Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 196
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Immunity ; 43(1): 80-91, 2015 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-26200012

RESUMEN

The orphan nuclear receptor estrogen-related receptor α (ERRα; NR3B1) is a key metabolic regulator, but its function in regulating inflammation remains largely unknown. Here, we demonstrate that ERRα negatively regulates Toll-like receptor (TLR)-induced inflammation by promoting Tnfaip3 transcription and fine-tuning of metabolic reprogramming in macrophages. ERRα-deficient (Esrra(-/-)) mice showed increased susceptibility to endotoxin-induced septic shock, leading to more severe pro-inflammatory responses than control mice. ERRα regulated macrophage inflammatory responses by directly binding the promoter region of Tnfaip3, a deubiquitinating enzyme in TLR signaling. In addition, Esrra(-/-) macrophages showed an increased glycolysis, but impaired mitochondrial respiratory function and biogenesis. Further, ERRα was required for the regulation of NF-κB signaling by controlling p65 acetylation via maintenance of NAD(+) levels and sirtuin 1 activation. These findings unravel a previously unappreciated role for ERRα as a negative regulator of TLR-induced inflammatory responses through inducing Tnfaip3 transcription and controlling the metabolic reprogramming.


Asunto(s)
Cisteína Endopeptidasas/biosíntesis , Inflamación/inmunología , Péptidos y Proteínas de Señalización Intracelular/biosíntesis , Macrófagos/metabolismo , Receptores de Estrógenos/genética , Receptor Toll-Like 4/inmunología , Acetilación , Animales , Calcio/metabolismo , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/metabolismo , Células Cultivadas , Cisteína Endopeptidasas/genética , Activación Enzimática/genética , Glucólisis/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Lipopolisacáridos , Macrófagos/inmunología , Ratones , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/metabolismo , NAD/metabolismo , Fosforilación Oxidativa , Regiones Promotoras Genéticas/genética , Regiones Promotoras Genéticas/inmunología , Choque Séptico/inmunología , Transducción de Señal , Sirtuina 1/metabolismo , Factor 6 Asociado a Receptor de TNF/metabolismo , Factor de Transcripción ReIA/metabolismo , Transcripción Genética/genética , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa , Ubiquitinación , Receptor Relacionado con Estrógeno ERRalfa
2.
Nat Immunol ; 12(8): 742-51, 2011 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-21725320

RESUMEN

The orphan nuclear receptor SHP (small heterodimer partner) is a transcriptional corepressor that regulates hepatic metabolic pathways. Here we identified a role for SHP as an intrinsic negative regulator of Toll-like receptor (TLR)-triggered inflammatory responses. SHP-deficient mice were more susceptible to endotoxin-induced sepsis. SHP had dual regulatory functions in a canonical transcription factor NF-κB signaling pathway, acting as both a repressor of transactivation of the NF-κB subunit p65 and an inhibitor of polyubiquitination of the adaptor TRAF6. SHP-mediated inhibition of signaling via the TLR was mimicked by macrophage-stimulating protein (MSP), a strong inducer of SHP expression, via an AMP-activated protein kinase-dependent signaling pathway. Our data identify a previously unrecognized role for SHP in the regulation of TLR signaling.


Asunto(s)
FN-kappa B/inmunología , Receptores Citoplasmáticos y Nucleares/inmunología , Sepsis/inmunología , Receptores Toll-Like/inmunología , Proteínas Quinasas Activadas por AMP/inmunología , Animales , Inmunoprecipitación de Cromatina , Femenino , Immunoblotting , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal , Factor 6 Asociado a Receptor de TNF/inmunología , Ubiquitinación/inmunología
3.
Semin Cell Dev Biol ; 101: 51-58, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31351226

RESUMEN

Xenophagy is a selective form of autophagy targeting intracellular pathogens for lysosomal degradation. Accordingly, bacteria have evolved multiple strategies to evade or minimize autophagy and xenophagy to survive and replicate in host cells. MicroRNAs (miRNAs) are small non-coding RNA molecules that play key roles in host cells by modulating immune and inflammatory responses during infection. Accumulating evidence shows that miRNAs influence the outcome of bacterial infection by regulating canonical autophagy and xenophagy responses in host cells. Despite recent advances, we are only just beginning to understand the role miRNAs play in autophagy processes and how it affects the outcome of host-pathogen interactions in various bacterial infections. In this review, we focus on how Mycobacteria, Listeria, and Helicobacter evade host protective immune responses using miRNA-dependent mechanisms to suppress autophagy. These efforts include recent insights into the crosstalk between miRNAs and autophagy pathways, and how these interactions may be targeted in the search for new therapeutics against bacterial infections.


Asunto(s)
Autofagia/inmunología , Infecciones Bacterianas/inmunología , Interacciones Huésped-Patógeno/inmunología , MicroARNs/inmunología , Animales , Infecciones Bacterianas/patología , Humanos
4.
Int J Mol Sci ; 23(21)2022 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-36362370

RESUMEN

Sirtuin 1 (SIRT1) regulates cellular processes by deacetylating non-histone targets, including transcription factors and intracellular signalling mediators; thus, its abnormal activation is closely linked to the pathophysiology of several diseases. However, its function in Toxoplasma gondii infection is unclear. We found that SIRT1 contributes to autophagy activation via the AMP-activated protein kinase (AMPK) and PI3K/AKT signalling pathways, promoting anti-Toxoplasma responses. Myeloid-specific Sirt1-/- mice exhibited an increased cyst burden in brain tissue compared to wild-type mice following infection with the avirulent ME49 strain. Consistently, the intracellular survival of T. gondii was markedly increased in Sirt1-deficient bone-marrow-derived macrophages (BMDMs). In contrast, the activation of SIRT1 by resveratrol resulted in not only the induction of autophagy but also a significantly increased anti-Toxoplasma effect. Notably, SIRT1 regulates the FoxO-autophagy axis in several human diseases. Importantly, the T. gondii-induced phosphorylation, acetylation, and cytosolic translocation of FoxO1 was enhanced in Sirt1-deficient BMDMs and the pharmacological inhibition of PI3K/AKT signalling reduced the cytosolic translocation of FoxO1 in BMDMs infected with T. gondii. Further, the CaMKK2-dependent AMPK signalling pathway is responsible for the effect of SIRT1 on the FoxO3a-autophagy axis and for its anti-Toxoplasma activity. Collectively, our findings reveal a previously unappreciated role for SIRT1 in Toxoplasma infection.


Asunto(s)
Toxoplasma , Animales , Humanos , Ratones , Proteínas Quinasas Activadas por AMP/metabolismo , Autofagia , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Sirtuina 1/genética , Toxoplasma/metabolismo , Factores de Transcripción Forkhead/metabolismo
5.
J Korean Med Sci ; 35(38): e343, 2020 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-32989935

RESUMEN

BACKGROUND: Observational studies of the ongoing coronavirus disease 2019 (COVID-19) outbreak suggest that a 'cytokine storm' is involved in the pathogenesis of severe illness. However, the molecular mechanisms underlying the altered pathological inflammation in COVID-19 are largely unknown. We report here that toll-like receptor (TLR) 4-mediated inflammatory signaling molecules are upregulated in peripheral blood mononuclear cells (PBMCs) from COVID-19 patients, compared with healthy controls (HC). METHODS: A total of 48 subjects including 28 COVID-19 patients (8 severe/critical vs. 20 mild/moderate cases) admitted to Chungnam National University Hospital, and age/sex-matched 20 HC were enrolled in this study. PBMCs from the subjects were processed for nCounter Human Immunology gene expression assay to analyze the immune related transcriptome profiles. Recombinant proteins of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) were used to stimulate the PBMCs and monocyte-derived macrophages, and real-time polymerase chain reaction was performed to quantify the mRNA expressions of the pro-inflammatory cytokines/chemokines. RESULTS: Among the most highly increased inflammatory mediators in severe/critically ill patients, S100A9, an alarmin and TLR4 ligand, was found as a noteworthy biomarker, because it inversely correlated with the serum albumin levels. We also observed that recombinant S2 and nucleocapsid proteins of SARS-CoV-2 significantly increased pro-inflammatory cytokines/chemokines and S100A9 in human primary PBMCs. CONCLUSION: These data support a link between TLR4 signaling and pathological inflammation during COVID-19 and contribute to develop therapeutic approaches through targeting TLR4-mediated inflammation.


Asunto(s)
Bacteriemia/etiología , Betacoronavirus , Infecciones por Coronavirus/inmunología , Inflamación/etiología , Neumonía Viral/inmunología , Sepsis/etiología , Receptor Toll-Like 4/fisiología , Adulto , Anciano , Anciano de 80 o más Años , COVID-19 , Diagnóstico Diferencial , Femenino , Humanos , Masculino , Persona de Mediana Edad , Pandemias , SARS-CoV-2 , Transducción de Señal/fisiología , Regulación hacia Arriba
6.
Int J Mol Sci ; 21(13)2020 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-32630319

RESUMEN

The inflammasome is a large intracellular protein complex that activates inflammatory caspase-1 and induces the maturation of interleukin (IL)-1ß and IL-18. Mitophagy plays an essential role in the maintenance of mitochondrial homeostasis during stress. Previous studies have indicated compelling evidence of the crosstalk between inflammasome and mitophagy. Mitophagy regulation of the inflammasome, or vice versa, is crucial for various biological functions, such as controlling inflammation and metabolism, immune and anti-tumor responses, and pyroptotic cell death. Uncontrolled regulation of the inflammasome often results in pathological inflammation and pyroptosis, and causes a variety of human diseases, including metabolic and inflammatory diseases, infection, and cancer. Here, we discuss how improved understanding of the interactions between inflammasome and mitophagy can lead to novel therapies against various disease pathologies, and how the inflammasome-mitophagy connection is currently being targeted pharmacologically by diverse agents and small molecules. A deeper understanding of the inflammasome-mitophagy connection will provide new insights into human health and disease through the balance between mitochondrial clearance and pathology.


Asunto(s)
Inflamasomas/metabolismo , Inflamasomas/fisiología , Mitofagia/fisiología , Animales , Caspasa 1/metabolismo , Enfermedad , Humanos , Inmunidad/inmunología , Inflamación/metabolismo , Inflamación/patología , Interleucina-1beta/metabolismo , Mitocondrias/metabolismo , Mitofagia/genética , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Piroptosis/inmunología , Piroptosis/fisiología
7.
Artículo en Inglés | MEDLINE | ID: mdl-30602512

RESUMEN

ClpC1 is an emerging new target for the treatment of Mycobacterium tuberculosis infections, and several cyclic peptides (ecumicin, cyclomarin A, and lassomycin) are known to act on this target. This study identified another group of peptides, the rufomycins (RUFs), as bactericidal to M. tuberculosis through the inhibition of ClpC1 and subsequent modulation of protein degradation of intracellular proteins. Rufomycin I (RUFI) was found to be a potent and selective lead compound for both M. tuberculosis (MIC, 0.02 µM) and Mycobacterium abscessus (MIC, 0.4 µM). Spontaneously generated mutants resistant to RUFI involved seven unique single nucleotide polymorphism (SNP) mutations at three distinct codons within the N-terminal domain of clpC1 (V13, H77, and F80). RUFI also significantly decreased the proteolytic capabilities of the ClpC1/P1/P2 complex to degrade casein, while having no significant effect on the ATPase activity of ClpC1. This represents a marked difference from ecumicin, which inhibits ClpC1 proteolysis but stimulates the ATPase activity, thereby providing evidence that although these peptides share ClpC1 as a macromolecular target, their downstream effects are distinct, likely due to differences in binding.


Asunto(s)
Proteasas ATP-Dependientes/antagonistas & inhibidores , Antituberculosos/farmacología , Mycobacterium abscessus/efectos de los fármacos , Mycobacterium tuberculosis/efectos de los fármacos , Oligopéptidos/farmacología , Proteínas Bacterianas/antagonistas & inhibidores , Pruebas de Sensibilidad Microbiana , Infecciones por Mycobacterium no Tuberculosas/tratamiento farmacológico , Infecciones por Mycobacterium no Tuberculosas/microbiología , Tuberculosis Pulmonar/tratamiento farmacológico , Tuberculosis Pulmonar/microbiología
8.
J Immunol ; 198(8): 3283-3295, 2017 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-28275133

RESUMEN

The role of peroxisome proliferator-activated receptor α (PPAR-α) in innate host defense is largely unknown. In this study, we show that PPAR-α is essential for antimycobacterial responses via activation of transcription factor EB (TFEB) transcription and inhibition of lipid body formation. PPAR-α deficiency resulted in an increased bacterial load and exaggerated inflammatory responses during mycobacterial infection. PPAR-α agonists promoted autophagy, lysosomal biogenesis, phagosomal maturation, and antimicrobial defense against Mycobacterium tuberculosis or M. bovis bacillus Calmette-Guérin. PPAR-α agonists regulated multiple genes involved in autophagy and lysosomal biogenesis, including Lamp2, Rab7, and Tfeb in bone marrow-derived macrophages. Silencing of TFEB reduced phagosomal maturation and antimicrobial responses, but increased macrophage inflammatory responses during mycobacterial infection. Moreover, PPAR-α activation promoted lipid catabolism and fatty acid ß-oxidation in macrophages during mycobacterial infection. Taken together, our data indicate that PPAR-α mediates antimicrobial responses to mycobacterial infection by inducing TFEB and lipid catabolism.


Asunto(s)
Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/inmunología , Inmunidad Innata/inmunología , Metabolismo de los Lípidos/inmunología , Infecciones por Mycobacterium/inmunología , PPAR alfa/inmunología , Animales , Autofagia/fisiología , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Immunoblotting , Inmunohistoquímica , Gotas Lipídicas/inmunología , Macrófagos/inmunología , Macrófagos/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mycobacterium , PPAR alfa/metabolismo , Reacción en Cadena de la Polimerasa
9.
Molecules ; 24(24)2019 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-31835481

RESUMEN

Mycobacterium abscessus is a rapid-growing, multidrug-resistant, non-tuberculous mycobacterial species responsible for a variety of human infections, such as cutaneous and pulmonary infections. M. abscessus infections are very difficult to eradicate due to the natural and acquired multidrug resistance profiles of M. abscessus. Thus, there is an urgent need for the development of effective drugs or regimens against M. abscessus infections. Here, we report the activity of a US Food and Drug Administration approved drug, thiostrepton, against M. abscessus. We found that thiostrepton significantly inhibited the growth of M. abscessus wild-type strains, subspecies, clinical isolates, and drug-resistant mutants in vitro and in macrophages. In addition, treatment of macrophages with thiostrepton significantly decreased proinflammatory cytokine production in a dose-dependent manner, suggesting an inhibitory effect of thiostrepton on inflammation induced during M. abscessus infection. We further showed that thiostrepton exhibits antimicrobial effects in vivo using a zebrafish model of M. abscessus infection.


Asunto(s)
Antibacterianos/farmacología , Infecciones por Mycobacterium no Tuberculosas/microbiología , Mycobacterium abscessus/efectos de los fármacos , Tioestreptona/farmacología , Animales , Antibacterianos/química , Antibacterianos/uso terapéutico , Línea Celular , Citocinas/biosíntesis , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Humanos , Mediadores de Inflamación/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/metabolismo , Pruebas de Sensibilidad Microbiana , Estructura Molecular , Infecciones por Mycobacterium no Tuberculosas/tratamiento farmacológico , Infecciones por Mycobacterium no Tuberculosas/metabolismo , Mycobacterium abscessus/clasificación , Mycobacterium abscessus/genética , Tioestreptona/uso terapéutico , Pez Cebra
10.
Cell Microbiol ; 19(1)2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27794209

RESUMEN

The fine-tuning of innate immune responses is an important aspect of host defenses against mycobacteria. MicroRNAs (miRNAs), small non-coding RNAs, play essential roles in regulating multiple biological pathways including innate host defenses against various infections. Accumulating evidence shows that many miRNAs regulate the complex interplay between mycobacterial survival strategies and host innate immune pathways. Recent studies have contributed to understanding the role of miRNAs, the levels of which can be modulated by mycobacterial infection, in tuning host autophagy to control bacterial survival and innate effector function. Despite considerable efforts devoted to miRNA profiling over the past decade, further work is needed to improve the selection of appropriate biomarkers for tuberculosis. Understanding the roles and mechanisms of miRNAs in regulating innate immune signaling and autophagy may provide insights into new therapeutic modalities for host-directed anti-mycobacterial therapies. Here, we present a comprehensive review of the recent literature regarding miRNA profiling in tuberculosis and the roles of miRNAs in modulating innate immune responses and autophagy defenses against mycobacterial infections.


Asunto(s)
Autofagia , Regulación de la Expresión Génica , Interacciones Huésped-Patógeno , Inmunidad Innata , MicroARNs/metabolismo , Tuberculosis/inmunología , Tuberculosis/microbiología , Animales , Humanos
11.
Int J Mol Sci ; 19(11)2018 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-30404221

RESUMEN

5'-AMP-activated protein kinase (AMPK) plays diverse roles in various physiological and pathological conditions. AMPK is involved in energy metabolism, which is perturbed by infectious stimuli. Indeed, various pathogens modulate AMPK activity, which affects host defenses against infection. In some viral infections, including hepatitis B and C viral infections, AMPK activation is beneficial, but in others such as dengue virus, Ebola virus, and human cytomegaloviral infections, AMPK plays a detrimental role. AMPK-targeting agents or small molecules enhance the antiviral response and contribute to the control of microbial and parasitic infections. In addition, this review focuses on the double-edged role of AMPK in innate and adaptive immune responses to infection. Understanding how AMPK regulates host defenses will enable development of more effective host-directed therapeutic strategies against infectious diseases.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Interacciones Huésped-Patógeno , Infecciones/etiología , Infecciones/metabolismo , Inmunidad Adaptativa , Animales , Resistencia a la Enfermedad , Interacciones Huésped-Parásitos , Humanos , Inmunidad Innata , Transducción de Señal
12.
Angew Chem Int Ed Engl ; 57(12): 3069-3073, 2018 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-29380472

RESUMEN

The first total syntheses of the bioactive cyclodepsipeptides ohmyungsamycin A and B are described. Key features of our synthesis include the concise preparation of a linear cyclization precursor that consists of N-methyl amides and non-proteinogenic amino acids, and its macrolactamization from a bent conformation. The proposed structure of ohmyungsamycin B was revised based on its synthesis. The cyclic core of the ohmyungsamycins was shown to be responsible for the excellent antituberculosis activity, and ohmyungsamycin variants with truncated chains were evaluated for their biological activity.


Asunto(s)
Péptidos Cíclicos/síntesis química , Modelos Moleculares , Conformación Molecular , Péptidos Cíclicos/química
13.
Artículo en Inglés | MEDLINE | ID: mdl-28674049

RESUMEN

Mycobacterium abscessus is a highly pathogenic drug-resistant rapidly growing mycobacterium. In this study, we evaluated the in vitro, intracellular, and in vivo activities of LCB01-0371, a novel and safe oxazolidinone derivative, for the treatment of M. abscessus infection and compared its resistance to that of other oxazolidinone drugs. LCB01-0371 was effective against several M. abscessus strains in vitro and in a macrophage model of infection. In the murine model, a similar efficacy to linezolid was achieved, especially in the lungs. We induced laboratory-generated resistance to LCB01-0371; sequencing analysis revealed mutations in rplC of T424C and G419A and a nucleotide insertion at the 503 position. Furthermore, LCB01-0371 inhibited the growth of amikacin-, cefoxitin-, and clarithromycin-resistant strains. Collectively, our data indicate that LCB01-0371 might represent a promising new class of oxazolidinones with improved safety, which may replace linezolid for the treatment of M. abscessus.


Asunto(s)
Antibacterianos/uso terapéutico , Infecciones por Mycobacterium no Tuberculosas/tratamiento farmacológico , Mycobacterium abscessus/efectos de los fármacos , Oxazolidinonas/uso terapéutico , Animales , Farmacorresistencia Bacteriana/genética , Bacterias Gramnegativas/efectos de los fármacos , Bacterias Grampositivas/efectos de los fármacos , Humanos , Linezolid/uso terapéutico , Ratones , Ratones Endogámicos C57BL , Pruebas de Sensibilidad Microbiana , Mycobacterium abscessus/aislamiento & purificación
14.
Immunol Cell Biol ; 95(7): 584-592, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28356568

RESUMEN

Inflammasomes are cytosolic multiprotein complexes that cause the release of biologically active interleukin-1ß. The best-characterized inflammasome is the NLRP3 (nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing-3 or Nod-like receptor protein 3) inflammasome. The NLRP3 inflammasome forms an assembly consisting of the ASC (apoptosis-associated speck-like protein containing a C-terminal caspase recruitment domain) adaptor protein and the effector, caspase-1 (cysteine-dependent aspartate-directed protease-1). Numerous agents and ligands derived from pathogens, modified self-cells and the environment induce NLRP3 inflammasome complex formation. NLRP3 inflammasome activation is tightly controlled at the transcriptional and post-translational levels to prevent unwanted excessive inflammation. Recent studies have highlighted the roles and mechanisms of several negative regulators that inhibit the assembly of NLRP3 inflammasome complexes and suppress inflammatory responses. The identification and characterization of new players in the regulation of NLRP3 inflammasome may lead to the development of inflammasome-targeting therapeutics against various inflammatory diseases related to NLRP3 inflammasome-associated pathogenesis.


Asunto(s)
Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Transducción de Señal , Animales , Humanos , Modelos Biológicos
15.
J Immunol ; 194(11): 5355-65, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-25917095

RESUMEN

MicroRNAs (miRNAs) are small noncoding nucleotides that play critical roles in the regulation of diverse biological functions, including the response of host immune cells. Autophagy plays a key role in activating the antimicrobial host defense against Mycobacterium tuberculosis. Although the pathways associated with autophagy must be tightly regulated at a posttranscriptional level, the contribution of miRNAs and whether they specifically influence the activation of macrophage autophagy during M. tuberculosis infection are largely unknown. In this study, we demonstrate that M. tuberculosis infection of macrophages leads to increased expression of miRNA-125a-3p (miR-125a), which targets UV radiation resistance-associated gene (UVRAG), to inhibit autophagy activation and antimicrobial responses to M. tuberculosis. Forced expression of miR-125a significantly blocked M. tuberculosis-induced activation of autophagy and phagosomal maturation in macrophages, and inhibitors of miR-125a counteracted these effects. Both TLR2 and MyD88 were required for biogenesis of miR-125a during M. tuberculosis infection. Notably, activation of the AMP-activated protein kinase significantly inhibited the expression of miR-125a in M. tuberculosis-infected macrophages. Moreover, either overexpression of miR-125a or silencing of UVRAG significantly attenuated the antimicrobial effects of macrophages against M. tuberculosis. Taken together, these data indicate that miR-125a regulates the innate host defense by inhibiting the activation of autophagy and antimicrobial effects against M. tuberculosis through targeting UVRAG.


Asunto(s)
Autofagia/genética , Macrófagos/inmunología , MicroARNs/fisiología , Mycobacterium tuberculosis/inmunología , Tuberculosis Pulmonar/inmunología , Proteínas Supresoras de Tumor/genética , Proteínas Quinasas Activadas por AMP/genética , Proteínas Quinasas Activadas por AMP/inmunología , Animales , Línea Celular , Regulación de la Expresión Génica , Macrófagos/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , MicroARNs/genética , Factor 88 de Diferenciación Mieloide , Interferencia de ARN , ARN Interferente Pequeño , Transducción de Señal/genética , Transducción de Señal/inmunología , Receptor Toll-Like 2 , Tuberculosis Pulmonar/genética
16.
Phytother Res ; 31(1): 152-163, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27859734

RESUMEN

Innate inflammations are dominant causes of poor health and high mortality. The pathogen-associated molecular pattern and lipopolysaccharide (LPS) are sensed by immune cells through activation of toll-like receptor 4 leading to mitogen-activated protein kinases (MAPKs) and NF-κB activations. Controlled MAPK and Nf-κB inhibitors have been proposed as potential antiinflammatory drugs. Withania somnifera is an important medicinal herb with known antiinflammatory activity. In this study, the selected Withania somnifera extracts and withanolides were analysed on LPS-induced macrophages comparatively. Molecular docking analysis revealed withaferin A, withanone and withanolide A as effective withanolides against inflammatory target molecules. In experiments, withaferin A and withanone treatment had prominent suppressions on LPS-induced expression of pro-inflammatory cytokines in bone marrow-derived macrophages. Withaferin A regulated all the major four pathways (MAPKs and NF-κB) involved in innate inflammations. Similarly among the Withania extracts analysed, the in vitro propagated leaf and field grown root extracts containing high withaferin A content suppressed the inflammatory molecules through NF-κB and MAPK pathways. Withaferin A was found to be best in suppressing the activated inflammatory pathways among all the analysed withanolides. Therefore, withaferin A and extracts with high withaferin A content can be used as promising drug candidates against innate inflammations. Copyright © 2016 John Wiley & Sons, Ltd.


Asunto(s)
Receptor Toll-Like 4/metabolismo , Withania/química , Witanólidos/química , Animales , Femenino , Humanos , Ratones , Simulación del Acoplamiento Molecular , Transducción de Señal , Witanólidos/farmacología
17.
Infect Immun ; 84(1): 339-50, 2016 01.
Artículo en Inglés | MEDLINE | ID: mdl-26553469

RESUMEN

The intracellular parasite Toxoplasma gondii has unique dense granule antigens (GRAs) that are crucial for host infection. Emerging evidence suggests that GRA7 of T. gondii is a promising serodiagnostic marker and an effective toxoplasmosis vaccine candidate; however, little is known about the intracellular regulatory mechanisms involved in the GRA7-induced host responses. Here we show that GRA7-induced MyD88 signaling through the activation of TRAF6 and production of reactive oxygen species (ROS) is required for the induction of NF-κB-mediated proinflammatory responses by macrophages. GRA7 stimulation resulted in the rapid activation of mitogen-activated protein kinases and an early burst of ROS in macrophages in a MyD88-dependent manner. GRA7 induced a physical association between GRA7 and TRAF6 via MyD88. Remarkably, the C terminus of GRA7 (GRA7-V) was sufficient for interaction with and ubiquitination of the RING domain of TRAF6, which is capable of inflammatory cytokine production. Interestingly, the generation of ROS and TRAF6 activation are mutually dependent on GRA7/MyD88-mediated signaling in macrophages. Furthermore, mice immunized with GRA7-V showed markedly increased Th1 immune responses and protective efficacy against T. gondii infection. Collectively, these results provide novel insight into the crucial role of GRA7-TRAF6 signaling in innate immune responses.


Asunto(s)
Antígenos de Protozoos/metabolismo , Macrófagos/inmunología , Factor 88 de Diferenciación Mieloide/inmunología , Proteínas Protozoarias/metabolismo , Factor 6 Asociado a Receptor de TNF/metabolismo , Toxoplasma/inmunología , Animales , Anticuerpos Antiprotozoarios/inmunología , Línea Celular , Citocinas/biosíntesis , Activación Enzimática , Células HEK293 , Humanos , Inmunidad Innata/inmunología , Macrófagos/metabolismo , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/genética , NADPH Oxidasa 2 , NADPH Oxidasas/genética , FN-kappa B/inmunología , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/inmunología , Receptor Toll-Like 2/genética , Receptor Toll-Like 4/genética , Toxoplasmosis Animal/inmunología , Toxoplasmosis Animal/parasitología , Ubiquitinación
18.
Biochem Biophys Res Commun ; 477(4): 1072-1077, 2016 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-27416759

RESUMEN

The strongest anaphylatoxin, C5a, plays a critical role in the proinflammatory responses, causing the pathogenesis of a number of inflammatory diseases including sepsis, asthma, and rheumatoid arthritis. Inhibitors of C5a thus have great potential as therapeutics for various inflammatory disorders. Herein, we present the development of a high-affinity repebody against human C5a (hC5a), which effectively suppresses the proinflammatory response. A repebody scaffold composed of leucine-rich repeat (LRR) modules was previously developed as an alternative protein scaffold. A repebody specifically binding to hC5a was selected through a phage display, and its affinity was increased up to 5 nM using modular engineering. The repebody was shown to effectively inhibit the production of C5a-induced proinflammatory cytokines by human monocytes. To obtain insight into a mode of action by the repebody, we determined its crystal structure in complex with hC5a. A structural analysis revealed that the repebody binds to the D1 and D3 regions of hC5a, overlapping several epitope residues with the hC5a receptor (hC5aR). It is thus likely that the repebody suppresses the hC5a-mediated immune response in monocytes by blocking the binding of hC5a to its receptor. The anti-hC5a repebody can be developed as a potential therapeutic for C5a-involved inflammatory diseases.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Complemento C5a/química , Complemento C5a/inmunología , Mediadores de Inflamación/inmunología , Receptor de Anafilatoxina C5a/química , Receptor de Anafilatoxina C5a/inmunología , Anticuerpos Monoclonales/inmunología , Sitios de Unión , Células Cultivadas , Humanos , Factores Inmunológicos/química , Factores Inmunológicos/inmunología , Mediadores de Inflamación/química , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/inmunología , Unión Proteica , Conformación Proteica
19.
J Immunol ; 190(12): 6368-77, 2013 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-23670194

RESUMEN

Intracellular reactive oxygen species (ROS) are essential secondary messengers in many signaling cascades governing innate immunity and cellular functions. TLR3 signaling is crucially involved in antiviral innate and inflammatory responses; however, the roles of ROS in TLR3 signaling remain largely unknown. In this study, we show that TLR3-induced ROS generation is required for the activation of NF-κB, IFN-regulatory factor 3, and STAT1-mediated innate immune responses in macrophages. TLR3 induction led to a rapid increase in ROS generation and a physical association between components of the NADPH oxidase (NOX) enzyme complex (NOX2 and p47(phox)) and TLR3 via a Ca(2+)-c-Src tyrosine kinase-dependent pathway. TLR3-induced ROS generation, NOX2, and p47(phox) were required for the phosphorylation and nuclear translocation of STAT1 and STAT2. TLR3-induced activation of STAT1 contributed to the generation of inflammatory mediators, which was significantly attenuated in NOX2- and p47(phox)-deficient macrophages, suggesting a role for ROS-STAT1 in TLR3-mediated innate immune responses. Collectively, these results provide a novel insight into the crucial role that TLR3-ROS signaling plays in innate immune responses by activating STAT1.


Asunto(s)
Inmunidad Innata/inmunología , Especies Reactivas de Oxígeno/inmunología , Factor de Transcripción STAT1/inmunología , Transducción de Señal/inmunología , Receptor Toll-Like 3/inmunología , Animales , Western Blotting , Células Cultivadas , Femenino , Técnica del Anticuerpo Fluorescente , Inmunoprecipitación , Inflamación/inmunología , Inflamación/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Confocal , Especies Reactivas de Oxígeno/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción STAT1/metabolismo , Receptor Toll-Like 3/metabolismo
20.
Mol Ther ; 22(7): 1254-1265, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24682171

RESUMEN

Interleukin-6 (IL-6) is a multifunctional cytokine that regulates immune responses for host defense and tumorigenic process. Upregulation of IL-6 is known to constitutively phosphorylate signal transducer and activator of transcription 3 (STAT3), leading to activation of multiple oncogene pathways and inflammatory cascade. Here, we present the development of a high-affinity protein binder, termed repebody, which effectively suppresses non-small cell lung cancer in vivo by blocking the IL-6/STAT3 signaling. We selected a repebody that prevents human IL-6 (hIL-6) from binding to its receptor by a competitive immunoassay, and modulated its binding affinity for hIL-6 up to a picomolar range by a modular approach that mimics the combinatorial assembly of diverse modules to form antigen-specific receptors in nature. The resulting repebody was highly specific for hIL-6, effectively inhibiting the STAT3 phosphorylation in a dose- and binding affinity-response manner in vitro. The repebody was shown to have a remarkable suppression effect on the growth of tumors and STAT3 phosphorylation in xenograft mice with non-small cell lung cancer by blocking the hIL-6/STAT3 signaling. Structural analysis of the repebody and IL-6 complex revealed that the repebody binds the site 2a of hIL-6, overlapping a number of epitope residues at site 2a with gp130, and consequently causes a steric hindrance to the formation of IL-6/IL-6Rα complex. Our results suggest that high-affinity repebody targeting the IL-6/STAT3 pathway can be developed as therapeutics for non-small cell lung cancer.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Interleucina-6/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Antineoplásicos/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Femenino , Humanos , Ratones , Ratones Desnudos , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA