Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
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
2.
PLoS Pathog ; 9(10): e1003646, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24098117

RESUMEN

Thioredoxin-interacting protein (TXNIP) has multiple functions, including tumor suppression and involvement in cell proliferation and apoptosis. However, its role in the inflammatory process remains unclear. In this report, we demonstrate that Txnip⁻/⁻ mice are significantly more susceptible to lipopolysaccharide (LPS)-induced endotoxic shock. In response to LPS, Txnip⁻/⁻ macrophages produced significantly higher levels of nitric oxide (NO) and inducible nitric oxide synthase (iNOS), and an iNOS inhibitor rescued Txnip⁻/⁻ mice from endotoxic shock-induced death, demonstrating that NO is a major factor in TXNIP-mediated endotoxic shock. This susceptibility phenotype of Txnip⁻/⁻ mice occurred despite reduced IL-1ß secretion due to increased S-nitrosylation of NLRP3 compared to wild-type controls. Taken together, these data demonstrate that TXNIP is a novel molecule that links NO synthesis and NLRP3 inflammasome activation during endotoxic shock.


Asunto(s)
Proteínas Portadoras/metabolismo , Inflamasomas/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Óxido Nítrico/metabolismo , Choque Séptico/metabolismo , Tiorredoxinas/metabolismo , Animales , Proteínas Portadoras/genética , Inflamasomas/genética , Lipopolisacáridos/toxicidad , Ratones , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR , Óxido Nítrico/genética , Óxido Nítrico Sintasa de Tipo II/genética , Choque Séptico/inducido químicamente , Choque Séptico/genética , Tiorredoxinas/genética
3.
Biochem Biophys Res Commun ; 438(2): 264-9, 2013 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-23880345

RESUMEN

The p53 protein plays a central role in cell cycle arrest and apoptosis in response to diverse stress stimuli. Human ecdysoneless (hEcd) is known for its role in stabilizing the p53 protein level and increasing p53-mediated transcription. Here, we report that thioredoxin interacting protein (TXNIP), a member of the tumor suppressor family, interacts with hEcd and decreases MDM2-mediated p53 ubiquitination, leading to p53 stabilization and an increase in p53 activity. The ectopic overexpression of both TXNIP and Ecd increased actinomycin D-mediated cell death in MCF-7 cells, whereas knockdown of TXNIP and Ecd decreased cell death. These results show that TXNIP is a new regulator of the Ecd-MDM2-p53 loop.


Asunto(s)
Proteínas Portadoras/metabolismo , Regulación Neoplásica de la Expresión Génica , Proteína p53 Supresora de Tumor/metabolismo , Apoptosis , Muerte Celular , Dactinomicina/farmacología , Células HCT116 , Células HEK293 , Células HeLa , Humanos , Células MCF-7 , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Tiorredoxinas/metabolismo , Transcripción Genética
5.
Cell Immunol ; 280(1): 1-9, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23246829

RESUMEN

Vitamin-D3 upregulated protein-1 (VDUP1) is a stress response protein. Pseudomonas aeruginosa (P. aeruginosa) infection is a leading cause of death. Mice infected with live P. aeruginosa exhibit significantly decreased VDUP1 expression. However, the function of VDUP1 during P. aeruginosa-induced mouse bacteremic shock is unknown. To address the function of VDUP1 in P. aeruginosa-infected mice, we constructed a bacteremic shock model wherein both wild-type and VDUP1-deficient mice were infected intra-peritoneally with live P. aeruginosa. We found that VDUP1-deficient mice were more resistant to P. aeruginosa-induced bacteremic shock than wild-type mice, as shown by the increased survival, accelerated bacterial clearance and suppression of cytokine overproduction of the VDUP1-deficient mice. VDUP1 promoted the recruitment of neutrophils into the peritoneal cavities of infected mice. VDUP1 impeded the phagocytosis of non-opsonized P. aeruginosa via phosphatidylinositide 3-kinase (PI3K) pathway in macrophages. P. aeruginosa infection induced the generation of reactive oxygen species (ROS), and the increased production of ROS by the peritoneal cells of VDUP1-deficient mice was advantageous in clearing the bacteria. Overall, VDUP1 aggravates bacteremic shock; thus, VDUP1 can be considered a target molecule for the inhibition of P. aeruginosa-induced bacteremic shock.


Asunto(s)
Proteínas Portadoras/fisiología , Peritonitis/fisiopatología , Infecciones por Pseudomonas/fisiopatología , Choque Séptico/fisiopatología , Tiorredoxinas/fisiología , Animales , Apoptosis/fisiología , Proteínas Portadoras/genética , Quimiotaxis de Leucocito/fisiología , Recuento de Colonia Microbiana , Citocinas/biosíntesis , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neutrófilos/patología , Cavidad Peritoneal/microbiología , Cavidad Peritoneal/patología , Peritonitis/microbiología , Fagocitosis/fisiología , Fosfatidilinositol 3-Quinasas/fisiología , Pseudomonas aeruginosa/crecimiento & desarrollo , Pseudomonas aeruginosa/aislamiento & purificación , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/fisiología , Organismos Libres de Patógenos Específicos , Bazo/microbiología , Tiorredoxinas/genética
6.
J Immunol ; 185(7): 3980-9, 2010 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-20826751

RESUMEN

Vitamin D(3) upregulated protein 1 (VDUP1) is a candidate tumor suppressor, the expression of which is dramatically reduced in various tumor tissues. In this study, we found that VDUP1 expression is suppressed during human hepatic carcinogenesis, and mice lacking VDUP1 are much more susceptible to diethylnitrosamine-induced hepatocarcinogenesis compared with wild type mice. VDUP1-deficient tumors proliferated significantly more than wild type tumors and had corresponding changes in the expression of key cell cycle regulatory proteins. In addition, the hepatomitogen-induced response was associated with a considerable increase in the release of TNF-α and subsequent enhancement of NF-κB activation in VDUP1-deficient mice. When cells were treated with TNF-α, the VDUP1 level was markedly reduced, concomitant with elevated NF-κB activation. Furthermore, the overexpression of VDUP1 resulted in the robust suppression of TNF-α-activated NF-κB activity via association with HDAC1 and HDAC3. These results indicate that VDUP1 negatively regulates hepatocarcinogenesis by suppressing TNF-α-induced NF-κB activation.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Proteínas Portadoras/metabolismo , Neoplasias Hepáticas/metabolismo , FN-kappa B/metabolismo , Tiorredoxinas/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Ensayo de Cambio de Movilidad Electroforética , Activación Enzimática/fisiología , Humanos , Inmunohistoquímica , Inmunoprecipitación , Etiquetado Corte-Fin in Situ , Ratones , Ratones Noqueados , Microscopía Fluorescente , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/fisiología
7.
Biochim Biophys Acta ; 1783(5): 838-48, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18062927

RESUMEN

Hypoxia-inducible factor 1alpha (HIF1alpha) is a critical transcriptional factor for inducing tumor metastasis, and stabilized under hypoxia but degraded by von Hippel-Lindau protein (pVHL) under normoxia. For the maximal degradation of HIF1alpha, it must be exported to the cytoplasm via an unidentified transporter. Here, we demonstrate that vitamin D3 up-regulated protein 1 (VDUP1) associates with the beta-domain of pVHL and enhances the interaction between pVHL and HIF1alpha to promote the nuclear export and degradation of HIF1alpha hypoxia-independently. Blocking of VDUP1 translocation either by leptomycin B or by nuclear export signal mutation inhibited the nuclear export of pVHL/HIF1alpha and relieved the destabilization of HIF1alpha. VDUP1 suppressed cell invasiveness and tumor metastasis, which were also recovered by blocking of nuclear export. Taken together, these findings indicate that VDUP1 is a novel tumor suppressor which mediates the nuclear export of pVHL/HIF1alpha complex to destabilize HIF1alpha.


Asunto(s)
Proteínas Portadoras/metabolismo , Núcleo Celular/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Carioferinas/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Transporte Activo de Núcleo Celular , Línea Celular , Células HeLa , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Neoplasias Gástricas/metabolismo , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/metabolismo , Proteína Exportina 1
8.
J Microbiol Biotechnol ; 29(9): 1401-1411, 2019 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-31434362

RESUMEN

Mycobacterial cell walls comprise thick and diverse lipids and glycolipids that act as a permeability barrier to antibiotics or other chemical agents. The use of OH radicals from a non-thermal plasma jet (NTPJ) for the inactivation of mycobacteria in aqueous solution was adopted as a novel approach. Addition of water vapor in a nitrogen plasma jet generated OH radicals, which converted to hydrogen peroxide (H2O2) that inactivated non-pathogenic Mycobacterium smegmatis and pathogenic Mycobacterium tuberculosis H37Rv. A stable plasma plume was obtained from a nitrogen plasma jet with 1.91 W of power, killing Escherichia coli and mycobacteria effectively, whereas addition of catalase decreased the effects of the former. Mycobacteria were more resistant than E. coli to NTPJ treatment. Plasma treatment enhanced intracellular ROS production and upregulation of genes related to ROS stress responses (thiolrelated oxidoreductases, such as SseA and DoxX, and ferric uptake regulator furA). Morphological changes of M. smegmatis and M. tuberculosis H37Rv were observed after 5 min treatment with N2+H2O plasma, but not of pre-incubated sample with catalase. This finding indicates that the bactericidal efficacy of NTPJ is related to the toxicity of OH and H2O2 radicals in cells. Therefore, our study suggests that NTPJ treatment may effectively control pulmonary infections caused by M. tuberculosis and nontuberculous mycobacteria (NTM) such as M. avium or M. abscessus in water.


Asunto(s)
Antibacterianos/farmacología , Radical Hidroxilo/farmacología , Mycobacterium/efectos de los fármacos , Gases em Plasma/farmacología , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Pared Celular/efectos de los fármacos , Pared Celular/metabolismo , Medios de Cultivo/química , Escherichia coli/efectos de los fármacos , Escherichia coli/fisiología , Peróxido de Hidrógeno/análisis , Peróxido de Hidrógeno/farmacología , Viabilidad Microbiana/efectos de los fármacos , Mycobacterium/fisiología , Nitrógeno/análisis , Nitrógeno/farmacología , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/genética , Gases em Plasma/química , Especies Reactivas de Oxígeno/metabolismo , Agua/análisis
9.
Autophagy ; 15(8): 1356-1375, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30774023

RESUMEN

SIRT3 (sirtuin 3), a mitochondrial protein deacetylase, maintains respiratory function, but its role in the regulation of innate immune defense is largely unknown. Herein, we show that SIRT3 coordinates mitochondrial function and macroautophagy/autophagy activation to promote anti-mycobacterial responses through PPARA (peroxisome proliferator activated receptor alpha). SIRT3 deficiency enhanced inflammatory responses and mitochondrial dysfunction, leading to defective host defense and pathological inflammation during mycobacterial infection. Antibody-mediated depletion of polymorphonuclear neutrophils significantly increased protection against mycobacterial infection in sirt3-/- mice. In addition, mitochondrial oxidative stress promoted excessive inflammation induced by Mycobacterium tuberculosis infection in sirt3-/- macrophages. Notably, SIRT3 was essential for the enhancement of PPARA, a key regulator of mitochondrial homeostasis and autophagy activation in the context of infection. Importantly, overexpression of either PPARA or TFEB (transcription factor EB) in sirt3-/- macrophages recovered antimicrobial activity through autophagy activation. Furthermore, pharmacological activation of SIRT3 enhanced antibacterial autophagy and functional mitochondrial pools during mycobacterial infection. Finally, the levels of SIRT3 and PPARA were downregulated and inversely correlated with TNF (tumor necrosis factor) levels in peripheral blood mononuclear cells from tuberculosis patients. Collectively, these data demonstrate a previously unappreciated function of SIRT3 in orchestrating mitochondrial and autophagic functions to promote antimycobacterial responses. Abbreviations: Ab: antibody; BCG: M. bovis Bacillus Calmette-Guérin; Baf-A1: bafilomycin A1; BMDMs: bone marrow-derived macrophages; CFU: colony forming unit; CXCL5: C-X-C motif chemokine ligand 5; EGFP: enhanced green fluorescent protein; ERFP: enhanced red fluorescent protein; FOXO3: forkhead box O3; HC: healthy controls; H&E: haematoxylin and eosin; HKL: honokiol; IHC: immunohistochemistry; IL1B: interleukin 1 beta; IL6: interleukin 6; IL12B: interleukin 12B; MDMs: monocyte-derived macrophages; MMP: mitochondrial membrane potential; Mtb: Mycobacterium tuberculosis; PBMC: peripheral blood mononuclear cells; PBS: phosphate buffered saline; PMN: polymorphonuclear neutrophil; PPARA: peroxisome proliferator activated receptor alpha; ROS: reactive oxygen species; SIRT3: sirtuin 3; TB: tuberculosis; TEM: transmission electron microscopy; TFEB: transcription factor EB; TNF: tumor necrosis factor.


Asunto(s)
Antibacterianos/metabolismo , Autofagia , Mitocondrias/metabolismo , Mycobacterium/metabolismo , Sirtuina 3/metabolismo , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Femenino , Homeostasis , Humanos , Inflamación/patología , Pulmón/microbiología , Pulmón/patología , Pulmón/ultraestructura , Lisosomas/metabolismo , Lisosomas/ultraestructura , Macrófagos/microbiología , Macrófagos/ultraestructura , Masculino , Persona de Mediana Edad , Mitocondrias/ultraestructura , Mycobacterium/ultraestructura , Neutrófilos/patología , Estrés Oxidativo , PPAR alfa/metabolismo , Fagosomas/metabolismo , Fagosomas/ultraestructura , Sirtuina 3/deficiencia , Tuberculosis/sangre , Tuberculosis/microbiología , Tuberculosis/patología , Factor de Necrosis Tumoral alfa/metabolismo
10.
Cell Mol Immunol ; 4(5): 345-51, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17976314

RESUMEN

Vitamin D3 up-regulated protein 1 (VDUP1) is a multifunctional protein involved in maintaining cellular homeostasis. VDUP1 is induced by a variety of stresses. Inversely, VDUP1 is often reduced in various tumor tissues and cell lines. Over-expression of VDUP1 inhibits cell proliferation through cell cycle arrest. VDUP1 interacts with thioredoxin (Trx) and negatively regulates the expression and antioxidant function of Trx which is involved in redox regulation. VDUP1-/- mice are more susceptible to carcinogenesis than wild-type mice and are defective in establishing immune system including the development and function of natural killer cells. Furthermore, VDUP1-/- mice show impaired Kreb cycle-mediated fatty acid utilization. In this review, we have discussed the multifunctional roles of VDUP1 in diverse cellular responses, in particular its relation to proliferation, apoptosis, differentiation, and diseases such as cancer and stress-related diseases.


Asunto(s)
Proteínas Portadoras/fisiología , Diferenciación Celular , Proliferación Celular , Enfermedades Metabólicas/metabolismo , Neoplasias/metabolismo , Tiorredoxinas/fisiología , Animales , Apoptosis , Colecalciferol/metabolismo , Humanos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Ratones , Ratones Mutantes , Oxidación-Reducción
11.
Immune Netw ; 17(2): 77-88, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28458619

RESUMEN

Mitochondria are key organelles involved in energy production, functioning as the metabolic hubs of cells. Recent findings emphasize the emerging role of the mitochondrion as a key intracellular signaling platform regulating innate immune and inflammatory responses. Several mitochondrial proteins and mitochondrial reactive oxygen species have emerged as central players orchestrating the innate immune responses to pathogens and damaging ligands. This review explores our current understanding of the roles played by mitochondria in regulation of innate immunity and inflammatory responses. Recent advances in our understanding of the relationship between autophagy, mitochondria, and inflammasome activation are also briefly discussed. A comprehensive understanding of mitochondrial role in toll-like receptor-mediated innate immune responses and NLRP3 inflammasome complex activation, will facilitate development of novel therapeutics to treat various infectious, inflammatory, and autoimmune disorders.

12.
Curr Med Chem ; 24(9): 898-910, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27915989

RESUMEN

Vitamin D exerts an anti-inflammatory effect in both health and disease. The importance of vitamin D in protection against various inflammatory and metabolic diseases, including inflammatory bowel diseases (IBDs), has been discussed. Considerable data indicate a role for vitamin D in the activation of autophagy, an intracellular renewal system that maintains homeostasis by eliminating large protein aggregates and damaged organelles. Recent studies have demonstrated an intricate interplay between autophagy and the regulation of inflammation, suggesting that autophagy-modulating agents could be used to treat IBDs. This review focuses on the role and mechanisms of vitamin D in autophagy and the regulation of intestinal inflammation. Vitamin D shows promise for the prevention and amelioration of pathologic responses in IBD, an effect that is mediated, at least in part, by the induction and modulation of autophagy.


Asunto(s)
Autofagia , Enfermedades Inflamatorias del Intestino/fisiopatología , Vitamina D/fisiología , Humanos , Inmunidad Innata , Inflamación/fisiopatología , Enfermedades Inflamatorias del Intestino/inmunología
13.
Autophagy ; 13(2): 423-441, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27764573

RESUMEN

Autophagy is an important antimicrobial effector process that defends against Mycobacterium tuberculosis (Mtb), the human pathogen causing tuberculosis (TB). MicroRNAs (miRNAs), endogenous noncoding RNAs, are involved in various biological functions and act as post-transcriptional regulators to target mRNAs. The process by which miRNAs affect antibacterial autophagy and host defense mechanisms against Mtb infections in human monocytes and macrophages is largely uncharacterized. In this study, we show that Mtb significantly induces the expression of MIR144*/hsa-miR-144-5p, which targets the 3'-untranslated region of DRAM2 (DNA damage regulated autophagy modulator 2) in human monocytes and macrophages. Mtb infection downregulated, whereas the autophagy activators upregulated, DRAM2 expression in human monocytes and macrophages by activating AMP-activated protein kinase. In addition, overexpression of MIR144* decreased DRAM2 expression and formation of autophagosomes in human monocytes, whereas inhibition of MIR144* had the opposite effect. Moreover, the levels of MIR144* were elevated, whereas DRAM2 levels were reduced, in human peripheral blood cells and tissues in TB patients, indicating the clinical significance of MIR144* and DRAM2 in human TB. Notably, DRAM2 interacted with BECN1 and UVRAG, essential components of the autophagic machinery, leading to displacement of RUBCN from the BECN1 complex and enhancement of Ptdlns3K activity. Furthermore, MIR144* and DRAM2 were critically involved in phagosomal maturation and enhanced antimicrobial effects against Mtb. Our findings identify a previously unrecognized role of human MIR144* in the inhibition of antibacterial autophagy and the innate host immune response to Mtb. Additionally, these data reveal that DRAM2 is a key coordinator of autophagy activation that enhances antimicrobial activity against Mtb.


Asunto(s)
Antiinfecciosos/farmacología , Autofagia/efectos de los fármacos , Macrófagos/microbiología , Proteínas de la Membrana/metabolismo , MicroARNs/metabolismo , Monocitos/microbiología , Mycobacterium tuberculosis/fisiología , Proteínas Relacionadas con la Autofagia/genética , Proteínas Relacionadas con la Autofagia/metabolismo , Secuencia de Bases , Beclina-1/metabolismo , Células Cultivadas , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Femenino , Humanos , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Macrófagos/patología , Masculino , MicroARNs/genética , Persona de Mediana Edad , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Monocitos/patología , Mycobacterium tuberculosis/efectos de los fármacos , Mycobacterium tuberculosis/crecimiento & desarrollo , Fagosomas/efectos de los fármacos , Fagosomas/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Unión Proteica/efectos de los fármacos , Tuberculosis/genética , Tuberculosis/microbiología , Tuberculosis/patología , Regulación hacia Arriba/efectos de los fármacos
14.
Nat Commun ; 5: 2958, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24389582

RESUMEN

The redox-dependent inhibition of thioredoxin (TRX) by thioredoxin-interacting protein (TXNIP) plays a pivotal role in various cancers and metabolic syndromes. However, the molecular mechanism of this regulation is largely unknown. Here, we present the crystal structure of the TRX-TXNIP complex and demonstrate that the inhibition of TRX by TXNIP is mediated by an intermolecular disulphide interaction resulting from a novel disulphide bond-switching mechanism. Upon binding to TRX, TXNIP undergoes a structural rearrangement that involves switching of a head-to-tail interprotomer Cys63-Cys247 disulphide between TXNIP molecules to an interdomain Cys63-Cys190 disulphide, and the formation of a de novo intermolecular TXNIP Cys247-TRX Cys32 disulphide. This disulphide-switching event unexpectedly results in a domain arrangement of TXNIP that is entirely different from those of other arrestin family proteins. We further show that the intermolecular disulphide bond between TRX and TXNIP dissociates in the presence of high concentrations of reactive oxygen species. This study provides insight into TRX and TXNIP-dependent cellular regulation.


Asunto(s)
Proteínas Portadoras/metabolismo , Disulfuros/metabolismo , Oxidación-Reducción , Especies Reactivas de Oxígeno , Tiorredoxinas/metabolismo , Cristalografía por Rayos X , Humanos , Modelos Moleculares , Estructura Terciaria de Proteína
15.
Cell Metab ; 18(1): 75-85, 2013 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-23823478

RESUMEN

Reactive oxygen species (ROS) are critical determinants of the fate of hematopoietic stem cells (HSCs) and hematopoiesis. Thioredoxin-interacting protein (TXNIP), which is induced by oxidative stress, is a known regulator of intracellular ROS. Txnip(-/-) old mice exhibited elevated ROS levels in hematopoietic cells and showed a reduction in hematopoietic cell population. Loss of TXNIP led to a dramatic reduction of mouse survival under oxidative stress. TXNIP directly regulated p53 protein by interfering with p53- mouse double minute 2 (MDM2) interactions and increasing p53 transcriptional activity. Txnip(-/-) mice showed downregulation of the antioxidant genes induced by p53. Introduction of TXNIP or p53 into Txnip(-/-) bone marrow cells rescued the HSC frequency and greatly increased survival in mice following oxidative stress. Overall, these data indicate that TXNIP is a regulator of p53 and plays a pivotal role in the maintenance of the hematopoietic cells by regulating intracellular ROS during oxidative stress.


Asunto(s)
Proteínas Portadoras/fisiología , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/citología , Estrés Oxidativo/fisiología , Transducción de Señal/fisiología , Tiorredoxinas/fisiología , Proteína p53 Supresora de Tumor/fisiología , Animales , Antioxidantes/fisiología , Células de la Médula Ósea/citología , Células de la Médula Ósea/fisiología , Trasplante de Médula Ósea , Proteínas Portadoras/genética , Células Cultivadas , Células Madre Hematopoyéticas/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Animales , Proteínas Proto-Oncogénicas c-mdm2/fisiología , Especies Reactivas de Oxígeno/metabolismo , Tiorredoxinas/genética , Proteína p53 Supresora de Tumor/deficiencia , Proteína p53 Supresora de Tumor/genética
16.
Int Immunopharmacol ; 10(4): 481-6, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20116458

RESUMEN

NK cells play crucial roles in innate immunity and adaptive immunity. The detailed mechanisms, however, governing NK cell development remains unclear. In this study, we report that YC-1 significantly enhances NK cell populations differentiated from human umbilical cord blood hematopoietic stem cells (HSCs). NK cells increased by YC-1 display both phenotypic and functional features of fully mature NK (mNK) cells, but YC-1 does not affect the activation of mNK cells. YC-1 did not affect cGMP production and phosphorylation of STAT-5 which is essential for IL-15R signaling. On the other hand, YC-1 increased p38 MAPK phosphorylation during NK cell differentiation. Furthermore, p38 inhibitor SB203580 inhibited the differentiation of NK cells enhanced by YC-1. Taken together, these data suggest that YC-1 enhances NK cell differentiation through the activation of p38 MAPK which is involved in NK cell differentiation.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Activadores de Enzimas/farmacología , Células Madre Hematopoyéticas/efectos de los fármacos , Indazoles/farmacología , Células Asesinas Naturales/efectos de los fármacos , Western Blotting , Línea Celular Tumoral , GMP Cíclico/metabolismo , Inhibidores Enzimáticos/farmacología , Sangre Fetal/citología , Citometría de Flujo , Humanos , Imidazoles/farmacología , Indazoles/antagonistas & inhibidores , Fosforilación , Piridinas/farmacología , Receptores de Interleucina-15/efectos de los fármacos , Factor de Transcripción STAT5/biosíntesis , Transducción de Señal/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
17.
Immunol Lett ; 129(2): 78-84, 2010 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-20156484

RESUMEN

The detailed mechanism driving the germinal center (GC) reaction to B cell lymphomagenesis has not been clarified. Thioredoxin interacting protein (TXNIP), also known as vitamin D3 up-regulated protein 1 which is an important tumor repressor, is involved in stress responses, redox regulation, and cellular proliferation. Here, we report that TXNIP has a potential role in the formation of GC in peripheral lymphoid organs where B lymphocytes divide rapidly. First, we compared changes in GC from wild type mice and Txnip(-/-) mice. After immunization, Txnip(-/-) mice exhibited higher expression level of BCL-6 and larger percentage of GC B cells with the reduction in antibody production and plasma cell numbers. In addition, Txnip(-/-) spleens had a much larger population which expressed Ki-67, a marker of cell proliferation, in the red pulp border than WT spleens. Furthermore, the expression of BCL-6 was decreased in TXNIP overexpressing cells and elevated in TXNIP deficient cells. Taken together, we conclude that TXNIP may contribute to the formation of GCs after immunization. During this process, TXNIP suppresses BCL-6 expression.


Asunto(s)
Linfocitos B/inmunología , Proteínas Portadoras/inmunología , Regulación hacia Abajo , Centro Germinal/citología , Centro Germinal/inmunología , Proteínas Proto-Oncogénicas c-bcl-6/inmunología , Animales , Secuencia de Bases , Western Blotting , Proliferación Celular , Citometría de Flujo , Inmunohistoquímica , Masculino , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Plásmidos/genética , Proteínas Proto-Oncogénicas c-bcl-6/genética
18.
Mol Biol Cell ; 19(2): 433-44, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18003981

RESUMEN

Interleukin-18 (IL-18) plays pivotal roles in linking inflammatory immune responses and tumor progression and metastasis, yet the manner in which this occurs remains to be sufficiently clarified. Here we report that hypoxia induces the transcription and secretion of IL-18, which subsequently induces the expression of hypoxia-inducible factor-1alpha (HIF-1alpha). Mechanistically, IL-18 induces HIF-1alpha through the activity of the GTPase Rac1, which inducibly associates with the IL-18 receptor beta (IL-18Rbeta) subunit, via a PI3K-AKT-NF-kappaB-dependent pathway. Importantly, the knockdown of the IL-18Rbeta subunit inhibited IL-18-driven tumor cell metastasis. Collectively, these findings demonstrate a feed-forward pathway in HIF-1alpha-mediated tumor progression, in which the induction of IL-18 by hypoxia or inflammatory cells augments the expression of both HIF-1alpha and tumor cell metastasis.


Asunto(s)
Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Interleucina-18/genética , FN-kappa B/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Animales , Hipoxia de la Célula/efectos de los fármacos , Línea Celular , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Interleucina-18/metabolismo , Interleucina-18/farmacología , Ratones , Metástasis de la Neoplasia , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Receptores de Interleucina-18/metabolismo , Transducción de Señal/efectos de los fármacos , Transcripción Genética/efectos de los fármacos
19.
Mol Cell Biochem ; 298(1-2): 187-94, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17131042

RESUMEN

Glutamate induces cell death by upsetting the cellular redox homeostasis, termed oxidative glutamate toxicity, in a mouse hippocampal cell line, HT22. Extracellular signal-regulated kinases (ERK) 1/2 are known key players in this process. Here we characterized the roles of both MAP kinases and cell cycle regulators in mediating oxidative glutamate toxicity and the neuroprotective mechanisms of curcumin in HT22 cells. c-Jun N-terminal kinase (JNK) and p38 kinase were activated during the glutamate-induced HT22 cell death, but at a later stage than ERK activation. Treatment with a JNK inhibitor, SP600125, or a p38 kinase inhibitor, SB203580, partly attenuated this cell death. Curcumin, a natural inhibitor of JNK signaling, protected the HT22 cells from glutamate-induced death at nanomolar concentrations more efficiently than SP600125. These doses of curcumin affected neither the level of intracellular glutathione nor the level of reactive oxygen species, but inactivated JNK and p38 significantly. Moreover, curcumin markedly upregulated a cell-cycle inhibitory protein, p21cip1, and downregulated cyclin D1 levels, which might help the cell death prevention. Our results suggest that curcumin has a neuroprotective effect against oxidative glutamate toxicity by inhibiting MAP kinase signaling and influencing cell-cycle regulation.


Asunto(s)
Curcumina/farmacología , Ácido Glutámico/toxicidad , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Animales , Antracenos/farmacología , Antioxidantes/metabolismo , Muerte Celular/efectos de los fármacos , Línea Celular , Ciclina D1/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Glutatión/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Ratones , Fármacos Neuroprotectores/farmacología , Estrés Oxidativo/efectos de los fármacos , Fosforilación/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Factor de Transcripción AP-1/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA