Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
PLoS Pathog ; 17(1): e1009111, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33411856

RESUMEN

Antiviral innate immune response to RNA virus infection is supported by Pattern-Recognition Receptors (PRR) including RIG-I-Like Receptors (RLR), which lead to type I interferons (IFNs) and IFN-stimulated genes (ISG) production. Upon sensing of viral RNA, the E3 ubiquitin ligase TNF Receptor-Associated Factor-3 (TRAF3) is recruited along with its substrate TANK-Binding Kinase (TBK1), to MAVS-containing subcellular compartments, including mitochondria, peroxisomes, and the mitochondria-associated endoplasmic reticulum membrane (MAM). However, the regulation of such events remains largely unresolved. Here, we identify TRK-Fused Gene (TFG), a protein involved in the transport of newly synthesized proteins to the endomembrane system via the Coat Protein complex II (COPII) transport vesicles, as a new TRAF3-interacting protein allowing the efficient recruitment of TRAF3 to MAVS and TBK1 following Sendai virus (SeV) infection. Using siRNA and shRNA approaches, we show that TFG is required for virus-induced TBK1 activation resulting in C-terminal IRF3 phosphorylation and dimerization. We further show that the ability of the TRAF3-TFG complex to engage mTOR following SeV infection allows TBK1 to phosphorylate mTOR on serine 2159, a post-translational modification shown to promote mTORC1 signaling. We demonstrate that the activation of mTORC1 signaling during SeV infection plays a positive role in the expression of Viperin, IRF7 and IFN-induced proteins with tetratricopeptide repeats (IFITs) proteins, and that depleting TFG resulted in a compromised antiviral state. Our study, therefore, identifies TFG as an essential component of the RLR-dependent type I IFN antiviral response.


Asunto(s)
Antivirales/metabolismo , Inmunidad Innata/inmunología , Interferón Tipo I/metabolismo , Proteínas/metabolismo , Infecciones por Rhabdoviridae/inmunología , Vías Secretoras , Vesiculovirus/inmunología , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Células HeLa , Humanos , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas/genética , Infecciones por Rhabdoviridae/metabolismo , Infecciones por Rhabdoviridae/virología , Transducción de Señal , Factor 3 Asociado a Receptor de TNF/genética , Factor 3 Asociado a Receptor de TNF/metabolismo , Vesiculovirus/fisiología
2.
Rheumatology (Oxford) ; 61(3): 1265-1275, 2022 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-34115840

RESUMEN

OBJECTIVE: SSc is an autoimmune connective tissue disorder characterized by inflammation and fibrosis. Although constitutive activation of fibroblasts is proposed to be responsible for the fibrotic and inflammatory features of the disease, the underlying mechanism remains elusive, and effective therapeutic targets are still lacking. The aim of this study was to evaluate the role of oxidative stress-induced senescence and its contribution to the pro-fibrotic and pro-inflammatory phenotypes of fibroblasts from SSc patients. METHODS: Dermal fibroblasts were isolated from SSc (n = 13) and healthy (n = 10) donors. Fibroblasts' intracellular and mitochondrial reactive oxygen species (ROS) were determined by flow cytometry. Mitochondrial function was measured by Seahorse XF24 analyser. Fibrotic and inflammatory gene expressions were assessed by qPCR and key pro-inflammatory components of the fibroblasts' secretome (IL-6 and IL-8) were quantified by ELISA. RESULTS: Compared with healthy fibroblasts, SSc fibroblasts displayed higher levels of both intracellular and mitochondrial ROS. Oxidative stress in SSc fibroblasts induced the expression of fibrotic genes and activated the TGF-ß-activated kinase 1 (TAK1)-IκB kinase ß (IKKß)-IFN regulatory factor 5 (IRF5) inflammatory signalling cascade. These cellular responses paralleled the presence of a DNA damage response, a senescence-associated secretory phenotype and a fibrotic response. Treatment of SSc fibroblasts with ROS scavengers reduced their pro-inflammatory secretome production and fibrotic gene expression. CONCLUSIONS: Oxidative stress-induced cellular senescence in SSc fibroblasts underlies their pro-inflammatory and pro-fibrotic phenotypes. Targeting redox imbalance of SSc fibroblasts enhances their in vitro functions and could be of relevance for SSc therapy.


Asunto(s)
Envejecimiento/metabolismo , Fibroblastos/metabolismo , Inflamación/metabolismo , Estrés Oxidativo , Esclerodermia Sistémica/metabolismo , Enfermedades de la Piel/metabolismo , Humanos , Fenotipo
3.
Circ Res ; 122(2): 255-266, 2018 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-29113965

RESUMEN

RATIONALE: Mesenchymal stromal cells (MSCs) are promising therapeutic strategies for coronary artery disease; however, donor-related variability in cell quality is a main cause of discrepancies in preclinical studies. In vitro, MSCs from individuals with coronary artery disease have reduced ability to suppress activated T-cells. The mechanisms underlying the altered immunomodulatory capacity of MSCs in the context of atherosclerosis remain elusive. OBJECTIVE: The aim of this study was to assess the role of mitochondrial dysfunction in the impaired immunomodulatory properties of MSCs from patients with atherosclerosis. METHODS AND RESULTS: Adipose tissue-derived MSCs were isolated from atherosclerotic (n=38) and nonatherosclerotic (n=42) donors. MSCs:CD4+T-cell suppression was assessed in allogeneic coculture systems. Compared with nonatherosclerotic-MSCs, atherosclerotic-MSCs displayed higher levels of both intracellular (P=0.006) and mitochondrial (P=0.03) reactive oxygen species reflecting altered mitochondrial function. The increased mitochondrial reactive oxygen species levels of atherosclerotic-MSCs promoted a phenotypic switch characterized by enhanced glycolysis and an altered cytokine secretion (interleukin-6 P<0.0001, interleukin-8/C-X-C motif chemokine ligand 8 P=0.04, and monocyte chemoattractant protein-1/chemokine ligand 2 P=0.01). Furthermore, treatment of atherosclerotic-MSCs with the reactive oxygen species scavenger N-acetyl-l-cysteine reduced the levels of interleukin-6, interleukin-8/C-X-C motif chemokine ligand 8, and monocyte chemoattractant protein-1/chemokine ligand 2 in the MSC secretome and improved MSCs immunosuppressive capacity (P=0.03). CONCLUSIONS: An impaired mitochondrial function of atherosclerotic-MSCs underlies their altered secretome and reduced immunopotency. Interventions aimed at restoring the mitochondrial function of atherosclerotic-MSCs improve their in vitro immunosuppressive ability and may translate into enhanced therapeutic efficiency.


Asunto(s)
Enfermedad de la Arteria Coronaria/metabolismo , Células Madre Mesenquimatosas/metabolismo , Mitocondrias/metabolismo , Estrés Oxidativo/fisiología , Adulto , Anciano , Aterosclerosis/inmunología , Aterosclerosis/metabolismo , Células Cultivadas , Enfermedad de la Arteria Coronaria/inmunología , Femenino , Humanos , Masculino , Células Madre Mesenquimatosas/inmunología , Persona de Mediana Edad , Mitocondrias/inmunología , Especies Reactivas de Oxígeno/inmunología , Especies Reactivas de Oxígeno/metabolismo , Adulto Joven
4.
FASEB J ; 28(8): 3679-90, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24803542

RESUMEN

Glycogen synthase kinase 3 (GSK-3) is associated with several cellular systems, including immune response. Lithium, a widely used pharmacological treatment for bipolar disorder, is a GSK-3 inhibitor. GSK-3α is the predominant isoform in human neutrophils. In this study, we examined the effect of GSK-3 inhibition on the production of TNF-α by neutrophils. In the murine air pouch model of inflammation, lithium chloride (LiCl) amplified TNF-α release. In lipopolysaccharide-stimulated human neutrophils, GSK-3 inhibitors mimicked the effect of LiCl, each potentiating TNF-α release after 4 h, in a concentration-dependent fashion, by up to a 3-fold increase (ED50 of 1 mM for lithium). LiCl had no significant effect on cell viability. A positive association was revealed between GSK-3 inhibition and prolonged activation of the p38/MNK1/eIF4E pathway of mRNA translation. Using lysine and arginine labeled with stable heavy isotopes followed by quantitative mass spectrometry, we determined that GSK-3 inhibition markedly increases (by more than 3-fold) de novo TNF-α protein synthesis. Our findings shed light on a novel mechanism of control of TNF-α expression in neutrophils with GSK-3 regulating mRNA translation and raise the possibility that lithium could be having a hitherto unforeseen effect on inflammatory diseases.


Asunto(s)
Glucógeno Sintasa Quinasa 3/fisiología , Cloruro de Litio/farmacología , Neutrófilos/metabolismo , Factor de Necrosis Tumoral alfa/biosíntesis , Animales , Células Cultivadas , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Citocinas/biosíntesis , Citocinas/genética , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Humanos , Indoles/farmacología , Inflamación , Lipopolisacáridos/farmacología , Maleimidas/farmacología , Ratones , Modelos Animales , FN-kappa B/metabolismo , Infiltración Neutrófila , Neutrófilos/enzimología , Biosíntesis de Proteínas/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Tejido Subcutáneo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
5.
PLoS Pathog ; 8(7): e1002747, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22792062

RESUMEN

Tumor Necrosis Factor receptor-associated factor-3 (TRAF3) is a central mediator important for inducing type I interferon (IFN) production in response to intracellular double-stranded RNA (dsRNA). Here, we report the identification of Sec16A and p115, two proteins of the ER-to-Golgi vesicular transport system, as novel components of the TRAF3 interactome network. Notably, in non-infected cells, TRAF3 was found associated with markers of the ER-Exit-Sites (ERES), ER-to-Golgi intermediate compartment (ERGIC) and the cis-Golgi apparatus. Upon dsRNA and dsDNA sensing however, the Golgi apparatus fragmented into cytoplasmic punctated structures containing TRAF3 allowing its colocalization and interaction with Mitochondrial AntiViral Signaling (MAVS), the essential mitochondria-bound RIG-I-like Helicase (RLH) adaptor. In contrast, retention of TRAF3 at the ER-to-Golgi vesicular transport system blunted the ability of TRAF3 to interact with MAVS upon viral infection and consequently decreased type I IFN response. Moreover, depletion of Sec16A and p115 led to a drastic disorganization of the Golgi paralleled by the relocalization of TRAF3, which under these conditions was unable to associate with MAVS. Consequently, upon dsRNA and dsDNA sensing, ablation of Sec16A and p115 was found to inhibit IRF3 activation and anti-viral gene expression. Reciprocally, mild overexpression of Sec16A or p115 in Hec1B cells increased the activation of IFNß, ISG56 and NF-κB -dependent promoters following viral infection and ectopic expression of MAVS and Tank-binding kinase-1 (TBK1). In line with these results, TRAF3 was found enriched in immunocomplexes composed of p115, Sec16A and TBK1 upon infection. Hence, we propose a model where dsDNA and dsRNA sensing induces the formation of membrane-bound compartments originating from the Golgi, which mediate the dynamic association of TRAF3 with MAVS leading to an optimal induction of innate immune responses.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Retículo Endoplásmico/metabolismo , Aparato de Golgi/metabolismo , Inmunidad Innata , Factor 3 Asociado a Receptor de TNF/genética , Factor 3 Asociado a Receptor de TNF/metabolismo , Línea Celular , ADN/metabolismo , Perfilación de la Expresión Génica , Proteínas de la Matriz de Golgi , Células HEK293 , Células HeLa , Humanos , Factor 3 Regulador del Interferón/antagonistas & inhibidores , Factor 3 Regulador del Interferón/metabolismo , Interferón beta/biosíntesis , Interferón beta/genética , Mitocondrias/metabolismo , FN-kappa B/genética , FN-kappa B/metabolismo , Regiones Promotoras Genéticas , Proteínas Serina-Treonina Quinasas/metabolismo , Transporte de Proteínas , Proteoma , Interferencia de ARN , ARN Bicatenario/metabolismo , ARN Interferente Pequeño , Proteínas de Unión al ARN , Transducción de Señal , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/metabolismo
6.
Arterioscler Thromb Vasc Biol ; 33(12): 2850-7, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24135021

RESUMEN

OBJECTIVE: Angiotensin II (Ang II) is implicated in processes underlying the development of arterial wall remodeling events, including cellular hypertrophy and inflammation. We previously documented the activation of IκB kinase-ß (IKKß) in Ang II-treated cells, a kinase involved in inflammatory reactions. In light of a study suggesting a role of IKKß in angiogenesis through its effect on the tuberous sclerosis (TSC)1/2-mammalian target of rapamycin complex 1 pathway in cancer cells, we hypothesized that targeting IKKß could reduce arterial remodeling events by affecting both the inflammatory and the growth-promoting response of Ang II. APPROACH AND RESULTS: Treatment of aortic vascular smooth muscle cells with Ang II induced the rapid and sustained phosphorylation of TSC1 on Ser511, which paralleled the activation of effectors of the mammalian target of rapamycin complex 1 pathway. Furthermore, we show that Ser511 of TSC1 acted as a phosphoacceptor site for Ang II-activated IKKß. Consistent with this, the use of different short hairpin RNA constructs targeting IKKß reduced Ang II-induced TSC1, S6 kinase, and eukaryotic translation initiation factor 4E-binding protein 1 phosphorylation and the rate of protein synthesis. Overexpression of TSC1 lacking Ser511 in vascular smooth muscle cells also exerted detrimental effects on the hypertrophic effect of Ang II. Furthermore, the selective IKKß inhibitor N-(6-chloro-7-methoxy-9H-ß-carbolin-8-yl)-2 methylnicotinamide reduced the inflammatory response and dose-dependently diminished Ang II-induced TSC1 phosphorylation and effectors of the mammalian target of rapamycin complex 1 pathway, leading to inhibition of protein synthesis in vitro and in rat arteries in vivo. CONCLUSIONS: Our findings provide new insights into the molecular understanding of the pathological role of Ang II and assist in identifying the beneficial effects of IKKß inhibition for the treatment of cardiovascular diseases.


Asunto(s)
Angiotensina II/farmacología , Proliferación Celular/efectos de los fármacos , Quinasa I-kappa B/metabolismo , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Angiotensina II/administración & dosificación , Animales , Proteínas Portadoras/metabolismo , Células Cultivadas , Relación Dosis-Respuesta a Droga , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Hipertrofia , Quinasa I-kappa B/antagonistas & inhibidores , Quinasa I-kappa B/genética , Mediadores de Inflamación/metabolismo , Infusiones Subcutáneas , Péptidos y Proteínas de Señalización Intracelular , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina , Complejos Multiproteicos/metabolismo , Músculo Liso Vascular/enzimología , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/enzimología , Miocitos del Músculo Liso/patología , Fosfoproteínas/metabolismo , Fosforilación , Biosíntesis de Proteínas/efectos de los fármacos , Interferencia de ARN , Ratas , Ratas Sprague-Dawley , Proteínas Quinasas S6 Ribosómicas/metabolismo , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Factores de Tiempo , Transfección , Proteína 1 del Complejo de la Esclerosis Tuberosa , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
7.
ACS Nano ; 16(12): 21583-21599, 2022 12 27.
Artículo en Inglés | MEDLINE | ID: mdl-36516979

RESUMEN

Drug nanocarriers (NCs) capable of crossing the vascular endothelium and deeply penetrating into dense tissues of the CNS could potentially transform the management of neurological diseases. In the present study, we investigated the interaction of bottle-brush (BB) polymers with different biological barriers in vitro and in vivo and compared it to nanospheres of similar composition. In vitro internalization and permeability assays revealed that BB polymers are not internalized by brain-associated cell lines and translocate much faster across a blood-brain barrier model compared to nanospheres of similar hydrodynamic diameter. These observations performed under static, no-flow conditions were complemented by dynamic assays performed in microvessel arrays on chip and confirmed that BB polymers can escape the vasculature compartment via a paracellular route. BB polymers injected in mice and zebrafish larvae exhibit higher penetration in brain tissues and faster extravasation of microvessels located in the brain compared to nanospheres of similar sizes. The superior diffusivity of BBs in extracellular matrix-like gels combined with their ability to efficiently cross endothelial barriers via a paracellular route position them as promising drug carriers to translocate across the blood-brain barrier and penetrate dense tissue such as the brain, two unmet challenges and ultimate frontiers in nanomedicine.


Asunto(s)
Polímeros , Pez Cebra , Ratones , Animales , Polímeros/metabolismo , Pez Cebra/metabolismo , Barrera Hematoencefálica/metabolismo , Encéfalo/metabolismo , Transporte Biológico
8.
J Biol Chem ; 285(40): 30708-18, 2010 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-20659889

RESUMEN

Activation of NF-κB transcription factors by locally produced angiotensin II (Ang II) is proposed to be involved in chronic inflammatory reactions leading to atherosclerosis development. However, a clear understanding of the signaling cascades coupling the Ang II AT1 receptors to the activation of NF-κB transcription factors is still lacking. Using primary cultured aortic vascular smooth muscle cells, we show that activation of the IKK complex and NF-κB transcription factors by Ang II is regulated by phosphorylation of the catalytic subunit IKKß on serine residues 177 and 181 in the activation T-loop. The use of pharmacological inhibitors against conventional protein kinases C (PKCs), mitogen-activated/extracellular signal-regulated kinase (MEK) 1/2, ribosomal S6 kinase (RSK), and silencing RNA technology targeting PKCα, IKKß subunit, tumor growth factor ß-activating kinase-1 (TAK1), the E3 ubiquitin ligase tumor necrosis factor receptor-associated factor-6 (TRAF6), and RSK isoforms, demonstrates the requirement of two distinct signaling pathway for the phosphorylation of IKKß and the activation of the IKK complex by Ang II. Rapid phosphorylation of IKKß requires a second messenger-dependent pathway composed of PKCα-TRAF6-TAK1, whereas sustained phosphorylation and activation of IKKß requires the MEK1/2-ERK1/2-RSK pathway. Importantly, simultaneously targeting components of these two pathways completely blunts the phosphorylation of IKKß and the proinflammatory effect of the octapeptide. This is the first report demonstrating activation of TAK1 by the AT1R. We propose a model whereby TRAF6-TAK1 and ERK-RSK intracellular pathways independently and sequentially converge to the T-loop phosphorylation for full activation of IKKß, which is an essential step in the proinflammatory activity of Ang II.


Asunto(s)
Quinasa I-kappa B/metabolismo , Sistema de Señalización de MAP Quinasas , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Receptor de Angiotensina Tipo 1/metabolismo , Proteínas Quinasas S6 Ribosómicas 90-kDa/metabolismo , Sistemas de Mensajero Secundario , Factor 6 Asociado a Receptor de TNF/metabolismo , Animales , Aorta/metabolismo , Aorta/patología , Línea Celular , Activación Enzimática/efectos de los fármacos , Humanos , Inflamación/metabolismo , Inflamación/patología , MAP Quinasa Quinasa 1/antagonistas & inhibidores , MAP Quinasa Quinasa 1/metabolismo , MAP Quinasa Quinasa 2/antagonistas & inhibidores , MAP Quinasa Quinasa 2/metabolismo , Quinasas Quinasa Quinasa PAM/antagonistas & inhibidores , Quinasas Quinasa Quinasa PAM/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Modelos Biológicos , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/patología , Fosforilación/efectos de los fármacos , Proteína Quinasa C-alfa/antagonistas & inhibidores , Proteína Quinasa C-alfa/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Ratas
9.
J Biol Chem ; 285(4): 2227-31, 2010 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-19948723

RESUMEN

Vitamin D signaling through its nuclear vitamin D receptor has emerged as a key regulator of innate immunity in humans. Here we show that hormonal vitamin D, 1,25-dihydroxyvitamin D(3), robustly stimulates expression of pattern recognition receptor NOD2/CARD15/IBD1 gene and protein in primary human monocytic and epithelial cells. The vitamin D receptor signals through distal enhancers in the NOD2 gene, whose function was validated by chromatin immunoprecipitation and chromatin conformation capture assays. A key downstream signaling consequence of NOD2 activation by agonist muramyl dipeptide is stimulation of NF-kappaB transcription factor function, which induces expression of the gene encoding antimicrobial peptide defensin beta2 (DEFB2/HBD2). Pretreatment with 1,25-dihydroxyvitamin D(3) synergistically induced NF-kappaB function and expression of genes encoding DEFB2/HBD2 and antimicrobial peptide cathelicidin in the presence of muramyl dipeptide. Importantly, this synergistic response was also seen in macrophages from a donor wild type for NOD2 but was absent in macrophages from patients with Crohn disease homozygous for non-functional NOD2 variants. These studies provide strong molecular links between vitamin D deficiency and the genetics of Crohn disease, a chronic incurable inflammatory bowel condition, as Crohn's pathogenesis is associated with attenuated NOD2 or DEFB2/HBD2 function.


Asunto(s)
Calcitriol/farmacología , Enfermedad de Crohn , Proteína Adaptadora de Señalización NOD2/genética , Deficiencia de Vitamina D , beta-Defensinas/genética , Acetilmuramil-Alanil-Isoglutamina/farmacología , Adyuvantes Inmunológicos/farmacología , Calcitriol/metabolismo , Enfermedad de Crohn/genética , Enfermedad de Crohn/inmunología , Enfermedad de Crohn/metabolismo , Sinergismo Farmacológico , Células Epiteliales/inmunología , Humanos , Macrófagos/inmunología , Macrófagos/metabolismo , Monocitos/inmunología , Monocitos/metabolismo , FN-kappa B/metabolismo , Transducción de Señal/inmunología , Activación Transcripcional/inmunología , Deficiencia de Vitamina D/genética , Deficiencia de Vitamina D/inmunología , Deficiencia de Vitamina D/metabolismo
10.
Cardiovasc Res ; 117(3): 756-766, 2021 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-32339220

RESUMEN

AIMS: Diabetes is a conventional risk factor for atherosclerotic cardiovascular disease and myocardial infarction (MI) is the most common cause of death among these patients. Mesenchymal stromal cells (MSCs) in patients with type 2 diabetes mellitus (T2DM) and atherosclerosis have impaired ability to suppress activated T-cells (i.e. reduced immunopotency). This is mediated by an inflammatory shift in MSC-secreted soluble factors (i.e. pro-inflammatory secretome) and can contribute to the reduced therapeutic effects of autologous T2DM and atherosclerosis-MSC post-MI. The signalling pathways driving the altered secretome of atherosclerosis- and T2DM-MSC are unknown. Specifically, the effect of IκB kinase ß (IKKß) modulation, a key regulator of inflammatory responses, on the immunopotency of MSCs from T2DM patients with advanced atherosclerosis has not been studied. METHODS AND RESULTS: MSCs were isolated from adipose tissue obtained from patients with (i) atherosclerosis and T2DM (atherosclerosis+T2DM MSCs, n = 17) and (ii) atherosclerosis without T2DM (atherosclerosis MSCs, n = 17). MSCs from atherosclerosis+T2DM individuals displayed an inflammatory senescent phenotype and constitutively expressed active forms of effectors of the canonical IKKß nuclear factor-κB transcription factors inflammatory pathway. Importantly, this constitutive pro-inflammatory IKKß signature resulted in an altered secretome and impaired in vitro immunopotency and in vivo healing capacity in an acute MI model. Notably, treatment with a selective IKKß inhibitor or IKKß knockdown (KD) (clustered regularly interspaced short palindromic repeats/Cas9-mediated IKKß KD) in atherosclerosis+T2DM MSCs reduced the production of pro-inflammatory secretome, increased survival, and rescued their immunopotency both in vitro and in vivo. CONCLUSIONS: Constitutively active IKKß reduces the immunopotency of atherosclerosis+T2DM MSC by changing their secretome composition. Modulation of IKKß in atherosclerosis+T2DM MSCs enhances their myocardial repair ability.


Asunto(s)
Aterosclerosis/enzimología , Diabetes Mellitus Tipo 2/enzimología , Quinasa I-kappa B/metabolismo , Mediadores de Inflamación/metabolismo , Células Madre Mesenquimatosas/enzimología , Anciano , Animales , Aterosclerosis/genética , Aterosclerosis/inmunología , Estudios de Casos y Controles , Proliferación Celular , Células Cultivadas , Senescencia Celular , Técnicas de Cocultivo , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/inmunología , Modelos Animales de Enfermedad , Femenino , Humanos , Quinasa I-kappa B/antagonistas & inhibidores , Quinasa I-kappa B/genética , Activación de Linfocitos , Masculino , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/inmunología , Ratones Endogámicos C57BL , Persona de Mediana Edad , Infarto del Miocardio/enzimología , Infarto del Miocardio/inmunología , Infarto del Miocardio/cirugía , Fenotipo , Inhibidores de Proteínas Quinasas/farmacología , Secretoma , Transducción de Señal , Linfocitos T/inmunología , Linfocitos T/metabolismo
11.
Oncogene ; 40(2): 452-464, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33177649

RESUMEN

Interleukin-17 receptor D (IL-17RD), also known as similar expression to Fgf genes (SEF), is proposed to act as a signaling hub that negatively regulates mitogenic signaling pathways, like the ERK1/2 MAP kinase pathway, and innate immune signaling. The expression of IL-17RD is downregulated in certain solid tumors, which has led to the hypothesis that it may exert tumor suppressor functions. However, the role of IL-17RD in tumor biology remains to be studied in vivo. Here, we show that genetic disruption of Il17rd leads to the increased formation of spontaneous tumors in multiple tissues of aging mice. Loss of IL-17RD also promotes tumor development in a model of colitis-associated colorectal cancer, associated with an exacerbated inflammatory response. Colon tumors from IL-17RD-deficient mice are characterized by a strong enrichment in inflammation-related gene signatures, elevated expression of pro-inflammatory tumorigenic cytokines, such as IL-17A and IL-6, and increased STAT3 tyrosine phosphorylation. We further show that RNAi depletion of IL-17RD enhances Toll-like receptor and IL-17A signaling in colon adenocarcinoma cells. No change in the proliferation of normal or tumor intestinal epithelial cells was observed upon genetic inactivation of IL-17RD. Our findings establish IL-17RD as a tumor suppressor in mice and suggest that the protein exerts its function mainly by limiting the extent and duration of inflammation.


Asunto(s)
Carcinogénesis/patología , Colitis/complicaciones , Neoplasias del Colon/patología , Inflamación/complicaciones , Receptores de Interleucina/fisiología , Animales , Carcinogénesis/metabolismo , Proliferación Celular , Neoplasias del Colon/etiología , Neoplasias del Colon/metabolismo , Citocinas/metabolismo , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación , Factor de Transcripción STAT3/metabolismo , Transcriptoma , Tirosina/metabolismo
12.
Cells ; 9(4)2020 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-32272583

RESUMEN

The rapid activation of the type I interferon (IFN) antiviral innate immune response relies on ubiquitously expressed RNA and DNA sensors. Once engaged, these nucleotide-sensing receptors use distinct signaling modules for the rapid and robust activation of mitogen-activated protein kinases (MAPKs), the IκB kinase (IKK) complex, and the IKK-related kinases IKKε and TANK-binding kinase 1 (TBK1), leading to the subsequent activation of the activator protein 1 (AP1), nuclear factor-kappa B (NF-κB), and IFN regulatory factor 3 (IRF3) transcription factors, respectively. They, in turn, induce immunomodulatory genes, allowing for a rapid antiviral cellular response. Unlike the MAPKs, the IKK complex and the IKK-related kinases, ubiquitously expressed glycogen synthase kinase 3 (GSK-3) α and ß isoforms are active in unstimulated resting cells and are involved in the constitutive turnover of ß-catenin, a transcriptional coactivator involved in cell proliferation, differentiation, and lineage commitment. Interestingly, studies have demonstrated the regulatory roles of both GSK-3 and ß-catenin in type I IFN antiviral innate immune response, particularly affecting the activation of IRF3. In this review, we summarize current knowledge on the mechanisms by which GSK-3 and ß-catenin control the antiviral innate immune response to RNA and DNA virus infections.


Asunto(s)
Glucógeno Sintasa Quinasa 3/metabolismo , Inmunidad Innata/genética , Ácidos Nucleicos/metabolismo , beta Catenina/metabolismo , Animales , Humanos , Transducción de Señal
13.
J Virol ; 82(8): 3984-96, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18272581

RESUMEN

The IkappaB kinase-related kinases, TBK1 and IKKi, were recently shown to be responsible for the C-terminal phosphorylation of IRF-3. However, the identity of the phosphoacceptor site(s) targeted by these two kinases remains unclear. Using a biological assay based on the IRF-3-mediated production of antiviral cytokines, we demonstrate here that all Ser/Thr clusters of IRF-3 are required for its optimal transactivation capacity. In vitro kinase assays using full-length His-IRF-3 as a substrate combined with mass spectrometry analysis revealed that serine 402 and serine 396 are directly targeted by TBK1. Analysis of Ser/Thr-to-Ala mutants revealed that the S396A mutation, located in cluster II, abolished IRF-3 homodimerization, CBP association, and nuclear accumulation. However, production of antiviral cytokines was still present in IRF-3 S396A-expressing cells. Interestingly, mutation of serine 339, which is involved in IRF-3 stability, also abrogated CBP association and dimerization without affecting gene transactivation as long as serine 396 remained available for phosphorylation. Complementation of IRF-3-knockout mouse embryonic fibroblasts also revealed a compensatory mechanism of serine 339 and serine 396 in the ability of IRF-3 to induce expression of the interferon-stimulated genes ISG56 and ISG54. These data lead us to reconsider the current model of IRF-3 activation. We propose that conventional biochemical assays used to measure IRF-3 activation are not sensitive enough to detect the small fraction of IRF-3 needed to elicit a biological response. Importantly, our study establishes a molecular link between the role of serine 339 in IRF-3 homodimerization, CBP association, and its destabilization.


Asunto(s)
Factor 3 Regulador del Interferón/metabolismo , Factores de Transcripción p300-CBP/metabolismo , Sustitución de Aminoácidos/genética , Animales , Antivirales/metabolismo , Línea Celular , Núcleo Celular/química , Células Cultivadas , Chlorocebus aethiops , Citocinas/biosíntesis , Dimerización , Fibroblastos , Eliminación de Gen , Prueba de Complementación Genética , Humanos , Factor 3 Regulador del Interferón/genética , Espectrometría de Masas , Ratones , Ratones Noqueados , Mutagénesis Sitio-Dirigida , Fosforilación , Unión Proteica , Proteínas Serina-Treonina Quinasas/metabolismo , Serina/metabolismo , Factores de Transcripción/biosíntesis
14.
Cell Res ; 18(9): 889-99, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19160540

RESUMEN

Over the past four years, the field of the innate immune response has been highly influenced by the discovery of the IkappaB kinase (IKK)-related kinases, TANK Binding Kinase 1 (TBK1) and IKKi, which regulate the activity of interferon regulatory factor (IRF)-3/IRF-7 and NF-kappaB transcription factors. More recently, additional essential components of the signaling pathways that activate these IKK homologues have been discovered. These include the RNA helicases RIGi and MDA5, and the downstream mitochondrial effector known as CARDIF/MAVS/VISA/IPS-1. In addition to their essential functions in controlling the innate immune response, recent studies have highlighted a role of these kinases in cell proliferation and oncogenesis. The canonical IKKs are well recognized to be a bridge linking chronic inflammation to cancer. New findings now suggest that the IKK-related kinases TBK1 and IKKi also participate in signaling pathways that impact on cell transformation and tumor progression. This review will therefore summarize and discuss the role of TBK1 and IKKi in cellular transformation and oncogenesis by focusing on their regulation and substrate specificity.


Asunto(s)
Transformación Celular Neoplásica , Quinasa I-kappa B/metabolismo , Inmunidad Innata/inmunología , Neoplasias/enzimología , Neoplasias/patología , Animales , Humanos , FN-kappa B/metabolismo , Viroterapia Oncolítica , Transducción de Señal
15.
Cytokine ; 43(3): 359-67, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18707898

RESUMEN

Post-translational protein modifications are involved in all functions of living cells. This includes the ability of cells to recognize pathogens and regulate genes involved in their clearance, a concept known as innate immunity. While phosphorylation mechanisms play essential roles in regulating different aspects of the innate immune response, ubiquitination is now recognized as another post-translational modification that works in parallel with phosphorylation to orchestrate the final proper innate immune response against invading pathogens. More precisely, this review will discuss the most recent advances that address the role of ubiquitination in pattern-recognition receptors and type I interferon receptor signaling.


Asunto(s)
Receptor de Interferón alfa y beta/fisiología , Receptores de Reconocimiento de Patrones/fisiología , Transducción de Señal/fisiología , Ubiquitinación/fisiología , Proteínas Adaptadoras Transductoras de Señales/fisiología , Animales , Proteínas de Unión al ADN , Enzima Desubiquitinante CYLD , Humanos , Péptidos y Proteínas de Señalización Intracelular/fisiología , Proteínas de la Membrana/fisiología , Proteínas Nucleares/fisiología , Procesamiento Proteico-Postraduccional/fisiología , Péptidos y Proteínas Asociados a Receptores de Factores de Necrosis Tumoral/fisiología , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa , Proteínas Supresoras de Tumor/fisiología , Ubiquitina-Proteína Ligasas/fisiología
16.
Cardiovasc Res ; 71(1): 61-8, 2006 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-16580653

RESUMEN

OBJECTIVE: Endothelin (ET) modulates cellular processes relevant to vascular remodeling, but there is still some debate as to the potential of ET to be a trophic factor or a mitogen. Moreover, the signaling of ET in vivo to produce these effects is largely unknown. METHODS: 3H-leucine and 3H-thymidine incorporation in rat small mesenteric arteries was studied with several doses of ET-1 (0.1-10 pmol/kg/min) administered for 26 h in vivo. RESULTS: The EC50 for protein synthesis was four times lower than that of DNA synthesis, with maximal effects around 1 and 3 pmol/kg/min, respectively. At 5 pmol/kg/min, ET enhanced CDK2 activity by reducing the binding of its inhibitor p27(Kip1). In contrast, the binding was enhanced at 0.5 pmol/kg/min. The reduced binding observed at 5 pmol/kg/min could not be explained by changes of p27(Kip1) or CDK2 content. Phosphorylation of p27(Kip1) on serine 10 was significantly reduced at 5 pmol/kg/min ET. Although the phosphoinositide 3-kinase pathway was activated, it did not contribute to the protein or DNA synthesis responses. Administration of 1 or 5 pmol/kg/min ET-1 for 28 days increased the thickness and cross-sectional area of the small mesenteric artery due to hypertrophy and hyperplasia, respectively, thus confirming the results obtained in acute conditions. CONCLUSION: ET modulates p27(Kip1) binding to CDK2, producing hypertrophy at low and hyperplasia at higher concentrations. Taken together, these results suggest that ET can act both as a trophic factor and as a mitogen in an in vivo environment, depending on its local concentration.


Asunto(s)
Endotelina-1/farmacología , Endotelio Vascular/metabolismo , Mitógenos/farmacología , Transducción de Señal/efectos de los fármacos , Animales , Ciclo Celular/efectos de los fármacos , Quinasa 2 Dependiente de la Ciclina/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , ADN/biosíntesis , Relación Dosis-Respuesta a Droga , Implantes de Medicamentos , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/patología , Marcaje Isotópico , Leucina/metabolismo , Arterias Mesentéricas , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación , Unión Proteica/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Timidina/metabolismo , Factores de Tiempo , Túnica Íntima/efectos de los fármacos , Túnica Íntima/metabolismo , Túnica Íntima/patología
17.
Mol Cell Biol ; 35(17): 3029-43, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26100021

RESUMEN

Induction of an antiviral innate immune response relies on pattern recognition receptors, including retinoic acid-inducible gene 1-like receptors (RLR), to detect invading pathogens, resulting in the activation of multiple latent transcription factors, including interferon regulatory factor 3 (IRF3). Upon sensing of viral RNA and DNA, IRF3 is phosphorylated and recruits coactivators to induce type I interferons (IFNs) and selected sets of IRF3-regulated IFN-stimulated genes (ISGs) such as those for ISG54 (Ifit2), ISG56 (Ifit1), and viperin (Rsad2). Here, we used wild-type, glycogen synthase kinase 3α knockout (GSK-3α(-/-)), GSK-3ß(-/-), and GSK-3α/ß double-knockout (DKO) embryonic stem (ES) cells, as well as GSK-3ß(-/-) mouse embryonic fibroblast cells in which GSK-3α was knocked down to demonstrate that both isoforms of GSK-3, GSK-3α and GSK-3ß, are required for this antiviral immune response. Moreover, the use of two selective small-molecule GSK-3 inhibitors (CHIR99021 and BIO-acetoxime) or ES cells reconstituted with the catalytically inactive versions of GSK-3 isoforms showed that GSK-3 activity is required for optimal induction of antiviral innate immunity. Mechanistically, GSK-3 isoform activation following Sendai virus infection results in phosphorylation of ß-catenin at S33/S37/T41, promoting IRF3 DNA binding and activation of IRF3-regulated ISGs. This study identifies the role of a GSK-3/ß-catenin axis in antiviral innate immunity.


Asunto(s)
Glucógeno Sintasa Quinasa 3/genética , Virus Sendai/inmunología , Virus de la Estomatitis Vesicular Indiana/inmunología , beta Catenina/genética , Animales , Línea Celular Tumoral , Proteína 58 DEAD Box , ARN Helicasas DEAD-box/inmunología , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3 beta , Células HEK293 , Células HeLa , Humanos , Inmunidad Innata/inmunología , Factor 3 Regulador del Interferón/inmunología , Factor 3 Regulador del Interferón/metabolismo , Interferón Tipo I/biosíntesis , Interferón Tipo I/inmunología , Ratones , Ratones Noqueados , Fosforilación , Interferencia de ARN , ARN Interferente Pequeño , Receptores Inmunológicos , Infecciones por Respirovirus/inmunología , Infecciones por Rhabdoviridae/inmunología , beta Catenina/metabolismo
18.
J Interferon Cytokine Res ; 22(1): 49-58, 2002 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11846975

RESUMEN

Recent molecular, biochemical, and gene disruption studies have demonstrated the essential role of interferon (IFN) regulatory factor-3, (IRF-3) and IRF-7 in the activation of type I IFN gene expression and the induction of the antiviral state. Both transcription factors share structural and functional properties, as well as a common mechanism of activation through C-terminal phosphorylation. The purpose of this review is to summarize recent investigations indicating that similar signalling pathways are likely involved in the activation of IRF-3 and IRF-7. Moreover, unique biochemical events, such as coactivator association and differential recognition of cis-acting elements, also illustrate the capacity of IRF-3 and IRF-7 to selectively regulate type I IFN and IFN-stimulated gene (ISG) expression.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Factores de Transcripción/metabolismo , Secuencia de Aminoácidos , Animales , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/fisiología , Humanos , Factor 3 Regulador del Interferón , Factor 7 Regulador del Interferón , Fosforilación , Estructura Terciaria de Proteína , Elementos de Respuesta , Transducción de Señal , Factores de Transcripción/química , Factores de Transcripción/fisiología , Activación Transcripcional , Virus/patogenicidad
19.
Biochem Pharmacol ; 64(5-6): 985-92, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12213596

RESUMEN

Virus infection of susceptible cells activates multiple signaling pathways that orchestrate the activation of genes, such as cytokines, involved in the antiviral and innate immune response. Among the kinases induced are the mitogen-activated protein (MAP) kinases, Jun-amino terminal kinases (JNK) and p38, the IkappaB kinase (IKK) and DNA-PK. In addition, virus infection also activates an uncharacterized VAK responsible for the C-terminal phosphorylation and subsequent activation of interferon regulatory factor 3 (IRF-3). Virus-mediated activation of IRF-3 through VAK is dependent on viral entry and transcription, since replication deficient virus failed to induce IRF-3 activity. The pathways leading to VAK activation are not well characterized, but IRF-3 appears to represent a novel cellular detection pathway that recognizes viral nucleocapsid (N) structure. Recently, the range of inducers responsible for IRF-3 activation has increased. In addition to virus infection, recognition of bacterial infection mediated through lipopolysaccharide by Toll-like receptor 4 has also been reported. Furthermore, MAP kinase kinase kinase (MAP KKK)-related pathways and DNA-PK induce N-terminal phosphorylation of IRF-3. This review summarizes recent observations in the identification of novel signaling pathways leading to IRF-3 activation.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Transducción de Señal/fisiología , Factores de Transcripción/metabolismo , Animales , Humanos , Factor 3 Regulador del Interferón , Lipopolisacáridos/farmacología , FN-kappa B/metabolismo , Fosforilación , ARN Bicatenario/metabolismo , Estrés Fisiológico/metabolismo , Virosis/metabolismo , Virus/metabolismo
20.
Ann N Y Acad Sci ; 1010: 237-48, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15033728

RESUMEN

The ubiquitously expressed interferon regulatory factor 3 (IRF-3) is directly activated following virus infection and functions as a key activator of the immediate-early Type 1 interferon (IFN) genes. Using DNA microarray analysis (8,556 genes) in Jurkat T cells inducibly expressing constitutively active IRF-3, several target genes directly regulated by IRF-3 were identified. Among the genes upregulated by IRF-3 were transcripts for a subset of known IFN-stimulated genes (ISGs), including ISG56, which functions as an inhibitor of translation initiation. Phosphorylation of C-terminal Ser/Thr residues--(382)GGASSLENTVDLHISNSHPLSLTSDQY(408)-is required for IRF-3 activation. Using C-terminal point mutations and a novel phosphospecific antibody, Ser396 was characterized as the minimal phosphoacceptor site required in vivo for IRF-3 activation following Sendai virus (SeV) infection, expression of viral nucleocapsid, or double-stranded RNA (dsRNA) treatment. The identity of the virus-activated kinase (VAK) activity that targets and activates IRF-3 and IRF-7 has remained a critical missing link in the understanding of interferon signaling. We report that the IKK-related kinases-IKKepsilon/TBK-1-are components of VAK that mediate IRF-3 and IRF-7 phosphorylation and thus functionally link the NF-kappaB and IRF pathways in the development of the antiviral response.


Asunto(s)
Apoptosis/fisiología , Proteínas de Unión al ADN/genética , Interferones/fisiología , FN-kappa B/fisiología , Transducción de Señal/fisiología , Factores de Transcripción/genética , Virus/patogenicidad , Secuencia de Aminoácidos , Humanos , Factor 3 Regulador del Interferón , Células Jurkat , Datos de Secuencia Molecular , Análisis de Secuencia por Matrices de Oligonucleótidos , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA