Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Blood ; 143(3): 243-257, 2024 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-37922454

RESUMEN

ABSTRACT: Regulation of lineage biases in hematopoietic stem and progenitor cells (HSPCs) is pivotal for balanced hematopoietic output. However, little is known about the mechanism behind lineage choice in HSPCs. Here, we show that messenger RNA (mRNA) decay factors regnase-1 (Reg1; Zc3h12a) and regnase-3 (Reg3; Zc3h12c) are essential for determining lymphoid fate and restricting myeloid differentiation in HSPCs. Loss of Reg1 and Reg3 resulted in severe impairment of lymphopoiesis and a mild increase in myelopoiesis in the bone marrow. Single-cell RNA sequencing analysis revealed that Reg1 and Reg3 regulate lineage directions in HSPCs via the control of a set of myeloid-related genes. Reg1- and Reg3-mediated control of mRNA encoding Nfkbiz, a transcriptional and epigenetic regulator, was essential for balancing lymphoid/myeloid lineage output in HSPCs in vivo. Furthermore, single-cell assay for transposase-accessible chromatin sequencing analysis revealed that Reg1 and Reg3 control the epigenetic landscape on myeloid-related gene loci in early stage HSPCs via Nfkbiz. Consistently, an antisense oligonucleotide designed to inhibit Reg1- and Reg3-mediated Nfkbiz mRNA degradation primed hematopoietic stem cells toward myeloid lineages by enhancing Nfkbiz expression. Collectively, the collaboration between posttranscriptional control and chromatin remodeling by the Reg1/Reg3-Nfkbiz axis governs HSPC lineage biases, ultimately dictating the fate of lymphoid vs myeloid differentiation.


Asunto(s)
Médula Ósea , Células Madre Hematopoyéticas , Linaje de la Célula/genética , Células Madre Hematopoyéticas/metabolismo , Médula Ósea/metabolismo , Hematopoyesis/genética , ARN Mensajero/metabolismo , Diferenciación Celular/genética
2.
J Immunol ; 209(6): 1083-1094, 2022 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-35977797

RESUMEN

Asthma is more common in females than males after adolescence. However, the mechanism of the sex bias in the prevalence of asthma remains unknown. To test whether sex steroid hormones have some roles in T cells during development of asthma, we analyzed airway inflammation in T cell-specific androgen receptor (AR)- and estrogen receptor (ER)-deficient mice. T cell-specific AR-deficient male mice developed severer house dust mite-induced allergic airway inflammation than did control male mice, whereas T cell-specific ERα- and ERß-deficient female mice exhibited a similar degree of inflammation as for control female mice. Furthermore, administration of dihydrotestosterone reduced cytokine production of Th2 cells from control, but not AR-deficient, naive T cells. Transfer of OT-II transgenic AR-deficient Th2 cells into wild-type mice induced severer allergic airway inflammation by OVA than transfer of control Th2 cells. Gene expression profiling suggested that the expression of genes related with cell cycle and Th2 differentiation was elevated in AR-deficient Th2 cells, whereas expression of dual specificity phosphatase (DUSP)-2, a negative regulator of p38, was downregulated. In addition, a chromatin immunoprecipitation assay suggested that AR bound to an AR motif in the 5' untranslated region of the Dusp2 gene in Th2 cells. Furthermore, the Dusp2 promoter with a wild-type AR motif, but not a mutated motif, was transactivated by dihydrotestosterone in a reporter assay. Finally, forced expression of DUSP-2 by retrovirus vector reduced IL-4 expression in Th2 cells. Thus, these results suggest that androgen signaling suppresses cytokine production of Th2 cells by inducing DUSP-2, explaining, in part, the sex bias of asthma after adolescence.


Asunto(s)
Asma , Hipersensibilidad , Regiones no Traducidas 5' , Andrógenos/metabolismo , Animales , Asma/genética , Asma/metabolismo , Dihidrotestosterona , Modelos Animales de Enfermedad , Fosfatasas de Especificidad Dual/metabolismo , Receptor alfa de Estrógeno/genética , Receptor beta de Estrógeno/genética , Femenino , Hipersensibilidad/metabolismo , Inflamación/metabolismo , Interleucina-4/genética , Interleucina-4/metabolismo , Masculino , Ratones , Ratones Noqueados , Receptores Androgénicos/genética , Receptores de Estrógenos/genética , Células Th17/metabolismo , Células Th2/metabolismo
3.
Circulation ; 146(13): 1006-1022, 2022 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-35997026

RESUMEN

BACKGROUND: Pulmonary arterial hypertension (PAH) is a type of pulmonary hypertension (PH) characterized by obliterative pulmonary vascular remodeling, resulting in right-sided heart failure. Although the pathogenesis of PAH is not fully understood, inflammatory responses and cytokines have been shown to be associated with PAH, in particular, with connective tissue disease-PAH. In this sense, Regnase-1, an RNase that regulates mRNAs encoding genes related to immune reactions, was investigated in relation to the pathogenesis of PH. METHODS: We first examined the expression levels of ZC3H12A (encoding Regnase-1) in peripheral blood mononuclear cells from patients with PH classified under various types of PH, searching for an association between the ZC3H12A expression and clinical features. We then generated mice lacking Regnase-1 in myeloid cells, including alveolar macrophages, and examined right ventricular systolic pressures and histological changes in the lung. We further performed a comprehensive analysis of the transcriptome of alveolar macrophages and pulmonary arteries to identify genes regulated by Regnase-1 in alveolar macrophages. RESULTS: ZC3H12A expression in peripheral blood mononuclear cells was inversely correlated with the prognosis and severity of disease in patients with PH, in particular, in connective tissue disease-PAH. The critical role of Regnase-1 in controlling PAH was also reinforced by the analysis of mice lacking Regnase-1 in alveolar macrophages. These mice spontaneously developed severe PAH, characterized by the elevated right ventricular systolic pressures and irreversible pulmonary vascular remodeling, which recapitulated the pathology of patients with PAH. Transcriptomic analysis of alveolar macrophages and pulmonary arteries of these PAH mice revealed that Il6, Il1b, and Pdgfa/b are potential targets of Regnase-1 in alveolar macrophages in the regulation of PAH. The inhibition of IL-6 (interleukin-6) by an anti-IL-6 receptor antibody or platelet-derived growth factor by imatinib but not IL-1ß (interleukin-1ß) by anakinra, ameliorated the pathogenesis of PAH. CONCLUSIONS: Regnase-1 maintains lung innate immune homeostasis through the control of IL-6 and platelet-derived growth factor in alveolar macrophages, thereby suppressing the development of PAH in mice. Furthermore, the decreased expression of Regnase-1 in various types of PH implies its involvement in PH pathogenesis and may serve as a disease biomarker, and a therapeutic target for PH as well.


Asunto(s)
Hipertensión Pulmonar , Hipertensión Arterial Pulmonar , Animales , Biomarcadores , Citocinas , Hipertensión Pulmonar Primaria Familiar , Hipertensión Pulmonar/metabolismo , Mesilato de Imatinib , Proteína Antagonista del Receptor de Interleucina 1 , Interleucina-1beta , Interleucina-6/genética , Interleucina-6/metabolismo , Leucocitos Mononucleares/metabolismo , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/patología , Ratones , Factor de Crecimiento Derivado de Plaquetas , Arteria Pulmonar , Estabilidad del ARN , Ribonucleasas/genética , Ribonucleasas/metabolismo , Remodelación Vascular
4.
Sci Transl Med ; 14(644): eabo2137, 2022 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-35544597

RESUMEN

Regnase-1 is an ribonuclease that plays essential roles in restricting inflammation through degrading messenger RNAs (mRNAs) involved in immune reactions via the recognition of stem-loop (SL) structures in the 3' untranslated regions (3'UTRs). Dysregulated expression of Regnase-1 is associated with the pathogenesis of inflammatory and autoimmune diseases in mice and humans. Here, we developed a therapeutic strategy to suppress inflammatory responses by blocking Regnase-1 self-regulation, which was mediated by the simultaneous use of two antisense phosphorodiamidate morpholino oligonucleotides (MOs) to alter the binding of Regnase-1 toward the SL structures in its 3'UTR. Regnase-1-targeting MOs not only enhanced Regnase-1 expression by stabilizing mRNAs but also effectively reduced the expression of multiple proinflammatory transcripts that were controlled by Regnase-1 in macrophages. Intratracheal administration of Regnase-1-targeting MOs ameliorated acute respiratory distress syndrome and chronic fibrosis through suppression of inflammatory cascades. In addition, intracranial treatment with Regnase-1-targeting MOs attenuated the development of experimental autoimmune encephalomyelitis by promoting the expansion of homeostatic microglia and regulatory T cell populations. Regnase-1 expression was inversely correlated with disease severity in patients with multiple sclerosis, and MOs targeting human Regnase-1 SL structures were effective in mitigating cytokine production in human immune cells. Collectively, MO-mediated disruption of the Regnase-1 self-regulation pathway is a potential therapeutic strategy to enhance Regnase-1 abundance, which, in turn, provides therapeutic benefits for treating inflammatory diseases by suppressing inflammation.


Asunto(s)
Enfermedades Autoinmunes , Oligonucleótidos Antisentido , Regiones no Traducidas 3'/genética , Animales , Endorribonucleasas , Humanos , Inflamación , Ratones , Oligonucleótidos Antisentido/farmacología , Oligonucleótidos Antisentido/uso terapéutico , ARN Mensajero/genética , ARN Mensajero/metabolismo
5.
Sci Signal ; 15(729): eabm5011, 2022 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-35412849

RESUMEN

Toll-like receptor (TLR) stimulation induces glycolysis and the production of mitochondrial reactive oxygen species (ROS), both of which are critical for inflammatory responses in macrophages. Here, we demonstrated that cyclin J, a TLR-inducible member of the cyclin family, reduced cytokine production in macrophages by coordinately controlling glycolysis and mitochondrial functions. Cyclin J interacted with cyclin-dependent kinases (CDKs), which increased the phosphorylation of a subset of CDK substrates, including the transcription factor FoxK1 and the GTPase Drp1. Cyclin J-dependent phosphorylation of FoxK1 decreased the transcription of glycolytic genes and Hif-1α activation, whereas hyperactivation of Drp1 by cyclin J-dependent phosphorylation promoted mitochondrial fragmentation and impaired the production of mitochondrial ROS. In mice, cyclin J in macrophages limited the growth of tumor xenografts and protected against LPS-induced shock but increased the susceptibility to bacterial infection. Collectively, our findings indicate that cyclin J-CDK signaling promotes antitumor immunity and the resolution of inflammation by opposing the metabolic changes that drive inflammatory responses in macrophages.


Asunto(s)
Inmunidad Innata , Macrófagos , Animales , Quinasas Ciclina-Dependientes/genética , Quinasas Ciclina-Dependientes/metabolismo , Ciclinas/metabolismo , Factores de Transcripción Forkhead/metabolismo , Humanos , Macrófagos/metabolismo , Ratones , Especies Reactivas de Oxígeno/metabolismo , Receptores Toll-Like/genética , Receptores Toll-Like/metabolismo
6.
Int Immunol ; 33(12): 859-865, 2021 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-34320195

RESUMEN

Regulation of messenger RNA (mRNA) decay plays a crucial role in the control of gene expression. Canonical mRNA decay pathways are initiated by deadenylation and decapping and are followed by exonucleolytic degradation. However, recent studies revealed that endoribonucleolytic cleavage also mediates mRNA decay, and both exoribonucleolytic and endoribonucleolytic decay pathways are important for the regulation of immune responses. Regnase-1 functions as an endoribonuclease to control immunity by damping mRNAs. Particularly, Regnase-1 controls cytokines and other inflammatory mediators by recognizing their mRNAs via stem-loop structures present in the 3' untranslated regions. Regnase-1 was found to be critical for human inflammatory diseases such as ulcerative colitis and idiopathic pulmonary fibrosis. Furthermore, a set of Regnase-1-related RNases contribute to immune regulation as well as antiviral host defense. In this review, we provide an overview of recent findings as to immune-related RNA-binding proteins (RBPs) with an emphasis on stem-loop-mediated mRNA decay via Regnase-1 and related RNases and discuss how the function of these RBPs is regulated and contributes to inflammatory disorders.


Asunto(s)
Ribonucleasas/inmunología , Factores de Transcripción/inmunología , Colitis Ulcerosa/inmunología , Humanos , Fibrosis Pulmonar Idiopática/inmunología , Procesamiento Postranscripcional del ARN
7.
Nucleic Acids Res ; 49(9): 5369-5381, 2021 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-33950203

RESUMEN

The CCCH-type zinc finger (ZnF) containing ZC3H12 ribonucleases are crucial in post-transcriptional immune homoeostasis with ZC3H12A being the only structurally studied member of the family. In this study, we present a structural-biochemical characterization of ZC3H12C, which is linked with chronic immune disorders like psoriasis. We established that the RNA substrate is cooperatively recognized by the PIN and ZnF domains of ZC3H12C and analyzed the crystal structure of ZC3H12C bound to a single-stranded RNA substrate. The RNA engages in hydrogen-bonded contacts and stacking interactions with the PIN and ZnF domains simultaneously. The ZC3H12 ZnF shows unprecedented structural features not previously observed in any member of the CCCH-ZnF family and utilizes stacking interactions via a unique combination of spatially conserved aromatic residues to align the target transcript in a bent conformation onto the ZnF scaffold. Further comparative structural analysis of ZC3H12 CCCH-ZnF suggests that a trinucleotide sequence is recognized by ZC3H12 ZnF in target RNA. Our work not only describes the initial structure-biochemical study on ZC3H12C, but also provides the first molecular insight into RNA recognition by a ZC3H12 family member. Finally, our work points to an evolutionary code for RNA recognition adopted by CCCH-type ZnF proteins.


Asunto(s)
ARN/química , Ribonucleasas/química , Regiones no Traducidas 3' , Animales , Cristalografía por Rayos X , Células HEK293 , Humanos , Interleucina-6/genética , Interleucina-6/metabolismo , Magnesio , Ratones , Modelos Moleculares , Unión Proteica , Dominios Proteicos , ARN/metabolismo , Ribonucleasas/metabolismo , Dedos de Zinc
8.
Eur Respir J ; 57(3)2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32978308

RESUMEN

Regnase-1 is an RNase critical for post-transcriptional control of pulmonary immune homeostasis in mice by degrading immune-related mRNAs. However, little is known about the cell types Regnase-1 controls in the lung, and its relevance to human pulmonary diseases.Regnase-1-dependent changes in lung immune cell types were examined by a competitive bone marrow transfer mouse model, and group 2 innate lymphoid cells (ILC2s) were identified. Then the associations between Regnase-1 in ILC2s and human diseases were investigated by transcriptome analysis and a bleomycin-induced pulmonary fibrosis mouse model. The clinical significance of Regnase-1 in ILC2s was further assessed using patient-derived cells.Regnase-1-deficiency resulted in the spontaneous proliferation and activation of ILC2s in the lung. Intriguingly, genes associated with pulmonary fibrosis were highly upregulated in Regnase-1-deficient ILC2s compared with wild-type, and supplementation of Regnase-1-deficient ILC2s augmented bleomycin-induced pulmonary fibrosis in mice. Regnase-1 suppresses mRNAs encoding transcription factors Gata3 and Egr1, which are potent to regulate fibrosis-associated genes. Clinically, Regnase-1 protein levels in ILC2 negatively correlated with the ILC2 population in bronchoalveolar lavage fluid. Furthermore, idiopathic pulmonary fibrosis (IPF) patients with ILC2s >1500 cells·mL-1 peripheral blood exhibited poorer prognosis than patients with lower numbers, implying the contribution of Regnase-1 in ILC2s for the progression of IPF.Collectively, Regnase-1 was identified as a critical post-transcriptional regulator of the profibrotic function of ILC2s both in mouse and human, suggesting that Regnase-1 may be a novel therapeutic target for IPF.


Asunto(s)
Linfocitos , Fibrosis Pulmonar , Animales , Líquido del Lavado Bronquioalveolar , Humanos , Inmunidad Innata , Pulmón , Ratones , Ratones Noqueados , Fibrosis Pulmonar/inducido químicamente
9.
Sci Immunol ; 5(51)2020 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-32887843

RESUMEN

Cell type-specific gene expression is driven by the interplay between lineage-specific transcription factors and cis-regulatory elements to which they bind. Adaptive immunity relies on RAG-mediated assembly of T cell receptor (TCR) and immunoglobulin (Ig) genes. Although Rag1 and Rag2 expression is largely restricted to adaptive lymphoid lineage cells, it remains unclear how Rag gene expression is regulated in a cell lineage-specific manner. Here, we identified three distinct cis-regulatory elements, a T cell lineage-specific enhancer (R-TEn) and the two B cell-specific elements, R1B and R2B By generating mice lacking either R-TEn or R1B and R2B, we demonstrate that these distinct sets of regulatory elements drive the expression of Rag genes in developing T and B cells. What these elements have in common is their ability to bind the transcription factor E2A. By generating a mouse strain that carries a mutation within the E2A binding site of R-TEn, we demonstrate that recruitment of E2A to this site is essential for orchestrating changes in chromatin conformation that drive expression of Rag genes in T cells. By mapping cis-regulatory elements and generating multiple mouse strains lacking distinct enhancer elements, we demonstrate expression of Rag genes in developing T and B cells to be driven by distinct sets of E2A-dependent cis-regulatory modules.


Asunto(s)
Linfocitos B/inmunología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/inmunología , Proteínas de Unión al ADN/inmunología , Proteínas de Homeodominio/inmunología , Linfocitos T/inmunología , Animales , Animales Modificados Genéticamente , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Proteínas de Unión al ADN/genética , Femenino , Proteínas de Homeodominio/genética , Masculino , Ratones
10.
Cells ; 9(7)2020 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-32708595

RESUMEN

RNA acts as an immunostimulatory molecule in the innate immune system to activate nucleic acid sensors. It functions as an intermediate, conveying genetic information to control inflammatory responses. A key mechanism for RNA sensing is discriminating self from non-self nucleic acids to initiate antiviral responses reliably, including the expression of type I interferon (IFN) and IFN-stimulated genes. Another important aspect of the RNA-mediated inflammatory response is posttranscriptional regulation of gene expression, where RNA-binding proteins (RBPs) have essential roles in various RNA metabolisms, including splicing, nuclear export, modification, and translation and mRNA degradation. Recent evidence suggests that the control of mRNA stability is closely involved in signal transduction and orchestrates immune responses. In this study, we review the current understanding of how RNA is sensed by host RNA sensing machinery and discuss self/non-self-discrimination in innate immunity focusing on mammalian species. Finally, we discuss how posttranscriptional regulation by RBPs shape immune reactions.


Asunto(s)
Regulación de la Expresión Génica , Inmunidad Innata/genética , ARN/genética , Transcripción Genética , Animales , Enfermedad/genética , Humanos , ARN Viral/metabolismo
11.
Nat Genet ; 51(9): 1369-1379, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31477927

RESUMEN

Promoters and enhancers are key cis-regulatory elements, but how they operate to generate cell type-specific transcriptomes is not fully understood. We developed a simple and robust method, native elongating transcript-cap analysis of gene expression (NET-CAGE), to sensitively detect 5' ends of nascent RNAs in diverse cells and tissues, including unstable transcripts such as enhancer-derived RNAs. We studied RNA synthesis and degradation at the transcription start site level, characterizing the impact of differential promoter usage on transcript stability. We quantified transcription from cis-regulatory elements without the influence of RNA turnover, and show that enhancer-promoter pairs are generally activated simultaneously on stimulation. By integrating NET-CAGE data with chromatin interaction maps, we show that cis-regulatory elements are topologically connected according to their cell type specificity. We identified new enhancers with high sensitivity, and delineated primary locations of transcription within super-enhancers. Our NET-CAGE dataset derived from human and mouse cells expands the FANTOM5 atlas of transcribed enhancers, with broad applicability to biomedical research.


Asunto(s)
Regiones no Traducidas 5'/genética , Biología Computacional/métodos , Elementos de Facilitación Genéticos , Regulación de la Expresión Génica , Regiones Promotoras Genéticas , ARN/genética , Transcripción Genética , Perfilación de la Expresión Génica , Células HeLa , Células Hep G2 , Humanos , Células MCF-7 , Sitio de Iniciación de la Transcripción , Transcriptoma
12.
Nucleic Acids Res ; 47(16): 8838-8859, 2019 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-31329944

RESUMEN

Regnase-1-mediated mRNA decay (RMD), in which inflammatory mRNAs harboring specific stem-loop structures are degraded, is a critical part of proper immune homeostasis. Prior to initial translation, Regnase-1 associates with target stem-loops but does not carry out endoribonucleolytic cleavage. Single molecule imaging revealed that UPF1 is required to first unwind the stem-loops, thus licensing Regnase-1 to proceed with RNA degradation. Following translation, Regnase-1 physically associates with UPF1 using two distinct points of interaction: The Regnase-1 RNase domain binds to SMG1-phosphorylated residue T28 in UPF1; in addition, an intrinsically disordered segment in Regnase-1 binds to the UPF1 RecA domain, enhancing the helicase activity of UPF1. The SMG1-UPF1-Regnase-1 axis targets pioneer rounds of translation and is critical for rapid resolution of inflammation through restriction of the number of proteins translated by a given mRNA. Furthermore, small-molecule inhibition of SMG1 prevents RNA unwinding in dendritic cells, allowing post-transcriptional control of innate immune responses.


Asunto(s)
Macrófagos Peritoneales/inmunología , Degradación de ARNm Mediada por Codón sin Sentido/inmunología , Proteínas Serina-Treonina Quinasas/genética , ARN Mensajero/genética , Ribonucleasas/genética , Transactivadores/genética , Animales , Fibroblastos/citología , Fibroblastos/inmunología , Células HEK293 , Células HeLa , Homeostasis/genética , Homeostasis/inmunología , Humanos , Inmunidad Innata , Inflamación , Secuencias Invertidas Repetidas , Macrófagos/citología , Macrófagos/inmunología , Macrófagos Peritoneales/citología , Ratones , Ratones Noqueados , Mutación , Cultivo Primario de Células , Unión Proteica , Biosíntesis de Proteínas , Dominios y Motivos de Interacción de Proteínas , Proteínas Serina-Treonina Quinasas/inmunología , ARN Mensajero/metabolismo , Ribonucleasas/deficiencia , Ribonucleasas/inmunología , Imagen Individual de Molécula , Transactivadores/inmunología
13.
J Exp Med ; 216(6): 1431-1449, 2019 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-31072819

RESUMEN

Regnase-1 (also known as Zc3h12a or MCPIP-1) is an endoribonuclease involved in mRNA degradation of inflammation-associated genes. Regnase-1 is inactivated in response to external stimuli through post-translational modifications including phosphorylation, yet the precise role of phosphorylation remains unknown. Here, we demonstrate that interleukin (IL)-17 induces phosphorylation of Regnase-1 in an Act1-TBK1/IKKi-dependent manner, especially in nonhematopoietic cells. Phosphorylated Regnase-1 is released from the endoplasmic reticulum (ER) into the cytosol, thereby losing its mRNA degradation function, which leads to expression of IL-17 target genes. By using CRISPR/Cas-9 technology, we generated Regnase-1 mutant mice, in which IL-17-induced Regnase-1 phosphorylation is completely blocked. Mutant mice (Regnase-1AA/AA and Regnase-1ΔCTD/ΔCTD ) were resistant to the IL-17-mediated inflammation caused by T helper 17 (Th17) cells in vivo. Thus, Regnase-1 plays a critical role in the development of IL-17-mediated inflammatory diseases via the Act1-TBK1-IKKi axis, and blockade of Regnase-1 phosphorylation sites may be promising for treatment of Th17-associated diseases.


Asunto(s)
Retículo Endoplásmico/metabolismo , Interleucina-17/farmacología , Ribonucleasas/metabolismo , Animales , Citosol/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Retículo Endoplásmico/efectos de los fármacos , Quinasa I-kappa B/metabolismo , Inflamación/patología , Interleucina-6/genética , Interleucina-6/metabolismo , Ratones , Mutación/genética , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Estabilidad Proteica/efectos de los fármacos , Proteolisis/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ribonucleasas/química , Ribonucleasas/genética , Índice de Severidad de la Enfermedad , Transducción de Señal/efectos de los fármacos
14.
Nat Microbiol ; 4(9): 1532-1544, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31133753

RESUMEN

RNA-modulating factors not only regulate multiple steps of cellular RNA metabolism, but also emerge as key effectors of the immune response against invading viral pathogens including human immunodeficiency virus type-1 (HIV-1). However, the cellular RNA-binding proteins involved in the establishment and maintenance of latent HIV-1 reservoirs have not been extensively studied. Here, we screened a panel of 62 cellular RNA-binding proteins and identified NEDD4-binding protein 1 (N4BP1) as a potent interferon-inducible inhibitor of HIV-1 in primary T cells and macrophages. N4BP1 harbours a prototypical PilT N terminus-like RNase domain and inhibits HIV-1 replication by interacting with and degrading viral mRNA species. Following activation of CD4+ T cells, however, N4BP1 undergoes rapid cleavage at Arg 509 by the paracaspase named mucosa-associated lymphoid tissue lymphoma translocation 1 (MALT1). Mutational analyses and knockout studies revealed that MALT1-mediated inactivation of N4BP1 facilitates the reactivation of latent HIV-1 proviruses. Taken together, our findings demonstrate that the RNase N4BP1 is an efficient restriction factor of HIV-1 and suggest that inactivation of N4BP1 by induction of MALT1 activation might facilitate elimination of latent HIV-1 reservoirs.


Asunto(s)
Infecciones por VIH/virología , VIH-1/fisiología , Proteína 1 de la Translocación del Linfoma del Tejido Linfático Asociado a Mucosas/metabolismo , Proteínas Nucleares/metabolismo , Proteínas de Unión al ARN/metabolismo , Activación Viral/genética , Animales , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/virología , Línea Celular , Expresión Génica/efectos de los fármacos , Infecciones por VIH/metabolismo , Humanos , Interferón-alfa/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Macrófagos/virología , Ratones , Proteína 1 de la Translocación del Linfoma del Tejido Linfático Asociado a Mucosas/genética , Proteínas Nucleares/química , Proteínas Nucleares/genética , Dominios Proteicos , ARN Mensajero/metabolismo , ARN Viral/metabolismo , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Latencia del Virus
15.
Mucosal Immunol ; 11(4): 1203-1218, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29695841

RESUMEN

Inhaled pathogens including Pseudomonas aeruginosa initially encounter airway epithelial cells (AECs), which are poised to evoke cell-intrinsic innate defense, affecting second tier of hematopoietic cell-mediated immune reaction. However, it is largely unknown how pulmonary immune responses mediated by a variety of immune cells are coordinated. Here we show that Regnase-1, an endoribonuclease expressed in AECs and immune cells, plays an essential role in coordinating innate responses and adaptive immunity against P. aeruginosa infection. Intratracheal treatment of mice with heat-killed P. aeruginosa resulted in prolonged disappearance of Regnase-1 consistent with sustained expression of Regnase-1 target inflammatory genes, whereas the transcription factor NF-κB was only transiently activated. AEC-specific deletion of Regnase-1 not only augmented innate defenses against P. aeruginosa but also enhanced secretion of Pseudomonas-specific IgA and Th17 accumulation in the lung, culminating in conferring significant resistance against P. aeruginosa re-infection in vivo. Although Regnase-1 directly controls distinct sets of genes in each of AECs and T cells, degradation of Regnase-1 in both cell types is beneficial for maximizing acquired immune responses. Collectively, these results demonstrate that Regnase-1 orchestrates AEC-mediated and immune cell-mediated host defense against pulmonary bacterial infection.


Asunto(s)
Pulmón/inmunología , Neumonía Bacteriana/inmunología , Infecciones por Pseudomonas/inmunología , Pseudomonas aeruginosa/fisiología , Mucosa Respiratoria/metabolismo , Ribonucleasas/metabolismo , Células Th17/inmunología , Inmunidad Adaptativa , Animales , Anticuerpos Antibacterianos/metabolismo , Inmunidad Innata , Inmunoglobulina A/metabolismo , Pulmón/microbiología , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/metabolismo , Ribonucleasas/genética , Transducción de Señal
16.
J Immunol ; 199(12): 4066-4077, 2017 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-29127149

RESUMEN

Regnase-1 and Roquin are RNA binding proteins that are essential for degradation of inflammatory mRNAs and maintenance of immune homeostasis. Although deficiency of either of the proteins leads to enhanced T cell activation, their functional relationship in T cells has yet to be clarified because of lethality upon mutation of both Regnase-1 and Roquin. By using a Regnase-1 conditional allele, we show that mutations of both Regnase-1 and Roquin in T cells leads to massive lymphocyte activation. In contrast, mutation of either Regnase-1 or Roquin affected T cell activation to a lesser extent than the double mutation, indicating that Regnase-1 and Roquin function nonredundantly in T cells. Interestingly, Regnase-1 and Roquin double-mutant mice suffered from severe inflammation and early formation of fibrosis, especially in the heart, along with the increased expression of Ifng, but not Il4 or Il17a Consistently, mutation of both Regnase-1 and Roquin leads to a huge increase in the Th1, but not the Th2 or Th17, population in spleens compared with T cells with a single Regnase-1 or Roquin deficiency. Regnase-1 and Roquin are capable of repressing the expression of a group of mRNAs encoding factors involved in Th1 differentiation, such as Furin and Il12rb1, via their 3' untranslated regions. Moreover, Regnase-1 is capable of repressing Roquin mRNA. This cross-regulation may contribute to the synergistic control of T cell activation/polarization. Collectively, our results demonstrate that Regnase-1 and Roquin maintain T cell immune homeostasis and regulate Th1 polarization synergistically.


Asunto(s)
Miocarditis/inmunología , Miocardio/patología , Ribonucleasas/fisiología , Células TH1/inmunología , Ubiquitina-Proteína Ligasas/fisiología , Regiones no Traducidas 3' , Animales , Fibrosis , Furina/biosíntesis , Furina/genética , Regulación de la Expresión Génica/inmunología , Células HeLa , Homeostasis , Humanos , Interferón gamma/biosíntesis , Interferón gamma/genética , Interleucina-17/biosíntesis , Interleucina-17/genética , Interleucina-4/biosíntesis , Interleucina-4/genética , Células Jurkat , Activación de Linfocitos , Linfopoyesis/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Mutantes , Miocarditis/genética , ARN Mensajero/biosíntesis , Receptores de Interleucina-12/biosíntesis , Receptores de Interleucina-12/genética , Proteínas Recombinantes de Fusión/metabolismo , Ribonucleasas/deficiencia , Ribonucleasas/genética , Organismos Libres de Patógenos Específicos , Bazo/citología , Bazo/inmunología , Células TH1/patología , Ubiquitina-Proteína Ligasas/deficiencia , Ubiquitina-Proteína Ligasas/genética
17.
J Interferon Cytokine Res ; 37(5): 220-229, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28475459

RESUMEN

Regnase-1, also known as Zc3h12a or MCPIP1, is responsible for endonucleolytic cleavage of mRNAs encoding proteins involved in inflammatory responses. Furthermore, Regnase-1-mediated mRNA decay (RMD) is critical for the maintenance of immune homeostasis. Regnase-1 controls the magnitude of innate and adaptive immune responses, and thereby dysfunction of this protein in mice leads to the development of spontaneous systemic inflammation. During the last several years, advances have been made in understanding molecular mechanisms of RMD. In this article, unique functions of Regnase-1 in controlling inflammation are discussed.


Asunto(s)
Homeostasis/inmunología , Ribonucleasas/metabolismo , Factores de Transcripción/metabolismo , Animales , Humanos , Inflamación/metabolismo , Ratones , ARN Mensajero/metabolismo
18.
Cell Rep ; 19(8): 1614-1630, 2017 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-28538180

RESUMEN

Iron metabolism is regulated by transcriptional and post-transcriptional mechanisms. The mRNA of the iron-controlling gene, transferrin receptor 1 (TfR1), has long been believed to be negatively regulated by a yet-unidentified endonuclease. Here, we show that the endonuclease Regnase-1 is critical for the degradation of mRNAs involved in iron metabolism in vivo. First, we demonstrate that Regnase-1 promotes TfR1 mRNA decay. Next, we show that Regnase-1-/- mice suffer from severe iron deficiency anemia, although hepcidin expression is downregulated. The iron deficiency anemia is induced by a defect in duodenal iron uptake. We reveal that duodenal Regnase-1 controls the expression of PHD3, which impairs duodenal iron uptake via HIF2α suppression. Finally, we show that Regnase-1 is a HIF2α-inducible gene and thus provides a positive feedback loop for HIF2α activation via PHD3. Collectively, these results demonstrate that Regnase-1-mediated regulation of iron-related transcripts is essential for the maintenance of iron homeostasis.


Asunto(s)
Antígenos CD/metabolismo , Homeostasis , Hierro/metabolismo , Procolágeno-Prolina Dioxigenasa/metabolismo , Estabilidad del ARN , Receptores de Transferrina/metabolismo , Ribonucleasas/metabolismo , Anemia/metabolismo , Anemia/patología , Animales , Antígenos CD/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Duodeno/metabolismo , Ferritinas/metabolismo , Ratones , Regiones Promotoras Genéticas/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Transferrina/genética , Elementos de Respuesta/genética , Ribonucleasas/deficiencia , Transcripción Genética
19.
Cell ; 161(5): 1058-1073, 2015 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-26000482

RESUMEN

Regnase-1 and Roquin are RNA binding proteins essential for degradation of inflammation-related mRNAs and maintenance of immune homeostasis. However, their mechanistic relationship has yet to be clarified. Here, we show that, although Regnase-1 and Roquin regulate an overlapping set of mRNAs via a common stem-loop structure, they function in distinct subcellular locations: ribosome/endoplasmic reticulum and processing-body/stress granules, respectively. Moreover, Regnase-1 specifically cleaves and degrades translationally active mRNAs and requires the helicase activity of UPF1, similar to the decay mechanisms of nonsense mRNAs. In contrast, Roquin controls translationally inactive mRNAs, independent of UPF1. Defects in both Regnase-1 and Roquin lead to large increases in their target mRNAs, although Regnase-1 tends to control the early phase of inflammation when mRNAs are more actively translated. Our findings reveal that differential regulation of mRNAs by Regnase-1 and Roquin depends on their translation status and enables elaborate control of inflammation.


Asunto(s)
Inflamación/metabolismo , Estabilidad del ARN , ARN Mensajero/metabolismo , Ribonucleasas/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Secuencia de Bases , Codón de Terminación , Células HeLa , Humanos , Inflamación/genética , Inflamación/inmunología , Ratones , Datos de Secuencia Molecular , Células 3T3 NIH , Conformación de Ácido Nucleico , Polirribosomas/metabolismo , Biosíntesis de Proteínas , ARN Mensajero/química , Proteínas Ribosómicas/metabolismo , Transactivadores/metabolismo
20.
Cell ; 153(5): 1036-49, 2013 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-23706741

RESUMEN

Regnase-1 (also known as Zc3h12a and MCPIP1) is an RNase that destabilizes a set of mRNAs, including Il6 and Il12b, through cleavage of their 3' UTRs. Although Regnase-1 inactivation leads to development of an autoimmune disease characterized by T cell activation and hyperimmunoglobulinemia in mice, the mechanism of Regnase-1-mediated immune regulation has remained unclear. We show that Regnase-1 is essential for preventing aberrant effector CD4(+) T cell generation cell autonomously. Moreover, in T cells, Regnase-1 regulates the mRNAs of a set of genes, including c-Rel, Ox40, and Il2, through cleavage of their 3' UTRs. Interestingly, T cell receptor (TCR) stimulation leads to cleavage of Regnase-1 at R111 by Malt1/paracaspase, freeing T cells from Regnase-1-mediated suppression. Furthermore, Malt1 protease activity is critical for controlling the mRNA stability of T cell effector genes. Collectively, these results indicate that dynamic control of Regnase-1 expression in T cells is critical for controlling T cell activation.


Asunto(s)
Caspasas/metabolismo , Activación de Linfocitos , Proteínas de Neoplasias/metabolismo , Ribonucleasas/metabolismo , Linfocitos T Colaboradores-Inductores/inmunología , Animales , Enfermedades Autoinmunes/inmunología , Humanos , Interleucina-2/genética , Células Jurkat , Glicoproteínas de Membrana/genética , Ratones , Proteína 1 de la Translocación del Linfoma del Tejido Linfático Asociado a Mucosas , Ligando OX40 , Proteínas Proto-Oncogénicas c-rel/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T Colaboradores-Inductores/metabolismo , Factores de Necrosis Tumoral/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA