS: ANKRD9 is associated with cancer susceptibility in a comparison of single-nucleotide polymorphisms between 1092 gastric cancer patients and 1206 healthy controls. ANKRD9 depletion accelerates tumor progression by increasing cellular proliferation, piling up, and anchorage-independent growth of MKN45 cells. We discovered that ANKRD9 is a ubiquitin ligase substrate receptor subunit and has an anti-proliferative activity. ANKRD9 associates with CUL5 (not CUL2), ELOB, ELOC, and presumably RNF7 subunits, which together assemble into a cullin-RING superfamily E3 ligase complex. ANKRD9 belongs to the ASB family of proteins, which are characterized by the presence of ankyrin repeats and a SOCS box. In addition to its interactions with the other E3 ligase subunits, ANKRD9 interacts with two isoforms of inosine monophosphate dehydrogenase (IMPDH). These IMPDH isoforms are cognate substrates of the ANKRD9-containing E3 enzyme, which ubiquitinates them for proteasomal degradation. Their ubiquitination and turnover require the presence of ANKRD9. CONCLUSION: ANKRD9, a previously unidentified E3 substrate receptor subunit, functions in tumor suppression by recognizing the oncoprotein IMPDH isoforms for E3 ubiquitination and proteasomal degradation.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Polimorfismo de Nucleotídeo Único , Neoplasias Gástricas/genética , Adulto , Idoso , Estudos de Casos e Controles , Linhagem Celular Tumoral , Proliferação de Células , Proteínas Culina/metabolismo , Progressão da Doença , Feminino , Estudos de Associação Genética , Predisposição Genética para Doença , Células HEK293 , Humanos , IMP Desidrogenase/metabolismo , Masculino , Pessoa de Meia-Idade , Proteólise , Proteômica , Neoplasias Gástricas/metabolismo , Proteínas Supressoras de Tumor
15.
Nucleic Acids Res ; 46(17): 9011-9026, 2018 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-30137501

RESUMO

DExD/H-box helicase 9 (DHX9), or RNA helicase A (RHA), is an abundant multifunctional nuclear protein. Although it was previously reported to act as a cytosolic DNA sensor in plasmacytoid dendritic cells (pDCs), the role and molecular mechanisms of action of DHX9 in cells that are not pDCs during DNA virus infection are not clear. Here, a macrophage-specific knockout and a fibroblast-specific knockdown of DHX9 impaired antiviral innate immunity against DNA viruses, leading to increased virus replication. DHX9 enhanced NF-κB-mediated transactivation in the nucleus, which required its ATPase-dependent helicase (ATPase/helicase) domain, but not the cytosolic DNA-sensing domain. In addition, DNA virus infection did not induce cytoplasmic translocation of nuclear DHX9 in macrophages and fibroblasts. Nuclear DHX9 was associated with a multiprotein complex including both NF-κB p65 and RNA polymerase II (RNAPII) in chromatin containing NF-κB-binding sites. DHX9 was essential for the recruitment of RNAPII rather than NF-κB p65, to the corresponding promoters; this function also required its ATPase/helicase activity. Taken together, our results show a critical role of nuclear DHX9 (as a transcription coactivator) in the stimulation of NF-κB-mediated innate immunity against DNA virus infection, independently of DHX9's DNA-sensing function.


Assuntos
RNA Helicases DEAD-box/genética , DNA Viral/genética , Interações Hospedeiro-Patógeno/genética , Imunidade Inata , NF-kappa B/genética , RNA Polimerase II/genética , Animais , Chlorocebus aethiops , RNA Helicases DEAD-box/deficiência , RNA Helicases DEAD-box/imunologia , DNA Viral/imunologia , Células Dendríticas/imunologia , Células Dendríticas/virologia , Feminino , Gammaherpesvirinae/genética , Gammaherpesvirinae/crescimento & desenvolvimento , Gammaherpesvirinae/imunologia , Regulação da Expressão Gênica , Células HEK293 , Células HeLa , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/crescimento & desenvolvimento , Herpesvirus Humano 1/imunologia , Interações Hospedeiro-Patógeno/imunologia , Humanos , Macrófagos/imunologia , Macrófagos/virologia , Masculino , Camundongos , Camundongos Transgênicos , Células-Tronco Embrionárias Murinas/imunologia , Células-Tronco Embrionárias Murinas/virologia , NF-kappa B/imunologia , Células NIH 3T3 , Cultura Primária de Células , RNA Polimerase II/imunologia , Transdução de Sinais , Células Vero , Replicação Viral
16.
PLoS One ; 13(7): e0200624, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29990347

RESUMO

[This corrects the article DOI: 10.1371/journal.pone.0126456.].

17.
Exp Mol Med ; 50(4): 1, 2018 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-29654313

RESUMO

The original version of this article unfortunately contained an error in the author name Dongchan Yang, which was incorrectly given as Dongchan Yang Sung.

18.
Biochem Biophys Res Commun ; 500(2): 497-503, 2018 06 02.
Artigo em Inglês | MEDLINE | ID: mdl-29673589

RESUMO

Stimulator of IFN genes (STING) is essential for the DNA-sensing innate immune pathway. Recently, evidence is emerging that suggests STING also plays important roles in autoimmunity, cancer therapy, and senescence. Although a multitude of post-translational modifications that regulate the STING pathway have been discovered, the cellular events that guide STING translocation remain unclear. Here, we show, paradoxically, that both BAPTA-AM-mediated calcium depletion and ionomycin-induced calcium elevation suppress STING translocation and STING-mediated IFN-ß production. We demonstrate that the mitochondria fission mediator DRP1 is crucial for ionomycin-induced inhibition of IFN-ß production. Furthermore, knockout of DRP1 suppressed ionomycin-induced increases in calcium as well as mitochondrial fragmentation. Collectively, our findings reveal that the induction of STING signaling is contingent on a fine-tuning of intracellular calcium levels.


Assuntos
Cálcio/metabolismo , Espaço Intracelular/metabolismo , Proteínas de Membrana/metabolismo , Transdução de Sinais , Animais , Dinaminas/deficiência , Dinaminas/metabolismo , Ácido Egtázico/análogos & derivados , Ácido Egtázico/farmacologia , Embrião de Mamíferos/citologia , Fibroblastos/metabolismo , Interferon beta/biossíntese , Ionomicina/farmacologia , Camundongos , Células RAW 264.7
19.
Exp Mol Med ; 50(3): e458, 2018 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-29546878

RESUMO

The spatiotemporal regulation of immune cells in lymph nodes (LNs) is crucial for mounting protective T-cell responses, which are orchestrated by dendritic cells (DCs). However, it is unclear how the DC subsets are altered by the inflammatory milieu of LNs. Here, we show that the inflamed LNs of Listeria-infected mice are characterized by the clustering of neutrophils and monocytes and IFN-γ production. Significantly, the early inflammatory responses are coupled with the differentiation of not one, but two types of CD64+CD11c+MHCII+ inflammatory DCs. Through the assessment of chemokine receptor dependency, gene expression profiles, growth factor requirements and DC-specific lineage mapping, we herein unveil a novel inflammatory DC population (we termed 'CD64+ cDCs') that arises from conventional DCs (cDCs), distinguishable from CD64+ monocyte-derived DCs (moDCs) in inflamed LNs. We determined that Listeria-induced type I IFN is a critical inflammatory cue for the development of CD64+ cDCs but not CD64+ moDCs. Importantly, CD64+ cDCs displayed a higher potential to activate T cells than CD64+ moDCs, whereas the latter showed more robust expression of inflammatory genes. Although CD64+ and CD64- cDCs were able to cross-present soluble antigens at a high dose to CD8+ T cells, CD64+ cDCs concentrated and cross-presented a minute amount of soluble antigens delivered via CD64 (FcγRI) as immune complexes. These findings reveal the role of early inflammatory responses in driving the differentiation of two inflammatory DC subsets empowered with distinct competencies.


Assuntos
Células Dendríticas/fisiologia , Inflamação/imunologia , Linfonodos/imunologia , Animais , Células Cultivadas , Células Dendríticas/metabolismo , Citometria de Fluxo , Imuno-Histoquímica , Inflamação/metabolismo , Linfonodos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...