Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
1.
J Cell Sci ; 136(14)2023 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-37350228

RESUMEN

Cells trigger the assembly of stress granules (SGs) under various stress conditions. Among the many proteins recruited to SGs are RNA-binding proteins and transcription regulators. Here, we report the translocation of human (h)Cdc73, a component of the PAF1 transcription complex, to cytosolic SGs in response to arsenic stress. The hCdc73 protein possesses a long intrinsically disordered region (IDR) from amino acids 256-416, the presence of which is required for the translocation of hCdc73 to cytosolic SGs. The purified hCdc73 IDR formed droplets in vitro, and the light-activated assembly of hCdc73-IDR-mCherry-CRY2 was verified. For translocation of hCdc73 to SGs, physical interactions with SG carrier proteins, such as FMR1, are also needed. Previously, we reported that the cytosolic hCdc73-eEF1Bγ complex controls the stability of p53 mRNA. Under arsenic stress, selective sequestration of cytosolic hCdc73, but not eEF1Bγ (EEF1G) or p53 (TP53) mRNA, was detected. As a result, a transient increase in p53 mRNA at the post-transcriptional level was observed. In conclusion, we propose that the availability of mRNAs for stress-responsive genes can be controlled by restraining their negative regulators within SGs.


Asunto(s)
Arsénico , Proteína p53 Supresora de Tumor , Humanos , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Arsénico/metabolismo , Gránulos de Estrés , Gránulos Citoplasmáticos/genética , Gránulos Citoplasmáticos/metabolismo , Estrés Fisiológico/genética , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/metabolismo , Factores de Transcripción/metabolismo
2.
EMBO Rep ; 24(8): e56538, 2023 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-37377038

RESUMEN

The ER regulates the spatiotemporal organization of endolysosomal systems by membrane contact. In addition to tethering via heterotypic interactions on both organelles, we present a novel ER-endosome tethering mechanism mediated by homotypic interactions. The single-pass transmembrane protein SCOTIN is detected in the membrane of the ER and endosomes. In SCOTIN-knockout (KO) cells, the ER-late endosome contacts are reduced, and the perinuclear positioning of endosomes is disturbed. The cytosolic proline-rich domain (PRD) of SCOTIN forms homotypic assemblies in vitro and is necessary for ER-endosome membrane tethering in cells. A region of 28 amino acids spanning 150-177 within the SCOTIN PRD is essential to elicit membrane tethering and endosomal dynamics, as verified by reconstitution in SCOTIN-KO cells. The assembly of SCOTIN (PRD) is sufficient to mediate membrane tethering, as purified SCOTIN (PRD), but not SCOTIN (PRDΔ150-177), brings two different liposomes closer in vitro. Using organelle-specific targeting of a chimeric PRD domain shows that only the presence on both organellar membranes enables the ER-endosome membrane contact, indicating that the assembly of SCOTIN on heterologous membranes mediates organelle tethering.


Asunto(s)
Retículo Endoplásmico , Membranas Intracelulares , Membranas Intracelulares/metabolismo , Retículo Endoplásmico/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Endosomas/metabolismo
3.
PLoS Genet ; 16(3): e1008617, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32130226

RESUMEN

The oligosaccharyl transferase (OST) protein complex mediates the N-linked glycosylation of substrate proteins in the endoplasmic reticulum (ER), which regulates stability, activity, and localization of its substrates. Although many OST substrate proteins have been identified, the physiological role of the OST complex remains incompletely understood. Here we show that the OST complex in C. elegans is crucial for ER protein homeostasis and defense against infection with pathogenic bacteria Pseudomonas aeruginosa (PA14), via immune-regulatory PMK-1/p38 MAP kinase. We found that genetic inhibition of the OST complex impaired protein processing in the ER, which in turn up-regulated ER unfolded protein response (UPRER). We identified vitellogenin VIT-6 as an OST-dependent glycosylated protein, critical for maintaining survival on PA14. We also showed that the OST complex was required for up-regulation of PMK-1 signaling upon infection with PA14. Our study demonstrates that an evolutionarily conserved OST complex, crucial for ER homeostasis, regulates host defense mechanisms against pathogenic bacteria.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/metabolismo , Retículo Endoplásmico/metabolismo , Proteostasis/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Hexosiltransferasas/metabolismo , Inmunidad Innata/fisiología , Sistema de Señalización de MAP Quinasas/fisiología , Proteínas de la Membrana/metabolismo , Pseudomonas aeruginosa/metabolismo , Transducción de Señal/fisiología , Factores de Transcripción/metabolismo , Respuesta de Proteína Desplegada/fisiología , Regulación hacia Arriba/fisiología , Vitelogeninas/metabolismo
4.
EMBO J ; 36(8): 1046-1065, 2017 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-28283579

RESUMEN

Mitochondria play key roles in cellular immunity. How mitochondria contribute to organismal immunity remains poorly understood. Here, we show that HSP-60/HSPD1, a major mitochondrial chaperone, boosts anti-bacterial immunity through the up-regulation of p38 MAP kinase signaling. We first identify 16 evolutionarily conserved mitochondrial components that affect the immunity of Caenorhabditis elegans against pathogenic Pseudomonas aeruginosa (PA14). Among them, the mitochondrial chaperone HSP-60 is necessary and sufficient to increase resistance to PA14. We show that HSP-60 in the intestine and neurons is crucial for the resistance to PA14. We then find that p38 MAP kinase signaling, an evolutionarily conserved anti-bacterial immune pathway, is down-regulated by genetic inhibition of hsp-60, and up-regulated by increased expression of hsp-60 Overexpression of HSPD1, the mammalian ortholog of hsp-60, increases p38 MAP kinase activity in human cells, suggesting an evolutionarily conserved mechanism. Further, cytosol-localized HSP-60 physically binds and stabilizes SEK-1/MAP kinase kinase 3, which in turn up-regulates p38 MAP kinase and increases immunity. Our study suggests that mitochondrial chaperones protect host eukaryotes from pathogenic bacteria by up-regulating cytosolic p38 MAPK signaling.


Asunto(s)
Caenorhabditis elegans/inmunología , Chaperonina 60/inmunología , Sistema de Señalización de MAP Quinasas/inmunología , Proteínas Mitocondriales/inmunología , Pseudomonas aeruginosa/inmunología , Proteínas Quinasas p38 Activadas por Mitógenos/inmunología , Animales , Animales Modificados Genéticamente/genética , Animales Modificados Genéticamente/inmunología , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/inmunología , Chaperonina 60/genética , Humanos , MAP Quinasa Quinasa 4/genética , MAP Quinasa Quinasa 4/inmunología , Sistema de Señalización de MAP Quinasas/genética , Proteínas Mitocondriales/genética , Proteínas Quinasas p38 Activadas por Mitógenos/genética
5.
J Theor Biol ; 527: 110816, 2021 10 21.
Artículo en Inglés | MEDLINE | ID: mdl-34161792

RESUMEN

We study the influence of spatial heterogeneity on the antiviral activity of mouse embryonic fibroblasts (MEF) infected with influenza A. MEF of type Ube1L-/- are composed of two distinct sub-populations, the strong type that sustains a strong viral infection and the weak type, sustaining a weak viral load. We present new data on the virus load infection of Ube1L-/-, which have been micro-printed in a checker board pattern of different sizes of the inner squares. Surprisingly, the total viral load at one day after inoculation significantly depends on the sizes of the inner squares. We explain this observation by using a reaction diffusion model and we show that mathematical homogenization can explain the observed inhomogeneities. If the individual patches are large, then the growth rate and the carrying capacity will be the arithmetic means of the patches. For finer and finer patches the average growth rate is still the arithmetic mean, however, the carrying capacity uses the harmonic mean. While fitting the PDE to the experimental data, we also predict that a discrepancy in virus load would be unobservable after only half a day. Furthermore, we predict the viral load in different inner squares that had not been measured in our experiment and the travelling distance the virions can reach after one day.


Asunto(s)
Virus de la Influenza A , Gripe Humana , Animales , Antivirales/uso terapéutico , Fibroblastos , Humanos , Gripe Humana/tratamiento farmacológico , Ratones , Carga Viral
6.
J Virol ; 93(20)2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31341044

RESUMEN

Proteoglycans function not only as structural components of the extracellular compartment but also as regulators of various cellular events, including cell migration, inflammation, and infection. Many microbial pathogens utilize proteoglycans to facilitate adhesion and invasion into host cells. Here we report a secreted form of a novel heparan sulfate proteoglycan that functions against virus infection. The expression of SPOCK2/testican-2 was significantly induced in virus-infected lungs or in interferon (IFN)-treated alveolar lung epithelial cells. Overexpression from a SPOCK2 expression plasmid alone or the treatment of cells with recombinant SPOCK2 protein efficiently blocked influenza virus infection at the step of viral attachment to the host cell and entry. Moreover, mice treated with purified SPOCK2 were protected against virus infection. Sialylated glycans and heparan sulfate chains covalently attached to the SPOCK2 core protein were critical for its antiviral activity. Neuraminidase (NA) of influenza virus cleaves the sialylated moiety of SPOCK2, thereby blocking its binding to the virus. Our data suggest that IFN-induced SPOCK2 functions as a decoy receptor to bind and block influenza virus infection, thereby restricting entry of the infecting virus into neighboring cells.IMPORTANCE Here we report a novel proteoglycan protein, testican-2/SPOCK2, that prevents influenza virus infection. Testican-2/SPOCK2 is a complex type of secreted proteoglycan with heparan sulfate GAG chains attached to the core protein. SPOCK2 expression is induced upon virus infection or by interferons, and the protein is secreted to an extracellular compartment, where it acts directly to block virus-cell attachment and entry. Treatment with purified testican-2/SPOCK2 protein can efficiently block influenza virus infection in vitro and in vivo We also identified the heparan sulfate moiety as a key regulatory module for this inhibitory effect. Based on its mode of action (cell attachment/entry blocker) and site of action (extracellular compartment), we propose testican-2/SPOCK2 as a potential antiviral agent that can efficiently control influenza virus infection.


Asunto(s)
Células Epiteliales Alveolares/metabolismo , Células Epiteliales Alveolares/virología , Resistencia a la Enfermedad/genética , Interacciones Huésped-Patógeno/genética , Proteoglicanos/genética , Virosis/etiología , Virosis/metabolismo , Animales , Antivirales/farmacología , Línea Celular , Modelos Animales de Enfermedad , Femenino , Expresión Génica , Proteoglicanos de Heparán Sulfato , Humanos , Virus de la Influenza A/efectos de los fármacos , Ratones , Proteoglicanos/metabolismo , Proteoglicanos/farmacología , Proteínas Recombinantes/farmacología , Acoplamiento Viral/efectos de los fármacos , Replicación Viral/efectos de los fármacos
7.
Nephrol Dial Transplant ; 34(9): 1481-1498, 2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-30544214

RESUMEN

BACKGROUND: Signal transducer and activator of transcription 3 (STAT3) is a latent transcription factor critical for T-cell function. Although inhibition of the Janus kinase 2 (JAK2)/STAT3 pathway has been reported to be protective against ischemia-reperfusion injury (IRI), the role of T cell-associated STAT3 in the pathogenesis of renal IRI has not been specifically defined. METHODS: We induced renal IRI in both mice with T cell-specific STAT3 knockout (Lck-Cre;STAT3flox/flox) and wild-type controls (C57BL/6) and assessed renal damage and inflammation at 48 h after IRI. Human proximal tubular epithelial cells grown under hypoxia were treated with a JAK2 inhibitor, caffeic acid 3,4-dihydroxy-phenylethyl ester, to determine the effect of JAK2/STAT3 inhibition on renal epithelia. Independently, we disrupted Cln 3-requiring 9 (Ctr9) to inhibit T helper 17 (Th17) activation via RNA interference and determined if Ctr9 inhibition aggravates renal injury through upregulated Th17 activation. RESULTS: The Lck-Cre;STAT3flox/flox mice exhibited significantly reduced kidney damage compared with controls. This protective effect was associated with reduced intrarenal Th17 infiltration and proinflammatory cytokines. Human proximal tubular epithelial cells under hypoxia exhibited significant upregulation of interleukin 17 receptors, and pharmacologic inhibition of JAK2 significantly ameliorated this change. RNA interference with Ctr9 in splenocytes enhanced differentiation into Th17 cells. In vivo knockdown of Ctr9 in mice with renal IRI further aggravated Th17-associated inflammation and kidney injury. CONCLUSIONS: STAT3 in T cells contributes to renal IRI through Th17 activation. Inhibition of Ctr9 further enhances Th17 activation and aggravates kidney injury, further supporting the role of Th17 cells in renal IRI.


Asunto(s)
Regulación de la Expresión Génica , Inflamación/prevención & control , Interleucina-17/genética , Riñón/inmunología , Daño por Reperfusión/prevención & control , Células Th17/inmunología , Animales , Citocinas/metabolismo , Humanos , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , Interleucina-17/metabolismo , Janus Quinasa 2/genética , Janus Quinasa 2/metabolismo , Janus Quinasa 3/genética , Janus Quinasa 3/metabolismo , Riñón/metabolismo , Riñón/patología , Ratones , Ratones Endogámicos C57BL , Daño por Reperfusión/inmunología , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Células Th17/metabolismo , Células Th17/patología
8.
Am J Hum Genet ; 96(2): 266-74, 2015 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-25620203

RESUMEN

Singleton-Merten syndrome (SMS) is an autosomal-dominant multi-system disorder characterized by dental dysplasia, aortic calcification, skeletal abnormalities, glaucoma, psoriasis, and other conditions. Despite an apparent autosomal-dominant pattern of inheritance, the genetic background of SMS and information about its phenotypic heterogeneity remain unknown. Recently, we found a family affected by glaucoma, aortic calcification, and skeletal abnormalities. Unlike subjects with classic SMS, affected individuals showed normal dentition, suggesting atypical SMS. To identify genetic causes of the disease, we performed exome sequencing in this family and identified a variant (c.1118A>C [p.Glu373Ala]) of DDX58, whose protein product is also known as RIG-I. Further analysis of DDX58 in 100 individuals with congenital glaucoma identified another variant (c.803G>T [p.Cys268Phe]) in a family who harbored neither dental anomalies nor aortic calcification but who suffered from glaucoma and skeletal abnormalities. Cys268 and Glu373 residues of DDX58 belong to ATP-binding motifs I and II, respectively, and these residues are predicted to be located closer to the ADP and RNA molecules than other nonpathogenic missense variants by protein structure analysis. Functional assays revealed that DDX58 alterations confer constitutive activation and thus lead to increased interferon (IFN) activity and IFN-stimulated gene expression. In addition, when we transduced primary human trabecular meshwork cells with c.803G>T (p.Cys268Phe) and c.1118A>C (p.Glu373Ala) mutants, cytopathic effects and a significant decrease in cell number were observed. Taken together, our results demonstrate that DDX58 mutations cause atypical SMS manifesting with variable expression of glaucoma, aortic calcification, and skeletal abnormalities without dental anomalies.


Asunto(s)
Enfermedades de la Aorta/genética , ARN Helicasas DEAD-box/genética , Hipoplasia del Esmalte Dental/genética , Glaucoma/genética , Metacarpo/anomalías , Modelos Moleculares , Enfermedades Musculares/genética , Odontodisplasia/genética , Osteoporosis/genética , Calcificación Vascular/genética , Adulto , Enfermedades de la Aorta/patología , Secuencia de Bases , Células Cultivadas , Preescolar , Proteína 58 DEAD Box , ARN Helicasas DEAD-box/química , Hipoplasia del Esmalte Dental/patología , Exoma/genética , Femenino , Genes Dominantes/genética , Humanos , Masculino , Metacarpo/patología , Datos de Secuencia Molecular , Enfermedades Musculares/patología , Anomalías Musculoesqueléticas/diagnóstico por imagen , Anomalías Musculoesqueléticas/genética , Mutación Missense/genética , Odontodisplasia/diagnóstico por imagen , Odontodisplasia/patología , Osteoporosis/patología , Linaje , Polimorfismo de Nucleótido Simple/genética , Radiografía , Receptores Inmunológicos , Análisis de Secuencia de ADN , Calcificación Vascular/patología
9.
J Biol Chem ; 290(11): 7087-96, 2015 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-25635053

RESUMEN

Dysbindin and DISC1 are schizophrenia susceptibility factors playing roles in neuronal development. Here we show that the physical interaction between dysbindin and DISC1 is critical for the stability of dysbindin and for the process of neurite outgrowth. We found that DISC1 forms a complex with dysbindin and increases its stability in association with a reduction in ubiquitylation. Furthermore, knockdown of DISC1 or expression of a deletion mutant, DISC1 lacking amino acid residues 403-504 of DISC1 (DISC1(Δ403-504)), effectively decreased levels of endogenous dysbindin. Finally, the neurite outgrowth defect induced by knockdown of DISC1 was partially reversed by coexpression of dysbindin. Taken together, these results indicate that dysbindin and DISC1 form a physiologically functional complex that is essential for normal neurite outgrowth.


Asunto(s)
Proteínas Asociadas a la Distrofina/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Animales , Células COS , Células Cultivadas , Corteza Cerebral/citología , Chlorocebus aethiops , Disbindina , Proteínas Asociadas a la Distrofina/química , Células HEK293 , Humanos , Ratones , Proteínas del Tejido Nervioso/química , Neuritas/metabolismo , Neuritas/patología , Neuronas/metabolismo , Neuronas/patología , Complejo de la Endopetidasa Proteasomal/metabolismo , Estabilidad Proteica , Ubiquitina
10.
Am J Physiol Renal Physiol ; 311(3): F505-19, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27306982

RESUMEN

T helper 17 (Th17) lymphocytes promote renal inflammation in anti-glomerular basement membrane glomerulonephritis (anti-GBM GN), and signal transducer and activator of transcription 3 (STAT3) mediates activation of Th17 lymphocytes by IL-6 and transforming growth factor-ß (TGF-ß). Cln 3-requiring 9 (Ctr9), a subunit of RNA polymerase-associated factor complex (PAFc), regulates the transcription of IL-6/STAT3-dependent genes. Here, we investigated the role of Ctr9 in regulating Th17-driven inflammation in anti-GBM GN. In mice, STAT3ß or IL-17 knockout ameliorated anti-GBM autoantibody-induced renal injury. This phenomenon was associated with decreases in retinoic acid receptor-related orphan receptor γt (RORγt), IL-17, phosphorylated STAT3, and proinflammatory cytokines. Compared with wild-type mice, Ctr9 increased in both STAT3ß(-/-) and IL-17(-/-) mice injected with anti-GBM IgG, showing a negative correlation with Th17-related transcripts. Small interfering RNA (siRNA)-mediated knockdown of Ctr9 in intrarenal lymphocytes further upregulated Th17-related transcripts, consistent with repression of Th17 differentiation by Ctr9. Interestingly, Ctr9 was also expressed in human and mouse mesangial cells and downregulated in response to anti-GBM IgG or to TGF-ß plus IL-17. Ctr9 in mesangial cells was even more repressed in the presence of both anti-GBM IgG and Th17-activating cytokines. Consistent with these findings, renal biopsies obtained from patients with anti-GBM GN showed consistent downregulation of Ctr9 and upregulation of phosphorylated STAT3 and IL-17 in the glomerulus. We conclude that Ctr9 is a negative regulator of Th17 differentiation in anti-GBM GN and repressed by anti-GBM IgG and IL-17 in mesangial cells.


Asunto(s)
Membrana Basal Glomerular/metabolismo , Glomerulonefritis/metabolismo , Inflamación/metabolismo , Riñón/metabolismo , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Animales , Membrana Basal Glomerular/patología , Glomerulonefritis/patología , Humanos , Inflamación/patología , Interleucina-17/metabolismo , Riñón/patología , Células Mesangiales/metabolismo , Células Mesangiales/patología , Ratones , Fosforilación , Factor de Transcripción STAT3/metabolismo
11.
J Immunol ; 192(4): 1440-8, 2014 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-24420920

RESUMEN

PAF complex is an evolutionarily conserved transcriptional complex that associates with RNA polymerase II in the coding region of actively transcribing genes. Although its transcriptional activity is closely related to diverse cellular processes, such as cell-cycle progression or development in mammals, its role in immune responses has not been addressed yet. In this study, we show that CTR9, a component of PAF complex, functions as a repressor of Th17 differentiation. Both mRNA and protein levels of CTR9 were significantly decreased during the differentiation processes of naive T into Th17 effector cells. When CTR9 was depleted, IL-17 expression was induced and differentiation into Th17 cells enhanced. In naive T cells, CTR9 occupied the coding region of Il17a, but dissociated under Th17 in vitro-polarizing conditions. In contrast, both CDC73 and PAF1 were recruited to the Il17a locus under Th17-differentiation conditions. In the IL-6-stimulated splenocytes, expression of CTR9 was decreased, and chromatin-bound CTR9 disappeared in the coding region of Il17a. IL-6 also directly repressed expression of CTR9 gene, as promoter activity of CTR9 was similarly repressed by IL-6 treatment. Moreover, in mice with collagen-induced arthritis, lentivirus-mediated CTR9 overexpression in the joints ameliorated arthritis severity, decreasing the frequency of CD4(+)IL-17(+) T cells in lymph nodes. In conclusion, our data propose a novel feed-forward loop of IL-17 transcriptional regulatory circuit, via IL-6-mediated repression of CTR9 which is a transcriptional repressor of IL-17.


Asunto(s)
Diferenciación Celular/genética , Regulación de la Expresión Génica , Interleucina-17/genética , Interleucina-6/metabolismo , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Células Th17/citología , Animales , Artritis Experimental/genética , Artritis Experimental/metabolismo , Proteínas Portadoras/metabolismo , Ganglios Linfáticos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Proteínas Nucleares/genética , Fosfoproteínas/genética , Regiones Promotoras Genéticas , Unión Proteica , ARN Mensajero/biosíntesis , Factor de Transcripción STAT3/metabolismo , Bazo/citología , Bazo/metabolismo , Células Th17/inmunología , Factores de Transcripción , Proteínas Supresoras de Tumor/metabolismo
12.
J Immunol ; 190(3): 1192-200, 2013 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-23284052

RESUMEN

In RNA virus-infected cells, retinoic acid-inducible gene-I-like receptors (RLRs) sense foreign RNAs and activate signaling cascades to produce IFN-α/ß. However, not every infected cell produces IFN-α/ß that exhibits cellular heterogeneity in antiviral immune responses. Using the IFN-ß-GFP reporter system, we observed bimodal IFN-ß production in the uniformly stimulated cell population with intracellular dsRNA. Mathematical simulation proposed the strength of autocrine loop via RLR as one of the contributing factor for biphasic IFN-ß expression. Bimodal IFN-ß production with intracellular dsRNA was disturbed by blockage of IFN-α/ß secretion or by silencing of the IFN-α/ß receptor. Amplification of RLRs was critical in the generation of bimodality of IFN-ß production, because IFN-ß(high) population expressed more RLRs than IFN-ß(low) population. In addition, bimodality in IFN-ß production results in biphasic cellular response against infection, because IFN-ß(high) population was more prone to apoptosis than IFN-ß(low) population. These results suggest that RLR-mediated biphasic cellular response may act to restrict the number of cells expressing IFN-ß and undergoing apoptosis in the infected population.


Asunto(s)
Apoptosis/inmunología , Comunicación Autocrina/inmunología , ARN Helicasas DEAD-box/fisiología , Regulación de la Expresión Génica/inmunología , Interferón beta/biosíntesis , Modelos Inmunológicos , Virosis/inmunología , Brefeldino A/farmacología , Proteína 58 DEAD Box , ARN Helicasas DEAD-box/antagonistas & inhibidores , ARN Helicasas DEAD-box/biosíntesis , ARN Helicasas DEAD-box/genética , Genes Reporteros , Células Hep G2/efectos de los fármacos , Células Hep G2/metabolismo , Humanos , Inductores de Interferón/farmacología , Helicasa Inducida por Interferón IFIH1 , Interferón beta/genética , Interferón beta/inmunología , Interferón beta/metabolismo , Poli I-C/farmacología , Transporte de Proteínas/efectos de los fármacos , Interferencia de ARN , ARN Bicatenario/farmacología , ARN Interferente Pequeño/farmacología , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/inmunología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Procesos Estocásticos , Transcripción Genética , Transfección
13.
Nucleic Acids Res ; 41(8): 4495-506, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23502002

RESUMEN

To determine the functional specificity of inflammation, it is critical to orchestrate the timely activation and repression of inflammatory responses. Here, we explored the PAF1 (RNA polymerase II associated factor)-mediated signal- and locus-specific repression of genes induced through the pro-inflammatory cytokine interleukin (IL)-1ß. Using microarray analysis, we identified the PAF1 target genes whose expression was further enhanced by PAF1 knockdown in IL-1ß-stimulated HepG2 hepatocarcinomas. PAF1 bound near the transcription start sites of target genes and dissociated on stimulation. In PAF1-deficient cells, more elongating RNA polymerase II and acetylated histones were observed, although IL-1ß-mediated activation and recruitment of nuclear factor κB (NF-κB) were not altered. Under basal conditions, PAF1 blocked histone acetyltransferase general control non-depressible 5 (GCN5)-mediated acetylation on H3K9 and H4K5 residues. On IL-1ß stimulation, activated GCN5 discharged PAF1 from chromatin, allowing productive transcription to occur. PAF1 bound to histones but not to acetylated histones, and the chromatin-binding domain of PAF1 was essential for target gene repression. Moreover, IL-1ß-induced cell migration was similarly controlled through counteraction between PAF1 and GCN5. These results suggest that the IL-1ß signal-specific exchange of PAF1 and GCN5 on the target locus limits inappropriate gene induction and facilitates the timely activation of inflammatory responses.


Asunto(s)
Proteínas Portadoras/metabolismo , Cromatina/metabolismo , Regulación de la Expresión Génica , Histona Acetiltransferasas/metabolismo , Mediadores de Inflamación/metabolismo , Interleucina-1beta/farmacología , Factores de Transcripción p300-CBP/metabolismo , Acetilación , Animales , Proteínas Portadoras/química , Línea Celular Tumoral , Movimiento Celular , Histonas/metabolismo , Humanos , Ratones , FN-kappa B/metabolismo , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Interferencia de ARN , Elongación de la Transcripción Genética , Factores de Transcripción , Sitio de Iniciación de la Transcripción , Transcripción Genética , Activador de Plasminógeno de Tipo Uroquinasa/genética
14.
Nucleic Acids Res ; 40(5): e38, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22187154

RESUMEN

Accurate prediction of transcription factor binding sites (TFBSs) is a prerequisite for identifying cis-regulatory modules that underlie transcriptional regulatory circuits encoded in the genome. Here, we present a computational framework for detecting TFBSs, when multiple position weight matrices (PWMs) for a transcription factor are available. Grouping multiple PWMs of a transcription factor (TF) based on their sequence similarity improves the specificity of TFBS prediction, which was evaluated using multiple genome-wide ChIP-Seq data sets from 26 TFs. The Z-scores of the area under a receiver operating characteristic curve (AUC) values of 368 TFs were calculated and used to statistically identify co-occurring regulatory motifs in the TF bound ChIP loci. Motifs that are co-occurring along with the empirical bindings of E2F, JUN or MYC have been evaluated, in the basal or stimulated condition. Results prove our method can be useful to systematically identify the co-occurring motifs of the TF for the given conditions.


Asunto(s)
Posición Específica de Matrices de Puntuación , Elementos Reguladores de la Transcripción , Análisis de Secuencia de ADN , Factores de Transcripción/metabolismo , Secuencia de Bases , Sitios de Unión , Secuencia Conservada , Factores de Transcripción E2F/metabolismo , Motivos de Nucleótidos , Proteínas Proto-Oncogénicas c-jun/metabolismo , Programas Informáticos
15.
Nucleic Acids Res ; 40(6): 2724-33, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22127865

RESUMEN

RIG-I is a cytosolic receptor for non-self RNA that mediates immune responses against viral infections through IFNα/ß production. In an attempt to identify novel tools that modulate IFNα/ß production, we used SELEX technology to screen RNA aptamers that specifically target RIG-I protein. Most of the selected RIG-I aptamers contained polyU motifs in the second half regions that played critical roles in the activation of RIG-I-mediated IFNß production. Unlike other known ligands, RIG-I aptamer bound and activated RIG-I in a 5'-triphosphate-independent manner. The helicase and RD domain of RIG-I were used for aptamer binding, but intact RIG-I protein was required to exert aptamer-mediated signaling activation. Furthermore, replication of NDV, VSV and influenza virus in infected host cells was efficiently blocked by pre- or post-treatment with RIG-I aptamer. Based on these data, we propose that RIG-I aptamer has strong potential to be an antiviral agent that specifically boosts the RIG-I-dependent signaling cascade.


Asunto(s)
Antivirales/farmacología , Aptámeros de Nucleótidos/farmacología , ARN Helicasas DEAD-box/metabolismo , Antivirales/química , Aptámeros de Nucleótidos/química , Secuencia de Bases , Línea Celular , Proteína 58 DEAD Box , Humanos , Interferón beta/biosíntesis , Datos de Secuencia Molecular , Poli U/química , Polifosfatos/química , ARN/química , Receptores Inmunológicos , Técnica SELEX de Producción de Aptámeros , Transducción de Señal/efectos de los fármacos , Replicación Viral/efectos de los fármacos
16.
J Arrhythm ; 39(3): 422-429, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37324764

RESUMEN

Background: Detecting high-risk arrhythmia is important in diagnosing patients with palpitations. We compared the diagnostic accuracies of 7-day patch-type electrocardiographic (ECG) monitoring and 24-h Holter monitoring for detecting significant arrhythmias in patients with palpitations. Methods: This was a single-center prospective trial with 58 participants who presented with palpitations, chest pain or syncope. Outcomes were defined as the detection of any one of six arrhythmias, including supraventricular tachycardia (SVT), atrial fibrillation or atrial flutter lasting more than 30 s, pauses of more than 3 s, high-degree atrioventricular block, ventricular tachycardia (VT) >3 beats, or polymorphic VT/ventricular fibrillation. The McNemar test for paired proportions was used to compare arrhythmia detection rates. Results: The overall arrhythmia detection rate was higher with 7-day ECG patch monitoring than with 24-h Holter monitoring (34.5% vs. 19.0%, p = .008). Compared with the use of 24-h Holter monitors, the use of 7-day ECG patch monitors was associated with higher detection of SVT (29.3% vs. 13.8%, p = .042). No serious adverse skin reactions were reported among the ECG patch-monitored participants. Conclusions: The results suggest that a 7-day patch-type continuous ECG monitor is more effective for the detection of supraventricular tachycardia than is a 24-h Holter monitor. However, the clinical significance of device detected arrhythmia should be consolidated.

17.
Dev Cell ; 58(19): 1950-1966.e8, 2023 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-37816329

RESUMEN

Newly synthesized proteins in the endoplasmic reticulum (ER) are sorted by coat protein complex II (COPII) at the ER exit site en route to the Golgi. Under cellular stresses, COPII proteins become targets of regulation to control the transport. Here, we show that the COPII outer coat proteins Sec31 and Sec13 are selectively sequestered into the biomolecular condensate of SCOTIN/SHISA-5, which interferes with COPII vesicle formation and inhibits ER-to-Golgi transport. SCOTIN is an ER transmembrane protein with a cytosolic intrinsically disordered region (IDR), which is required and essential for the formation of condensates. Upon IFN-γ stimulation, which is a cellular condition that induces SCOTIN expression and condensation, ER-to-Golgi transport was inhibited in a SCOTIN-dependent manner. Furthermore, cancer-associated mutations of SCOTIN perturb its ability to form condensates and control transport. Together, we propose that SCOTIN impedes the ER-to-Golgi transport through its ability to form biomolecular condensates at the ER membrane.


Asunto(s)
Retículo Endoplásmico , Proteínas de Transporte Vesicular , Proteínas de Transporte Vesicular/metabolismo , Transporte Biológico , Transporte de Proteínas/fisiología , Retículo Endoplásmico/metabolismo , Aparato de Golgi/metabolismo
18.
J Immunol ; 185(7): 4311-8, 2010 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-20810994

RESUMEN

ISG15 is a ubiquitin-like molecule whose expression is induced by type I IFN (IFN-α/ß) or in response to virus or bacterial infection. ISG15 or conjugation of ISG15 to target proteins was reported to play critical roles in the regulation of antiviral responses. IFN restricts replication of hepatitis C virus (HCV). However, molecular mechanism of IFN-α/ß that inhibits HCV replication is not clear yet. In the current study, we demonstrated that replication of HCV was inhibited by overexpression of ISG15 and ISG15-conjugation enzymes in the HCV subgenomic replicon cells. Among various nonstructural proteins of HCV, NS5A was identified as the substrate for ISGylation. Furthermore, protein stability of NS5A was decreased by overexpression of ISG15 or ISG15-conjugating enzymes. The inhibitory effect of ISG15 or ISGylation on NS5A was efficiently blocked by substitution of lysine at 379 residue to arginine within the C-terminal region, suggesting that ISGylation directly controls protein stability of NS5A. Finally, the inhibitory effect of IFN-α/ß on HCV replication was further enhanced by ISGylation, suggesting ISG15 as a therapeutic tool for combined therapy with IFN against HCV.


Asunto(s)
Citocinas/inmunología , Citocinas/metabolismo , Hepacivirus/inmunología , Proteínas no Estructurales Virales/metabolismo , Replicación Viral/fisiología , Animales , Células COS , Chlorocebus aethiops , Hepacivirus/metabolismo , Immunoblotting , Inmunoprecipitación , Interferón Tipo I/inmunología , Interferón Tipo I/metabolismo , Ratones , Reacción en Cadena de la Polimerasa , Ubiquitinas/inmunología , Ubiquitinas/metabolismo , Proteínas no Estructurales Virales/inmunología
19.
Autophagy ; 18(7): 1613-1628, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-34720018

RESUMEN

ABBREVIATIONS: ATG2: autophagy related 2; BECN1: beclin 1; COPII: coat protein II; DMSO: dimethyl sulfoxide; EBSS: Earle's balanced salt solution; EGFP: enhanced green fluorescent protein; ER: endoplasmic reticulum; ERES: ER exit site(s); GFP: green fluorescent protein; H89: H-89 dihydrochloride hydrate; LAMP1: lysosomal-associated membrane protein 1; MAP1LC3/LC3: microtubule-associated protein 1 light chain 3; MTORC1: mechanistic target of rapamycin kinase complex 1; NS5A: nonstructural protein 5A; PIK3C3/VPS34: phosphatidylinositol 3-kinase catalytic subunit type 3; PLA: proximity ligation assay; PtdIns3P: phosphatidylionositol-3-phosphate; RB1CC1/FIP200: RB1 inducible coiled-coil 1; RFP: red fluorescent protein; RPS6KB1/S6K: ribosomal protein S6 kinase B1; SBP: streptavidin binding protein; SEC16A: SEC16 homolog A, endoplasmic reticulum export factor; SEC31A: SEC31 homolog A, COPII coat complex component; siRNA: small interfering RNA; Str: streptavidin; ULK1: unc-51-like autophagy activating kinase 1; VSVG: vesicular stomatitis virus glycoprotein; WIPI2: WD repeat domain, phosphoinositide interacting 2; WT: wild type.


Asunto(s)
Autofagosomas , Autofagia , Autofagosomas/metabolismo , Fosfatidilinositol 3-Quinasas Clase III/metabolismo , Retículo Endoplásmico/metabolismo , Aparato de Golgi/metabolismo , Estreptavidina/metabolismo , Proteínas de Transporte Vesicular/metabolismo
20.
Biochem Biophys Res Commun ; 413(3): 487-93, 2011 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-21910972

RESUMEN

The cellular RIG-I-like receptor (RLR) senses pathogenic RNA molecular patterns and transmits signals for type I interferon (IFN) production. It acts as a center for antiviral responses, and large numbers of RIG-I (retinoic acid inducible gene-I) interacting proteins are identified as signaling regulators. In the present study, we report PRKRIR, a negative regulator of PKR inhibitor, as a novel RIG-I interacting protein. In HEK293FT cells, PRKRIR synergistically enhances type I IFN production mediated by a signal activated- or constitutively active form of RIG-I. The C-terminal domain of the PRKRIR was required for physical interaction and the signal augmentation. The PRKRIR blocks poly-ubiquitination and protein degradation of RIG-I, thereby increasing cellular levels of RIG-I proteins. Furthermore, overexpression of PRKRIR, along with a signal activated- or constitutively active form of RIG-I, efficiently inhibits virus replication in the infected host. In conclusion, PRKRIR provides a novel positive regulator controlling the RIG-I-IFN production system through protein stability control.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , ARN Helicasas DEAD-box/metabolismo , Interferón beta/inmunología , Virus ARN/inmunología , ARN Bicatenario/metabolismo , Replicación Viral/inmunología , Proteínas Adaptadoras Transductoras de Señales/genética , Proteína 58 DEAD Box , ARN Helicasas DEAD-box/genética , Células HEK293 , Humanos , Estabilidad Proteica , Receptores Inmunológicos , Ubiquitinación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA