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










Base de datos
Intervalo de año de publicación
1.
Clin Transl Med ; 14(3): e1631, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38504554

RESUMEN

BACKGROUND: Cold ischemia-reperfusion injury (IRI) is an unavoidable complication of kidney transplantation. We investigated the role of regulatory T cells (Treg) in cold IRI and whether the interleukin (IL)-2/anti-IL-2 antibody complex (IL-2C) can ameliorate cold IRI. METHODS: We developed a cold IRI mouse model using kidney transplantation and analyzed the IL-2C impact on cold IRI in acute, subacute and chronic phases. RESULTS: Treg transfer attenuated cold IRI, while Treg depletion aggravated cold IRI. Next, IL-2C administration prior to IRI mitigated acute renal function decline, renal tissue damage and apoptosis and inhibited infiltration of effector cells into kidneys and pro-inflammatory cytokine expression on day 1 after IRI. On day 7 after IRI, IL-2C promoted renal regeneration and reduced subacute renal damage. Furthermore, on day 28 following IRI, IL-2C inhibited chronic fibrosis. IL-2C decreased reactive oxygen species-mediated injury and improved antioxidant function. When IL-2C was administered following IRI, it also increased renal regeneration with Treg infiltration and suppressed renal fibrosis. In contrast, Treg depletion in the presence of IL-2C eliminated the positive effects of IL-2C on IRI. CONCLUSION: Tregs protect kidneys from cold IRI and IL-2C inhibited cold IRI by increasing the renal Tregs, suggesting a potential of IL-2C in treating cold IRI. KEY POINTS: Interleukin (IL)-2/anti-IL-2 antibody complex attenuated acute renal injury, facilitated subacute renal regeneration and suppressed chronic renal fibrosis after cold ischemia-reperfusion injury (IRI) by increasing the renal Tregs. IL-2/anti-IL-2 antibody complex decreased reactive oxygen species-mediated injury and improved antioxidant function. This study suggests the therapeutic potential of the IL-2/anti-IL-2 antibody complex in kidney transplantation-associated cold IR.


Asunto(s)
Lesión Renal Aguda , Trasplante de Riñón , Daño por Reperfusión , Animales , Ratones , Interleucina-2/metabolismo , Linfocitos T Reguladores , Complejo Antígeno-Anticuerpo , Trasplante de Riñón/efectos adversos , Antioxidantes/farmacología , Especies Reactivas de Oxígeno/metabolismo , Riñón , Daño por Reperfusión/tratamiento farmacológico , Daño por Reperfusión/metabolismo , Lesión Renal Aguda/tratamiento farmacológico , Lesión Renal Aguda/etiología , Lesión Renal Aguda/metabolismo , Fibrosis
2.
Phytomedicine ; 121: 155133, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37812852

RESUMEN

BACKGROUND: Uveitis is an inflammatory eye condition that threatens vision, and effective anti-inflammatory treatments with minimal side effects are necessary to treat uveitis. PURPOSE: This study aimed to investigate the effects of Lithospermum erythrorhizon Siebold & Zucc. against endotoxin-induced uveitis in rat and mouse models. METHODS: Endotoxin-induced uveitis models of rats and mice were used to evaluate the effects of l. erythrorhizon treatment. Clinical inflammation scores and retinal thickness were assessed in the extract of l. erythrorhizon-treated rats. Histopathological examination revealed inflammatory cell infiltration into the ciliary body. Protein concentration, cellular infiltration, and prostaglandin-E2 levels were measured in the aqueous humor of the extract of l. erythrorhizon-treated rats. Protective effects of l. erythrorhizon on the anterior segment of the eye were examined in mice with endotoxin-induced uveitis. Additionally, we investigated the effect of l. erythrorhizon on the expression of pro-inflammatory cytokines [tumor necrosis factor alpha, interleukin-6, and interleukin-8] in lipopolysaccharide-stimulated THP1 human macrophages and examined the involvement of nuclear factor kappaB/activator protein 1 and interferon regulatory factor signaling pathways. Furthermore, three components of l. erythrorhizon were identified and assessed for their inhibitory effects on LPS-induced inflammation in RAW264.7 macrophage cells. RESULTS: Treatment of the extract of l. erythrorhizon significantly reduced clinical inflammation scores and retinal thickening in rats with endotoxin-induced uveitis. Histopathological examination revealed decreased inflammatory cell infiltration into the ciliary body. The extract of l. erythrorhizon effectively reduced the protein concentration, cellular infiltration, and PG-E2 levels in the aqueous humor of rats with endotoxin-induced uveitis. In mice with endotoxin-induced uveitis, the extract of l. erythrorhizon demonstrated a protective effect on the anterior segment of the eye by reducing inflammation and retinal thickening. The extract of l. erythrorhizon suppressed the expression of pro-inflammatory cytokines (tumor necrosis factor alpha, interleukin-6, and interleukin-8) in lipopolysaccharide-induced inflammation in THP1 human macrophages, by modulating nuclear factor kappaB/activator protein 1 and interferon regulatory factor signaling pathways. Moreover, shikonin, acetylshikonin, and ß, ß-dimethylacryloylshikonin showed dose-dependent inhibition of nitric oxide, tumor necrosis factor alpha and interleukin-6 production in RAW264.7 macrophage cells. CONCLUSION: The extract of l. erythrorhizon is a potential therapeutic agent for uveitis management. Administration of the extract of l. erythrorhizon led to reduced inflammation, retinal thickening, and inflammatory cell infiltration in rat and mouse models of uveitis. The compounds (shikonin, acetylshikonin, and ß, ß-dimethylacryloylshikonin) identified in this study played crucial roles in mediating the anti-inflammatory effects of l. erythrorhizon. These findings indicate that the extract of l. erythrorhizon and its constituent compounds are promising candidates for further research and development of novel treatment modalities for uveitis.


Asunto(s)
Lithospermum , Uveítis , Ratas , Ratones , Humanos , Animales , Endotoxinas/efectos adversos , Lipopolisacáridos/efectos adversos , Interleucina-8/metabolismo , FN-kappa B/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Interleucina-6/metabolismo , Factor de Transcripción AP-1/metabolismo , Uveítis/inducido químicamente , Uveítis/tratamiento farmacológico , Uveítis/patología , Inflamación/tratamiento farmacológico , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Citocinas/metabolismo , Factores Reguladores del Interferón/metabolismo
3.
Biomed Res Int ; 2023: 1317998, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37206297

RESUMEN

This study is aimed at investigating the potential molecular features of allergic rhinitis (AR) and identifying gene signatures and related transcription factors using transcriptome analysis and in silico datasets. Transcriptome profiles were obtained using three independent cohorts (GSE101720, GSE19190, and GSE46171) comprising healthy controls (HC) and patients with AR. The pooled dataset (n = 82) was used to identify the critical signatures of AR compared with HC. Subsequently, key transcription factors were identified by a combined analysis using transcriptome and in silico datasets. Gene ontology: bioprocess (GO: BP) analysis using differentially expressed genes (DEGs) revealed that immune response-related genes were significantly enriched in AR compared with HC. Among them, IL1RL1, CD274, and CD44 were significantly higher in AR patients. We also identified key transcription factors between HC and AR using the in silico dataset and found that AR samples frequently express KLF transcription factor 4 (KLF4), which regulates immune response-related genes including IL1RL1, CD274, and CD44 in human nasal epithelial cells. Our integrative analysis of transcriptomic regulation provides new insights into AR, which may help in developing precision management for patients with AR.


Asunto(s)
Regulación de la Expresión Génica , Inmunidad , Factor 4 Similar a Kruppel , Rinitis Alérgica , Rinitis Alérgica/genética , Rinitis Alérgica/inmunología , Factores de Transcripción/genética , Factores de Transcripción/inmunología , Inmunidad/genética , Inmunidad/inmunología , Factor 4 Similar a Kruppel/genética , Factor 4 Similar a Kruppel/inmunología , Humanos , Regulación de la Expresión Génica/inmunología , Perfilación de la Expresión Génica , Línea Celular
4.
Int J Mol Sci ; 23(18)2022 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-36142314

RESUMEN

In East Asia, the dried root of Lithospermum erythrorhizon has been utilized as an anti-inflammatory, antipyretic, detoxifying, and anti-inflammatory agent. Recently, we reported that L. erythrorhizon protects against allergic rhinitis; however, the component within L. erythrorhizon that exerts antiallergic activity remains unknown. The purpose of the current study was to isolate and characterize the antiallergic active components in an ethanolic extract of L. erythrorhizon roots. We examined the antiallergic effects of L. erythrorhizon reflux ethanol extracts in an ovalbumin (OVA)-induced allergic rhinitis mouse model, and compared the chemical compounds extracted using the hot reflux and cold extraction methods. Chromatographic separation identified two novel anthraquinones, erythrin A and B, one newly discovered compound from the Lithospermum genus, N1″,N3″-dicoumaroylspermidine, and nineteen other recognized compounds. Their chemical structures were elucidated by single (1D) and 2D analysis of nuclear magnetic resonance (NMR) spectroscopic data, as well as high resolution mass spectrometry. Among the identified compounds, N,N'-dicoumaroylspermidine strongly inhibited the release of ß-hexosaminidase, as well as the production of IL-3, IL-4, and IL-13 by IgE-sensitized and BSA-stimulated RBL-2H3 cells. Using the OVA-induced allergic rhinitis mouse model, we showed that N,N'-dicoumaroylspermidine reduced the production of serum OVA-specific IgE and the number of inflammatory cells in nasal lavage fluid. N,N'-dicoumaroylspermidine isolated from L. erythrorhizon exhibits antiallergic properties, making it potentially effective for allergic rhinitis.


Asunto(s)
Antialérgicos , Antipiréticos , Lithospermum , Rinitis Alérgica , Animales , Antraquinonas/farmacología , Antialérgicos/farmacología , Antialérgicos/uso terapéutico , Antipiréticos/farmacología , Citocinas , Modelos Animales de Enfermedad , Etanol/farmacología , Inmunoglobulina E , Interleucina-13/farmacología , Interleucina-3/farmacología , Interleucina-4/farmacología , Mastocitos , Ratones , Ratones Endogámicos BALB C , Ovalbúmina/farmacología , Extractos Vegetales/efectos adversos , Rinitis Alérgica/patología , beta-N-Acetilhexosaminidasas
5.
Exp Eye Res ; 203: 108419, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33383026

RESUMEN

Lithospermum erythrorhizon (L. erythrorhizon), used in traditional medicine, is a potent wound healing, anti-inflammatory and antioxidant plant. However, the effects of L. erythrorhizon on retinal degenerative diseases remain unknown. Here, we explored the protective effects of L. erythrorhizon in in vitro and in vivo retinal degeneration. We found that ethanol extract of L. erythrorhizon (EELE) and the dichloromethane fraction of L. erythrorhizon (MCLE) significantly increased cell viability under glutamate/BSO-induced excitotoxicity/oxidative stress in R28 cells. Treatment with EELE and MCLE reduced the intracellular reactive oxygen species (ROS) and the levels of apoptotic proteins, such as cleaved PARP and cleaved caspase-3. Furthermore, oral administration of EELE and MCLE in an in vivo optic nerve crush mouse model decreased RGC cell death and increased retinal thickness. The major compound between EELE and MCLE was found to be lithospermic acid A (LAA), which has been shown to prevent the elevation of ROS in R28. Therefore, EELE and MCLE have protective effects against the death of retinal cells in vitro and in vivo, and the major compound, LAA, has an antioxidant effect on retinal cells, suggesting that EELE and MCLE could be beneficial agents for retinal degenerative diseases, including glaucoma.


Asunto(s)
Lithospermum/química , Traumatismos del Nervio Óptico/tratamiento farmacológico , Fitoterapia/métodos , Extractos Vegetales/uso terapéutico , Raíces de Plantas/química , Degeneración Retiniana/tratamiento farmacológico , Células Ganglionares de la Retina/efectos de los fármacos , Animales , Proteínas Reguladoras de la Apoptosis/metabolismo , Benzofuranos/farmacología , Técnicas de Cultivo de Célula , Supervivencia Celular/efectos de los fármacos , Cromatografía Líquida de Alta Presión , Depsidos/farmacología , Electroforesis en Gel de Poliacrilamida , Masculino , Ratones , Ratones Endogámicos C57BL , Compresión Nerviosa , Traumatismos del Nervio Óptico/metabolismo , Estrés Oxidativo/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Degeneración Retiniana/metabolismo , Células Ganglionares de la Retina/metabolismo , Tomografía de Coherencia Óptica
6.
Sci Rep ; 9(1): 18440, 2019 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-31804558

RESUMEN

WNT signaling activation in colorectal cancers (CRCs) occurs through APC inactivation or ß-catenin mutations. Both processes promote ß-catenin nuclear accumulation, which up-regulates epithelial-to-mesenchymal transition (EMT). We investigated ß-catenin localization, transcriptome, and phenotypic differences of HCT116 cells containing a wild-type (HCT116-WT) or mutant ß-catenin allele (HCT116-MT), or parental cells with both WT and mutant alleles (HCT116-P). We then analyzed ß-catenin expression and associated phenotypes in CRC tissues. Wild-type ß-catenin showed membranous localization, whereas mutant showed nuclear localization; both nuclear and non-nuclear localization were observed in HCT116-P. Microarray analysis revealed down-regulation of Claudin-7 and E-cadherin in HCT116-MT vs. HCT116-WT. Claudin-7 was also down-regulated in HCT116-P vs. HCT116-WT without E-cadherin dysregulation. We found that ZEB1 is a critical EMT factor for mutant ß-catenin-mediated loss of E-cadherin and Claudin-7 in HCT116-P and HCT116-MT cells. We also demonstrated that E-cadherin binds to both WT and mutant ß-catenin, and loss of E-cadherin releases ß-catenin from the cell membrane and leads to its degradation. Alteration of Claudin-7, as well as both Claudin-7 and E-cadherin respectively caused tight junction (TJ) impairment in HCT116-P, and dual loss of TJs and adherens junctions (AJs) in HCT116-MT. TJ loss increased cell motility, and subsequent AJ loss further up-regulated that. Immunohistochemistry analysis of 101 CRCs revealed high (14.9%), low (52.5%), and undetectable (32.6%) ß-catenin nuclear expression, and high ß-catenin nuclear expression was significantly correlated with overall survival of CRC patients (P = 0.009). Our findings suggest that ß-catenin activation induces EMT progression by modifying cell-cell junctions, and thereby contributes to CRC aggressiveness.


Asunto(s)
Neoplasias Colorrectales/genética , Transición Epitelial-Mesenquimal/genética , Regulación Neoplásica de la Expresión Génica , Vía de Señalización Wnt/genética , beta Catenina/genética , Uniones Adherentes/metabolismo , Uniones Adherentes/patología , Antígenos CD/genética , Antígenos CD/metabolismo , Cadherinas/genética , Cadherinas/metabolismo , Claudinas/genética , Claudinas/metabolismo , Neoplasias Colorrectales/patología , Regulación hacia Abajo , Perfilación de la Expresión Génica , Células HCT116 , Humanos , Mutación , Análisis de Secuencia por Matrices de Oligonucleótidos , Uniones Estrechas/metabolismo , Uniones Estrechas/patología , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/genética , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/metabolismo , beta Catenina/metabolismo
7.
BMB Rep ; 52(2): 133-138, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30078389

RESUMEN

Upon viral infection, the 2', 5'-oligoadenylate synthetase (OAS)-ribonuclease L (RNaseL) system works to cleave viral RNA, thereby blocking viral replication. However, it is unclear whether OAS proteins have a role in regulating gene expression. Here, we show that OAS1 and OAS3 act as negative regulators of the expression of chemokines and interferonresponsive genes in human macrophages. Clustered regularly interspaced short palindromic repeats (CRISPR)-CRISPR-associated protein-9 nuclease (Cas9) technology was used to engineer human myeloid cell lines in which the OAS1 or OAS3 gene was deleted. Neither OAS1 nor OAS3 was exclusively responsible for the degradation of rRNA in macrophages stimulated with poly(I:C), a synthetic surrogate for viral double-stranded (ds)RNA. An mRNA sequencing analysis revealed that genes related to type I interferon signaling and chemokine activity were increased in OAS1-/- and OAS3-/- macrophages treated with intracellular poly(I:C). Indeed, retinoic-acid-inducible gene (RIG)-I- and interferon-induced helicase C domain-containing protein (IFIH1 or MDA5)-mediated induction of chemokines and interferon-stimulated genes was regulated by OAS3, but Toll-like receptor 3 (TLR3)- and TLR4-mediated induction of those genes was modulated by OAS1 in macrophages. However, stimulation of these cells with type I interferons had no effect on OAS1- or OAS3-mediated chemokine secretion. These data suggest that OAS1 and OAS3 negatively regulate the expression of chemokines and interferon-responsive genes in human macrophages. [BMB Reports 2019; 52(2): 133-138].


Asunto(s)
2',5'-Oligoadenilato Sintetasa/genética , Quimiocinas/biosíntesis , Interferones/biosíntesis , Macrófagos/fisiología , 2',5'-Oligoadenilato Sintetasa/metabolismo , Sistemas CRISPR-Cas , Línea Celular , Quimiocinas/genética , Endorribonucleasas/genética , Endorribonucleasas/metabolismo , Regulación de la Expresión Génica , Técnicas de Inactivación de Genes , Humanos , Interferones/genética , Lipopolisacáridos/farmacología , Macrófagos/metabolismo , Poli I-C/biosíntesis , Poli I-C/genética , ARN Bicatenario/genética , ARN Bicatenario/metabolismo , ARN Viral/genética , ARN Viral/metabolismo , Transfección , Replicación Viral/genética
8.
Exp Mol Med ; 50(2): e439, 2018 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-29400702

RESUMEN

During mycobacteria infection, anti-inflammatory responses allow the host to avoid tissue damage caused by overactivation of the immune system; however, little is known about the negative modulators that specifically control mycobacteria-induced immune responses. Here we demonstrate that integrin CD11b is a critical negative regulator of mycobacteria cord factor-induced macrophage-inducible C-type lectin (Mincle) signaling. CD11b deficiency resulted in hyperinflammation following mycobacterial infection. Activation of Mincle by mycobacterial components turns on not only the Syk signaling pathway but also CD11b signaling and induces formation of a Mincle-CD11b signaling complex. The activated CD11b recruits Lyn, SIRPα and SHP1, which dephosphorylate Syk to inhibit Mincle-mediated inflammation. Furthermore, the Lyn activator MLR1023 effectively suppressed Mincle signaling, indicating the possibility of Lyn-mediated control of inflammatory responses. These results describe a new role for CD11b in fine-tuning the immune response against mycobacterium infection.


Asunto(s)
Antígeno CD11b/metabolismo , Lectinas Tipo C/metabolismo , Proteínas de la Membrana/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo , Receptores Inmunológicos/metabolismo , Transducción de Señal , Familia-src Quinasas/metabolismo , Animales , Antígeno CD11b/genética , Adhesión Celular/genética , Adhesión Celular/inmunología , Línea Celular , Citocinas/biosíntesis , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Lectinas Tipo C/genética , Macrófagos/metabolismo , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Complejos Multiproteicos/metabolismo , Infecciones por Mycobacterium/genética , Infecciones por Mycobacterium/metabolismo , Infecciones por Mycobacterium/microbiología , Neutrófilos/inmunología , Neutrófilos/metabolismo , Óxido Nítrico/metabolismo , Unión Proteica , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos
9.
Mol Cells ; 41(3): 214-223, 2018 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-29463066

RESUMEN

Oligoadenylate synthetase (OAS) protein family is the major interferon (IFN)-stimulated genes responsible for the activation of RNase L pathway upon viral infection. OAS-like (OASL) is also required for inhibition of viral growth in human cells, but the loss of one of its mouse homolog, OASL1, causes a severe defect in termination of type I interferon production. To further investigate the antiviral activity of OASL1, we examined its subcellular localization and regulatory roles in IFN production in the early and late stages of viral infection. We found OASL1, but not OASL2, formed stress granules trapping viral RNAs and promoted efficient RLR signaling in early stages of infection. Stress granule formation was dependent on RNA binding activity of OASL1. But in the late stages of infection, OASL1 interacted with IRF7 transcripts to inhibit translation resulting in down regulation of IFN production. These results implicate that OASL1 plays context dependent functions in the antiviral response for the clearance and resolution of viral infections.


Asunto(s)
2',5'-Oligoadenilato Sintetasa/inmunología , Gránulos Citoplasmáticos/inmunología , ARN Viral/metabolismo , Virosis/inmunología , 2',5'-Oligoadenilato Sintetasa/metabolismo , Animales , Gránulos Citoplasmáticos/enzimología , Gránulos Citoplasmáticos/metabolismo , Gránulos Citoplasmáticos/virología , Factor 7 Regulador del Interferón/genética , Factor 7 Regulador del Interferón/inmunología , Factor 7 Regulador del Interferón/metabolismo , Interferones/biosíntesis , Interferones/inmunología , Ratones , Células 3T3 NIH , Transfección , Virosis/metabolismo
10.
Transplantation ; 102(3): 406-416, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-28968355

RESUMEN

BACKGROUND: Macrophages play important roles in xenograft rejection. Here, we investigated whether overexpression of human CD200 or CD47 in porcine endothelial cells (PEC) can suppress macrophages activation in xenogeneic immune responses. METHODS: PECs and human macrophages were incubated together, harvested, and analyzed for in vitro macrophage phagocytic and cytotoxicity activity, and cytokine release. Next, PECs were injected into renal subcapsular space of humanized mice. On day 10 posttransplantation, we analyzed xenograft survival and perigraft inflammatory cell infiltrations in PEC-to-humanized mouse transplantation. RESULTS: PECs highly expressing human CD200, CD47, or both CD47/CD200 were established by lentiviral vector transduction. Both CD200 and CD47 suppressed in vitro macrophage phagocytic and cytotoxic activity against PECs; decreased TNF-α, IL-1ß, and IL-6 secretion; and increased IL-10 secretion. However, simultaneous overexpression of CD200 and CD47 did not show additive effects. Next, PECs were transplanted into NOD-scid IL-2Rg null mice, and human monocytes and lymphocytes were adoptively transferred 1 day after xenotransplantation. PEC xenograft cell death and apoptosis were decreased in the CD200-PEC and CD47/CD200-PEC groups. Perigraft infiltration of human T cells was suppressed by CD47; CD200 suppressed infiltration of human macrophages to a greater extent than CD47; and the CD47/CD200-PEC group exhibited the lowest level of leukocyte infiltration. In summary, overexpression of CD200 in PECs suppressed xenogeneic activation of human macrophages and improved survival of PEC xenografts in humanized mice; however, coexpression of CD200 and CD47 did not show additive effects. CONCLUSIONS: Therefore, overexpression of human CD200 in donor pigs could constitute a promising strategy for overcoming xenograft rejection.


Asunto(s)
Antígenos CD/fisiología , Antígeno CD47/fisiología , Activación de Macrófagos , Trasplante Heterólogo , Animales , Citocinas/biosíntesis , Supervivencia de Injerto , Humanos , Ratones , Porcinos
11.
Biochem Biophys Res Commun ; 493(4): 1491-1497, 2017 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-28988116

RESUMEN

Sepsis is a life-threatening condition caused by an uncontrolled response to bacterial infection. Impaired bactericidal activity in the host is directly associated with severe sepsis; however, the underlying regulatory mechanism(s) is largely unknown. Here, we show that MCL (macrophage C-type lectin) plays a crucial role in killing bacteria during Escherichia coli-induced peritonitis. MCL-deficient mice with E. coli-induced sepsis showed lower survival rates and reduced bacterial clearance when compared with control mice, despite similar levels of proinflammatory cytokine production. Although the ability of macrophages from MCL-deficient mice to kill bacteria was impaired, they showed normal phagocytic activity and production of reactive oxygen species. In addition, MCL-deficient macrophages showed defective phagosome maturation and phagosomal acidification after E. coli infection. Taken together, these results indicate that MCL plays an important role in host defense against E. coli infection by promoting phagosome maturation and acidification, thereby providing new insight into the role of MCL during pathogenesis of sepsis and offering new therapeutic options.


Asunto(s)
Infecciones por Escherichia coli/inmunología , Lectinas Tipo C/inmunología , Macrófagos/inmunología , Proteínas de la Membrana/inmunología , Peritonitis/inmunología , Animales , Infecciones por Escherichia coli/microbiología , Concentración de Iones de Hidrógeno , Inmunidad Innata , Lectinas Tipo C/deficiencia , Lectinas Tipo C/genética , Macrófagos/metabolismo , Macrófagos/microbiología , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Peritonitis/microbiología , Fagocitosis , Fagosomas/inmunología , Fagosomas/metabolismo , Fagosomas/microbiología , Especies Reactivas de Oxígeno/metabolismo , Sepsis/inmunología , Sepsis/microbiología
12.
Microbiol Immunol ; 61(12): 523-530, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28976590

RESUMEN

Trehalose 6,6'-dimycolate (TDM), or cord factor, is a crucial stimulus of immune responses during Mycobacterium tuberculosis infection. Although TDM has immuno-stimulatory properties, including adjuvant activity and the ability to induce granuloma formation, the mechanisms underlying these remain unknown. We hypothesized that TDM stimulates transendothelial migration of neutrophils, which are the first immune cells to infiltrate the tissue upon infection. In this study, it was shown that TDM enhances N-formylmethionyl-leucyl-phenylalanine (fMLP)-induced chemotaxis and transendothelial movement by prolonging AKT phosphorylation in human neutrophils. TDM induced expression of macrophage-inducible C-type lectin, a receptor for TDM, and induced secretion of pro-inflammatory cytokines and chemokines in differentiated HL-60 cells. In 2- and 3-D neutrophil migration assays, TDM-stimulated neutrophils showed increased fMLP-induced chemotaxis and transendothelial migration. Interestingly, following fMLP stimulation of TDM-activated neutrophils, AKT, a crucial kinase for neutrophil polarization and chemotaxis, showed prolonged phosphorylation at serine 473. Taken together, these data suggest that TDM modulates transendothelial migration of neutrophils upon mycobacterial infection through prolonged AKT phosphorylation. AKT may therefore be a promising therapeutic target for enhancing immune responses to mycobacterial infection.


Asunto(s)
Movimiento Celular , Factores Cordón/metabolismo , Mycobacterium tuberculosis/metabolismo , Neutrófilos/citología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Tuberculosis/enzimología , Secuencias de Aminoácidos , Células HL-60 , Interacciones Huésped-Patógeno , Humanos , Mycobacterium tuberculosis/genética , N-Formilmetionina Leucil-Fenilalanina/metabolismo , Neutrófilos/enzimología , Neutrófilos/metabolismo , Proteínas Proto-Oncogénicas c-akt/química , Proteínas Proto-Oncogénicas c-akt/genética , Tuberculosis/genética , Tuberculosis/microbiología , Tuberculosis/fisiopatología
13.
Sci Rep ; 7: 41106, 2017 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-28112221

RESUMEN

Sepsis is a systemic inflammatory response to bacterial infection. The therapeutic options for treating sepsis are limited. Impaired neutrophil recruitment into the infection site is directly associated with severe sepsis, but the precise mechanism is unclear. Here, we show that Mincle plays a key role in neutrophil migration and resistance during polymicrobial sepsis. Mincle-deficient mice exhibited lower survival rates in experimental sepsis from cecal ligation and puncture and Escherichia coli-induced peritonitis. Mincle deficiency led to higher serum inflammatory cytokine levels and reduced bacterial clearance and neutrophil recruitment. Transcriptome analyses revealed that trehalose dimycolate, a Mincle ligand, reduced the expression of G protein-coupled receptor kinase 2 (GRK2) in neutrophils. Indeed, GRK2 expression was upregulated, but surface expression of the chemokine receptor CXCR2 was downregulated in blood neutrophils from Mincle-deficient mice with septic injury. Moreover, CXCL2-mediated adhesion, chemotactic responses, and F-actin polymerization were reduced in Mincle-deficient neutrophils. Finally, we found that fewer Mincle-deficient neutrophils infiltrated from the blood circulation into the peritoneal fluid in bacterial septic peritonitis compared with wild-type cells. Thus, our results indicate that Mincle plays an important role in neutrophil infiltration and suggest that Mincle signaling may provide a therapeutic target for treating sepsis.


Asunto(s)
Coinfección/genética , Lectinas Tipo C/genética , Proteínas de la Membrana/genética , Peritonitis/genética , Sepsis/genética , Animales , Movimiento Celular/genética , Coinfección/microbiología , Factores Cordón/genética , Escherichia coli/patogenicidad , Quinasa 2 del Receptor Acoplado a Proteína-G/genética , Regulación de la Expresión Génica , Humanos , Ratones , Infiltración Neutrófila/genética , Neutrófilos/microbiología , Peritonitis/microbiología , Receptores de Interleucina-8B/genética , Sepsis/microbiología , Transcriptoma/genética
14.
Nat Commun ; 7: 11322, 2016 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-27089465

RESUMEN

In response to persistent mycobacteria infection, the host induces a granuloma, which often fails to eradicate bacteria and results in tissue damage. Diverse host receptors are required to control the formation and resolution of granuloma, but little is known concerning their regulatory interactions. Here we show that Mincle, the inducible receptor for mycobacterial cord factor, is the key switch for the transition of macrophages from cytokine expression to high nitric oxide production. In addition to its stimulatory role on TLR-mediated transcription, Mincle enhanced the translation of key genes required for nitric oxide synthesis through p38 and eIF5A hypusination, leading to granuloma resolution. Thus, Mincle has dual functions in the promotion and subsequent resolution of inflammation during anti-mycobacterial defence using both transcriptional and translational controls.


Asunto(s)
Inflamación/genética , Lectinas Tipo C/genética , Proteínas de la Membrana/genética , Óxido Nítrico/biosíntesis , Biosíntesis de Proteínas/genética , Animales , Línea Celular , Células Cultivadas , Factores Cordón/metabolismo , Factores Cordón/farmacología , Citocinas/metabolismo , Expresión Génica/efectos de los fármacos , Granuloma/genética , Granuloma/metabolismo , Immunoblotting , Inflamación/metabolismo , Lectinas Tipo C/metabolismo , Lisina/análogos & derivados , Lisina/metabolismo , Macrófagos/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mycobacterium tuberculosis/metabolismo , Células 3T3 NIH , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Factores de Iniciación de Péptidos/genética , Factores de Iniciación de Péptidos/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Receptores Toll-Like/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Factor 5A Eucariótico de Iniciación de Traducción
15.
Arterioscler Thromb Vasc Biol ; 35(9): 2020-31, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26229139

RESUMEN

OBJECTIVE: Modulating endothelial progenitor cells (EPCs) is essential for therapeutic angiogenesis, and thus various clinical trials involving EPCs are ongoing. However, the identification of environmental conditions and development of optimal methods are required to accelerate EPC-driven vasculogenesis. APPROACH AND RESULTS: We evaluated gene expression profiles of cord blood-derived EPCs and endothelial cells to identify the key factors in EPC→endothelial cell differentiation and to show that transforming growth factor-ß family members contribute to EPC differentiation. The expression levels of activin receptor-like kinase 1 (ALK1) and its high-affinity ligand, bone morphogenetic protein 9 (BMP9) were markedly changed in EPC→endothelial cell differentiation. Interestingly, BMP9 induced EPC→endothelial cell differentiation and EPC incorporation into vessel-like structures by acting on ALK1 expressed on EPCs in vitro. BMP9 also induced neovascularization in mice with hindlimb ischemia by increasing vessel formation and the incorporation of EPCs into vessels. Conversely, neovascularization was impaired when ALK1 signaling was blocked. Furthermore, EPCs exposed to either short- or long-term BMP9 stimulation demonstrated these functions in EPC-mediated neovascularization. CONCLUSIONS: Collectively, our results indicated that BMP9/ALK1 augmented vasculogenesis and angiogenesis, and thereby enhanced neovascularization. Thus, we suggest that BMP9/ALK1 may improve the efficacy of EPC-based therapies for treating ischemic diseases.


Asunto(s)
Receptores de Activinas Tipo I/genética , Células Progenitoras Endoteliales/patología , Sangre Fetal/citología , Regulación de la Expresión Génica , Factor 2 de Diferenciación de Crecimiento/genética , Isquemia/genética , Neovascularización Patológica/genética , Receptores de Activinas Tipo I/biosíntesis , Receptores de Activinas Tipo II , Animales , Diferenciación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Células Progenitoras Endoteliales/metabolismo , Citometría de Flujo , Factor 2 de Diferenciación de Crecimiento/biosíntesis , Humanos , Isquemia/metabolismo , Isquemia/patología , Masculino , Ratones , Ratones Desnudos , Neovascularización Patológica/sangre , Neovascularización Patológica/patología , ARN/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal
16.
PLoS One ; 9(4): e95805, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24756037

RESUMEN

To avoid excessive activation, immune signals are tightly controlled by diverse inhibitory proteins. TRIM30, a tripartite motif (TRIM)-containing protein is one of such inhibitors known to function in macrophages. To define the roles of TRIM30, we generated Trim30 knockout (Trim30-/-) mice. Trim30 deletion caused no major developmental defects in any organs, nor showed any discernable defect in the activation of macrophages. But, Trim30-/- mice showed increased CD4/CD8 ratio when aged and Trim30-/- CD4+ T cells exhibited an abnormal response upon TCR activation, in particular in the absence of a costimulatory signal. Adoptive transfer of wild-type and Trim30-/- CD4+ T cells together into lymphopenic hosts confirmed higher proliferation of the Trim30-/- CD4+ T cells in vivo. Despite the enhanced proliferation, Trim30-/- T cells showed decreased levels of NF-κB activation and IL-2 production compared to wild-type cells. These results indicate a distinct requirement for TRIM30 in modulation of NF-κB activation and cell proliferation induced by TCR stimulation.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Proteínas Portadoras/genética , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Factores de Edad , Animales , Relación CD4-CD8 , Proteínas Portadoras/metabolismo , Ciclo Celular/genética , Proteínas de Homeodominio/genética , Masculino , Ratones , Ratones Noqueados , FN-kappa B/metabolismo
17.
Mol Cells ; 35(6): 498-513, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23666259

RESUMEN

Lipopolysaccharide (LPS), the major causative agent of bacterial sepsis, has been used by many laboratories in genome-wide expression profiling of the LPS response. However, these studies have predominantly used in vitro cultured macrophages (Macs), which may not accurately reflect the LPS response of these innate immune cells in vivo. To overcome this limitation and to identify inflammatory genes in vivo, we have profiled genome-wide expression patterns in non-lymphoid, splenic myeloid cells extracted directly from LPS-treated mice. Genes encoding factors known to be involved in mediating or regulating inflammatory processes, such as cytokines and chemokines, as well as many genes whose immunological functions are not well known, were strongly induced by LPS after 3 h or 8 h of treatment. Most of the highly LPS-responsive genes that we randomly selected from the microarray data were independently confirmed by quantitative RT-PCR, implying that our microarray data are quite reliable. When our in vivo data were compared to previously reported microarray data for in vitro LPS-treated Macs, a significant proportion (∼20%) of the in vivo LPS-responsive genes defined in this study were specific to cells exposed to LPS in vivo, but a larger proportion of them (∼60%) were influenced by LPS in both in vitro and in vivo settings. This result indicates that our in vivo LPS-responsive gene set includes not only previously identified in vitro LPS-responsive genes but also novel LPS-responsive genes. Both types of genes would be a valuable resource in the future for understanding inflammatory responses in vivo.


Asunto(s)
Macrófagos/inmunología , Células Mieloides/inmunología , Sepsis/genética , Bazo/inmunología , Animales , Células Cultivadas , Quimiocinas/genética , Citocinas/genética , Perfilación de la Expresión Génica , Inmunidad Innata/genética , Inflamación/genética , Lipopolisacáridos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Análisis por Micromatrices
18.
PLoS Pathog ; 8(4): e1002614, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22496642

RESUMEN

Trehalose 6,6'-dimycolate (TDM), a cord factor of Mycobacterium tuberculosis (Mtb), is an important regulator of immune responses during Mtb infections. Macrophages recognize TDM through the Mincle receptor and initiate TDM-induced inflammatory responses, leading to lung granuloma formation. Although various immune cells are recruited to lung granulomas, the roles of other immune cells, especially during the initial process of TDM-induced inflammation, are not clear. In this study, Mincle signaling on neutrophils played an important role in TDM-induced lung inflammation by promoting adhesion and innate immune responses. Neutrophils were recruited during the early stage of lung inflammation following TDM-induced granuloma formation. Mincle expression on neutrophils was required for infiltration of TDM-challenged sites in a granuloma model induced by TDM-coated-beads. TDM-induced Mincle signaling on neutrophils increased cell adherence by enhancing F-actin polymerization and CD11b/CD18 surface expression. The TDM-induced effects were dependent on Src, Syk, and MAPK/ERK kinases (MEK). Moreover, coactivation of the Mincle and TLR2 pathways by TDM and Pam3CSK4 treatment synergistically induced CD11b/CD18 surface expression, reactive oxygen species, and TNFα production by neutrophils. These synergistically-enhanced immune responses correlated with the degree of Mincle expression on neutrophil surfaces. The physiological relevance of the Mincle-mediated anti-TDM immune response was confirmed by defective immune responses in Mincle⁻/⁻ mice upon aerosol infections with Mtb. Mincle-mutant mice had higher inflammation levels and mycobacterial loads than WT mice. Neutrophil depletion with anti-Ly6G antibody caused a reduction in IL-6 and monocyte chemotactic protein-1 expression upon TDM treatment, and reduced levels of immune cell recruitment during the initial stage of infection. These findings suggest a new role of Mincle signaling on neutrophils during anti-mycobacterial responses.


Asunto(s)
Adyuvantes Inmunológicos/efectos adversos , Factores Cordón/efectos adversos , Lectinas Tipo C/metabolismo , Proteínas de la Membrana/metabolismo , Infiltración Neutrófila/efectos de los fármacos , Neutrófilos/metabolismo , Neumonía/inducido químicamente , Neumonía/metabolismo , Transducción de Señal/efectos de los fármacos , Adyuvantes Inmunológicos/química , Adyuvantes Inmunológicos/farmacología , Animales , Antígeno CD11b/genética , Antígeno CD11b/inmunología , Antígeno CD11b/metabolismo , Antígenos CD18/genética , Antígenos CD18/inmunología , Antígenos CD18/metabolismo , Factores Cordón/química , Factores Cordón/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Regulación de la Expresión Génica/inmunología , Granuloma del Sistema Respiratorio/inducido químicamente , Granuloma del Sistema Respiratorio/genética , Granuloma del Sistema Respiratorio/inmunología , Granuloma del Sistema Respiratorio/metabolismo , Granuloma del Sistema Respiratorio/patología , Lectinas Tipo C/genética , Lectinas Tipo C/inmunología , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/patología , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Ratones , Ratones Noqueados , Mycobacterium tuberculosis/química , Mycobacterium tuberculosis/metabolismo , Infiltración Neutrófila/genética , Infiltración Neutrófila/inmunología , Neutrófilos/inmunología , Neutrófilos/patología , Neumonía/genética , Neumonía/inmunología , Neumonía/patología , Proteínas Quinasas/genética , Proteínas Quinasas/inmunología , Proteínas Quinasas/metabolismo , Transducción de Señal/genética , Transducción de Señal/inmunología , Receptor Toll-Like 2/genética , Receptor Toll-Like 2/inmunología , Receptor Toll-Like 2/metabolismo , Tuberculosis Pulmonar/genética , Tuberculosis Pulmonar/inmunología , Tuberculosis Pulmonar/metabolismo , Tuberculosis Pulmonar/patología
19.
PLoS Biol ; 5(9): e238, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17803358

RESUMEN

The activation of several transcription factors is required for the elimination of infectious pathogens via the innate immune response. The transcription factors NF-kappaB, AP-1, and STAT play major roles in the synthesis of immune effector molecules during innate immune responses. However, the fact that these immune responses can have cytotoxic effects requires their tight regulation to achieve restricted and transient activation, and mis-regulation of the damping process has pathological consequences. Here we show that AP-1 and STAT are themselves the major inhibitors responsible for damping NF-kappaB-mediated transcriptional activation during the innate immune response in Drosophila. As the levels of dAP-1 and Stat92E increase due to continuous immune signaling, they play a repressive role by forming a repressosome complex with the Drosophila HMG protein, Dsp1. The dAP-1-, Stat92E-, and Dsp1-containing complexes replace Relish at the promoters of diverse immune effector genes by binding to evolutionarily conserved cis-elements, and they recruit histone deacetylase to inhibit transcription. Reduction by mutation of dAP-1, Stat92E, or Dsp1 results in hyperactivation of Relish target genes and reduces the viability of bacterially infected flies despite more efficient pathogen clearance. These defects are rescued by reducing the Relish copy number, thus confirming that mis-regulation of Relish, not inadequate activation of dAP-1, Stat92E, or Dsp1 target genes, is responsible for the reduced survival of the mutants. We conclude that an inhibitory effect of AP-1 and STAT on NF-kappaB is required for properly balanced immune responses and appears to be evolutionarily conserved.


Asunto(s)
Regulación hacia Abajo , Proteínas de Drosophila/genética , Inmunidad Innata , FN-kappa B/genética , Factores de Transcripción STAT/fisiología , Factor de Transcripción AP-1/fisiología , Animales , Línea Celular , Regulación hacia Abajo/genética , Drosophila , FN-kappa B/antagonistas & inhibidores , Factores de Transcripción STAT/metabolismo , Factor de Transcripción AP-1/metabolismo
20.
Nat Immunol ; 6(2): 211-8, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15640802

RESUMEN

IkappaB kinase (IKK) and Jun N-terminal kinase (Jnk) signaling modules are important in the synthesis of immune effector molecules during innate immune responses against lipopolysaccharide and peptidoglycan. However, the regulatory mechanisms required for specificity and termination of these immune responses are unclear. We show here that crosstalk occurred between the drosophila Jnk and IKK pathways, which led to downregulation of each other's activity. The inhibitory action of Jnk was mediated by binding of drosophila activator protein 1 (AP1) to promoters activated by the transcription factor NF-kappaB. This binding led to recruitment of the histone deacetylase dHDAC1 to the promoter of the gene encoding the antibacterial protein Attacin-A and to local modification of histone acetylation content. Thus, AP1 acts as a repressor by recruiting the deacetylase complex to terminate activation of a group of NF-kappaB target genes.


Asunto(s)
Regulación hacia Abajo/efectos de los fármacos , Drosophila melanogaster/genética , Lipopolisacáridos/farmacología , FN-kappa B/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Transcripción AP-1/metabolismo , Animales , Línea Celular , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Histona Desacetilasas/metabolismo , Regiones Promotoras Genéticas/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...