Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
HGG Adv ; 5(4): 100333, 2024 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-39030910

RESUMEN

Heterozygous de novo or inherited gain-of-function mutations in the MTOR gene cause Smith-Kingsmore syndrome (SKS). SKS is a rare autosomal dominant condition, and individuals with SKS display macrocephaly/megalencephaly, developmental delay, intellectual disability, and seizures. A few dozen individuals are reported in the literature. Here, we report a cohort of 28 individuals with SKS that represent nine MTOR pathogenic variants. We conducted a detailed natural history study and found pathophysiological deficits among individuals with SKS in addition to the common neurodevelopmental symptoms. These symptoms include sleep-wake disturbance, hyperphagia, and hyperactivity, indicative of homeostatic imbalance. To characterize these variants, we developed cell models and characterized their functional consequences. We showed that these SKS variants display a range of mechanistic target of rapamycin (mTOR) activities and respond to the mTOR inhibitor, rapamycin, differently. For example, the R1480_C1483del variant we identified here and the previously known C1483F are more active than wild-type controls and less responsive to rapamycin. Further, we showed that SKS mutations dampened circadian rhythms and low-dose rapamycin improved the rhythm amplitude, suggesting that optimal mTOR activity is required for normal circadian function. As SKS is caused by gain-of-function mutations in MTOR, rapamycin was used to treat several patients. While higher doses of rapamycin caused delayed sleep-wake phase disorder in a subset of patients, optimized lower doses improved sleep. Our study expands the clinical and molecular spectrum of SKS and supports further studies for mechanism-guided treatment options to improve sleep-wake behavior and overall health.

2.
Cell Rep ; 42(1): 111982, 2023 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-36640301

RESUMEN

Cellular circadian clocks direct a daily transcriptional program that supports homeostasis and resilience. Emerging evidence has demonstrated age-associated changes in circadian functions. To define age-dependent changes at the systems level, we profile the circadian transcriptome in the hypothalamus, lung, heart, kidney, skeletal muscle, and adrenal gland in three age groups. We find age-dependent and tissue-specific clock output changes. Aging reduces the number of rhythmically expressed genes (REGs), indicative of weakened circadian control. REGs are enriched for the hallmarks of aging, adding another dimension to our understanding of aging. Analyzing differential gene expression within a tissue at four different times of day identifies distinct clusters of differentially expressed genes (DEGs). Increased variability of gene expression across the day is a common feature of aged tissues. This analysis extends the landscape for understanding aging and highlights the impact of aging on circadian clock function and temporal changes in gene expression.


Asunto(s)
Relojes Circadianos , Transcriptoma , Masculino , Animales , Ratones , Transcriptoma/genética , Ritmo Circadiano/genética , Relojes Circadianos/genética , Hipotálamo , Envejecimiento/genética , Envejecimiento/metabolismo
3.
Sleep ; 46(2)2023 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-36462188

RESUMEN

STUDY OBJECTIVES: Genetics impacts sleep, yet, the molecular mechanisms underlying sleep regulation remain elusive. In this study, we built machine learning models to predict sleep genes based on their similarity to genes that are known to regulate sleep. METHODS: We trained a prediction model on thousands of published datasets, representing circadian, immune, sleep deprivation, and many other processes, using a manually curated list of 109 sleep genes. RESULTS: Our predictions fit with prior knowledge of sleep regulation and identified key genes and pathways to pursue in follow-up studies. As an example, we focused on the NF-κB pathway and showed that chronic activation of NF-κB in a genetic mouse model impacted the sleep-wake patterns. CONCLUSION: Our study highlights the power of machine learning in integrating prior knowledge and genome-wide data to study genetic regulation of complex behaviors such as sleep.


Asunto(s)
FN-kappa B , Sueño , Animales , Ratones , Ritmo Circadiano/genética , Regulación de la Expresión Génica , FN-kappa B/genética , Sueño/genética , Sueño/fisiología , Privación de Sueño/genética
4.
JCI Insight ; 7(5)2022 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-35113810

RESUMEN

Infants born prematurely worldwide have up to a 50% chance of developing bronchopulmonary dysplasia (BPD), a clinical morbidity characterized by dysregulated lung alveolarization and microvascular development. It is known that PDGFR alpha-positive (PDGFRA+) fibroblasts are critical for alveolarization and that PDGFRA+ fibroblasts are reduced in BPD. A better understanding of fibroblast heterogeneity and functional activation status during pathogenesis is required to develop mesenchymal population-targeted therapies for BPD. In this study, we utilized a neonatal hyperoxia mouse model (90% O2 postnatal days 0-7, PN0-PN7) and performed studies on sorted PDGFRA+ cells during injury and room air recovery. After hyperoxia injury, PDGFRA+ matrix and myofibroblasts decreased and PDGFRA+ lipofibroblasts increased by transcriptional signature and population size. PDGFRA+ matrix and myofibroblasts recovered during repair (PN10). After 7 days of in vivo hyperoxia, PDGFRA+ sorted fibroblasts had reduced contractility in vitro, reflecting loss of myofibroblast commitment. Organoids made with PN7 PDGFRA+ fibroblasts from hyperoxia in mice exhibited reduced alveolar type 1 cell differentiation, suggesting reduced alveolar niche-supporting PDGFRA+ matrix fibroblast function. Pathway analysis predicted reduced WNT signaling in hyperoxia fibroblasts. In alveolar organoids from hyperoxia-exposed fibroblasts, WNT activation by CHIR increased the size and number of alveolar organoids and enhanced alveolar type 2 cell differentiation.


Asunto(s)
Displasia Broncopulmonar , Hiperoxia , Animales , Displasia Broncopulmonar/etiología , Fibroblastos/metabolismo , Humanos , Hiperoxia/complicaciones , Recién Nacido , Pulmón/patología , Ratones , Miofibroblastos/metabolismo
5.
PLoS Genet ; 17(11): e1009933, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34807912

RESUMEN

In mammals, the circadian clock coordinates cell physiological processes including inflammation. Recent studies suggested a crosstalk between these two pathways. However, the mechanism of how inflammation affects the clock is not well understood. Here, we investigated the role of the proinflammatory transcription factor NF-κB in regulating clock function. Using a combination of genetic and pharmacological approaches, we show that perturbation of the canonical NF-κB subunit RELA in the human U2OS cellular model altered core clock gene expression. While RELA activation shortened period length and dampened amplitude, its inhibition lengthened period length and caused amplitude phenotypes. NF-κB perturbation also altered circadian rhythms in the master suprachiasmatic nucleus (SCN) clock and locomotor activity behavior under different light/dark conditions. We show that RELA, like the clock repressor CRY1, repressed the transcriptional activity of BMAL1/CLOCK at the circadian E-box cis-element. Biochemical and biophysical analysis showed that RELA binds to the transactivation domain of BMAL1. These data support a model in which NF-kB competes with CRY1 and coactivator CBP/p300 for BMAL1 binding to affect circadian transcription. This is further supported by chromatin immunoprecipitation analysis showing that binding of RELA, BMAL1 and CLOCK converges on the E-boxes of clock genes. Taken together, these data support a significant role for NF-κB in directly regulating the circadian clock and highlight mutual regulation between the circadian and inflammatory pathways.


Asunto(s)
Factores de Transcripción ARNTL/genética , Proteínas CLOCK/genética , Inflamación/genética , Factor de Transcripción ReIA/genética , Animales , Proteínas de Unión al Calcio/genética , Línea Celular Tumoral , Relojes Circadianos/genética , Ritmo Circadiano/genética , Criptocromos/genética , Regulación del Desarrollo de la Expresión Génica/genética , Humanos , Inflamación/patología , FN-kappa B/genética , Núcleo Supraquiasmático/metabolismo
6.
Nat Commun ; 9(1): 430, 2018 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-29382851

RESUMEN

GTPase of immunity-associated protein 5 (Gimap5) is linked with lymphocyte survival, autoimmunity, and colitis, but its mechanisms of action are unclear. Here, we show that Gimap5 is essential for the inactivation of glycogen synthase kinase-3ß (GSK3ß) following T cell activation. In the absence of Gimap5, constitutive GSK3ß activity constrains c-Myc induction and NFATc1 nuclear import, thereby limiting productive CD4+ T cell proliferation. Additionally, Gimap5 facilitates Ser389 phosphorylation and nuclear translocation of GSK3ß, thereby limiting DNA damage in CD4+ T cells. Importantly, pharmacological inhibition and genetic targeting of GSK3ß can override Gimap5 deficiency in CD4+ T cells and ameliorates immunopathology in mice. Finally, we show that a human patient with a GIMAP5 loss-of-function mutation has lymphopenia and impaired T cell proliferation in vitro that can be rescued with GSK3 inhibitors. Given that the expression of Gimap5 is lymphocyte-restricted, we propose that its control of GSK3ß is an important checkpoint in lymphocyte proliferation.


Asunto(s)
Linfocitos T CD4-Positivos/fisiología , GTP Fosfohidrolasas/metabolismo , Proteínas de Unión al GTP/metabolismo , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Animales , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/patología , Muerte Celular , Proliferación Celular , Colitis/genética , Colitis/inmunología , Daño del ADN/inmunología , Activación Enzimática , Inhibidores Enzimáticos/farmacología , GTP Fosfohidrolasas/genética , Proteínas de Unión al GTP/genética , Proteínas de Unión al GTP/inmunología , Glucógeno Sintasa Quinasa 3 beta/antagonistas & inhibidores , Homeostasis , Humanos , Ratones Endogámicos C57BL , Ratones Transgénicos , Fosforilación
7.
Data Brief ; 13: 415-431, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28702480

RESUMEN

The following data are derived from key stages of acinar lung development and define the developmental role of lung interstitial fibroblasts expressing platelet-derived growth factor alpha (PDGFRα). This dataset is related to the research article entitled "Temporal, spatial, and phenotypical changes of PDGFRα expressing fibroblasts during late lung development" (Endale et al., 2017) [1]. At E16.5 (canalicular), E18.5 (saccular), P7 (early alveolar) and P28 (late alveolar), PDGFRαGFP mice, in conjunction with immunohistochemical markers, were utilized to define the spatiotemporal relationship of PDGFRα+ fibroblasts to endothelial, stromal and epithelial cells in both the proximal and distal acinar lung. Complimentary analysis with flow cytometry was employed to determine changes in cellular proliferation, define lipofibroblast and myofibroblast populations via the presence of intracellular lipid or alpha smooth muscle actin (αSMA), and evaluate the expression of CD34, CD29, and Sca-1. Finally, PDGFRα+ cells isolated at each stage of acinar lung development were subjected to RNA-Seq analysis, data was subjected to Bayesian timeline analysis and transcriptional factor promoter enrichment analysis.

8.
Dev Biol ; 425(2): 161-175, 2017 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-28408205

RESUMEN

Many studies have investigated the source and role of epithelial progenitors during lung development; such information is limited for fibroblast populations and their complex role in the developing lung. In this study, we characterized the spatial location, mRNA expression and Immunophenotyping of PDGFRα+ fibroblasts during sacculation and alveolarization. Confocal microscopy identified spatial association of PDGFRα expressing fibroblasts with proximal epithelial cells of the branching bronchioles and the dilating acinar tubules at E16.5; with distal terminal saccules at E18.5; and with alveolar epithelial cells at PN7 and PN28. Immunohistochemistry for alpha smooth muscle actin revealed that PDGFRα+ fibroblasts contribute to proximal peribronchiolar smooth muscle at E16.5 and to transient distal alveolar myofibroblasts at PN7. Time series RNA-Seq analyses of PDGFRα+ fibroblasts identified differentially expressed genes that, based on gene expression similarity were clustered into 7 major gene expression profile patterns. The presence of myofibroblast and smooth muscle precursors at E16.5 and PN7 was reflected by a two-peak gene expression profile on these days and gene ontology enrichment in muscle contraction. Additional molecular and functional differences between peribronchiolar smooth muscle cells at E16.5 and transient intraseptal myofibroblasts at PN7 were suggested by a single peak in gene expression at PN7 with functional enrichment in cell projection and muscle cell differentiation. Immunophenotyping of subsets of PDGFRα+ fibroblasts by flow cytometry confirmed the predicted increase in proliferation at E16.5 and PN7, and identified subsets of CD29+ myofibroblasts and CD34+ lipofibroblasts. These data can be further mined to develop novel hypotheses and valuable understanding of the molecular and cellular basis of alveolarization.


Asunto(s)
Fibroblastos/citología , Fibroblastos/metabolismo , Pulmón/embriología , Pulmón/metabolismo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Animales , Antígenos CD/metabolismo , Bronquios/citología , Diferenciación Celular/genética , Embrión de Mamíferos/citología , Epitelio/metabolismo , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Proteínas Fluorescentes Verdes/metabolismo , Inmunofenotipificación , Pulmón/citología , Ratones Transgénicos , Miocitos del Músculo Liso/metabolismo , Miofibroblastos/citología , Fenotipo , Regiones Promotoras Genéticas/genética , Alveolos Pulmonares/citología , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Factores de Tiempo , Factores de Transcripción/metabolismo
9.
Mediators Inflamm ; 2017: 7250968, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28316375

RESUMEN

Torilin, a sesquiterpene isolated from the fruits of Torilis japonica, has shown antimicrobial, anticancer, and anti-inflammatory properties. However, data on the mechanism of torilin action against inflammation is limited. This study aimed at determining the anti-inflammatory property of torilin in LPS-induced inflammation using in vitro model of inflammation. We examined torilin's effect on expression levels of inflammatory mediators and cytokines in LPS-stimulated RAW 264.7 macrophages. The involvement of NF-kB and AP-1, MAP kinases, and adaptor proteins were assessed. Torilin strongly inhibited LPS-induced NO release, iNOS, PGE2, COX-2, NF-α, IL-1ß, IL-6, and GM-CSF gene and protein expressions. In addition, MAPKs were also suppressed by torilin pretreatment. Involvement of ERK1/2, P38MAPK, and JNK1/2 was further confirmed by PD98059, SB203580, and SP600125 mediated suppression of iNOS and COX-2 proteins. Furthermore, torilin attenuated NF-kB and AP-1 translocation, DNA binding, and reporter gene transcription. Interestingly, torilin inhibited TAK1 kinase activation with the subsequent suppression of MAPK-mediated JNK, p38, ERK1/2, and AP-1 (ATF-2 and c-jun) activation and IKK-mediated I-κBα degradation, p65/p50 activation, and translocation. Together, the results revealed the suppression of NF-κB and AP-1 regulated inflammatory mediator and cytokine expressions, suggesting the test compound's potential as a candidate anti-inflammatory agent.


Asunto(s)
Inflamación/metabolismo , Quinasas Quinasa Quinasa PAM/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , FN-kappa B/metabolismo , Animales , Antiinflamatorios/farmacología , Ciclooxigenasa 2/metabolismo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Células RAW 264.7 , Sesquiterpenos de Guayano/farmacología , Transducción de Señal/efectos de los fármacos
10.
Am J Respir Cell Mol Biol ; 54(4): 532-45, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26414960

RESUMEN

Epithelial-mesenchymal cell interactions and factors that control normal lung development are key players in lung injury, repair, and fibrosis. A number of studies have investigated the roles and sources of epithelial progenitors during lung regeneration; such information, however, is limited in lung fibroblasts. Thus, understanding the origin, phenotype, and roles of fibroblast progenitors in lung development, repair, and regeneration helps address these limitations. Using a combination of platelet-derived growth factor receptor α-green fluorescent protein (PDGFRα-GFP) reporter mice, microarray, real-time polymerase chain reaction, flow cytometry, and immunofluorescence, we characterized two distinct interstitial resident fibroblasts, myo- and matrix fibroblasts, and identified a role for PDGFRα kinase activity in regulating their activation during lung regeneration. Transcriptional profiling of the two populations revealed a myo- and matrix fibroblast gene signature. Differences in proliferation, smooth muscle actin induction, and lipid content in the two subpopulations of PDGFRα-expressing fibroblasts during alveolar regeneration were observed. Although CD140α(+)CD29(+) cells behaved as myofibroblasts, CD140α(+)CD34(+) appeared as matrix and/or lipofibroblasts. Gain or loss of PDGFRα kinase activity using the inhibitor nilotinib and a dominant-active PDGFRα-D842V mutation revealed that PDGFRα was important for matrix fibroblast differentiation. We demonstrated that PDGFRα signaling promotes alveolar septation by regulating fibroblast activation and matrix fibroblast differentiation, whereas myofibroblast differentiation was largely PDGFRα independent. These studies provide evidence for the phenotypic and functional diversity as well as the extent of specificity of interstitial resident fibroblasts differentiation during regeneration after partial pneumonectomy.


Asunto(s)
Pulmón/citología , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Animales , Antígenos CD/inmunología , Proteínas Fluorescentes Verdes/genética , Inmunofenotipificación , Pulmón/enzimología , Pulmón/inmunología , Pulmón/fisiología , Ratones , Ratones Transgénicos , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Regeneración
11.
Mediators Inflamm ; 2015: 436017, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25944983

RESUMEN

Inflammatory bowel disease (IBD) including Crohn's disease and ulcerative colitis is often precipitated by an abnormal immune response to microbiota due to host genetic aberrancies. Recent studies highlight the importance of the host genome and microflora interactions in the pathogenesis of mucosal inflammation including IBD. Specifically, genome-wide (GWAS) and also next-generation sequencing (NGS)-including whole exome or genome sequencing-have uncovered a large number of susceptibility loci that predispose to autoimmune diseases and/or the two phenotypes of IBD. In addition, the generation of "IBD-prone" animal models using both reverse and forward genetic approaches has not only helped confirm the identification of susceptibility loci but also shed critical insight into the underlying molecular and cellular pathways that drive colitis development. In this review, we summarize recent findings derived from studies involving a novel early-onset model of colitis as it develops in GTPase of immunity-associated protein 5- (Gimap5-) deficient mice. In humans, GIMAP5 has been associated with autoimmune diseases although its function is poorly defined. Here, we discuss how defects in Gimap5 function impair immunological tolerance and lymphocyte survival and ultimately drive the development of CD4(+) T cell-mediated early-onset colitis.


Asunto(s)
GTP Fosfohidrolasas/fisiología , Proteínas de Unión al GTP/fisiología , Intestinos/inmunología , Tolerancia Periférica , Linfocitos T/citología , Animales , Colitis Ulcerosa/inmunología , Enfermedad de Crohn/inmunología , GTP Fosfohidrolasas/genética , Proteínas de Unión al GTP/genética , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Guanosina Trifosfato/química , Homeostasis , Humanos , Inmunidad , Enfermedades Inflamatorias del Intestino/inmunología , Ratones , Ratones Transgénicos , Fenotipo , Ratas
12.
Eur J Immunol ; 45(7): 2072-83, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25929249

RESUMEN

Absence of MHC class I expression is an important mechanism by which NK cells recognize a variety of target cells, yet the pathways underlying "missing-self" recognition, including the involvement of activating receptors, remain poorly understood. Using ethyl-N-nitrosourea mutagenesis in mice, we identified a germline mutant, designated Ace, with a marked defect in NK cell mediated recognition and elimination of "missing-self" targets. The causative mutation was linked to chromosome 11 and identified as a missense mutation (Thr428Ile) in the SH2 domain of Slp-76-a critical adapter molecule downstream of ITAM-containing surface receptors. The Slp-76 Ace mutation behaved as a hypomorphic allele-while no major defects were observed in conventional T-cell development/function, a marked defect in NK cell mediated elimination of ß2-microglobulin (ß2M) deficient target cells was observed. Further studies revealed Slp-76 to control NK-cell receptor expression and maturation; however, activation of Slp-76(ace/ace) NK cells through ITAM-containing NK-cell receptors or allogeneic/tumor target cells appeared largely unaffected. Imagestream analysis of the NK-ß2M(-/-) target cell synapse revealed a specific defect in actin recruitment to the conjugate synapse in Slp-76(ace/ace) NK cells. Overall these studies establish Slp-76 as a critical determinant of NK-cell development and NK cell mediated elimination of missing-self target cells in mice.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/inmunología , Células Asesinas Naturales/inmunología , Fosfoproteínas/inmunología , Autotolerancia/inmunología , Animales , Citometría de Flujo , Immunoblotting , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes
13.
Mediators Inflamm ; 2014: 748964, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24833816

RESUMEN

Despite a multitude of reports on anti-inflammatory properties of ginseng extracts or individual ginsenosides, data on antiarthritic effect of ginseng saponin preparation with mixed ginsenosides is limited. On the other hand, a combined therapy of safe and inexpensive plant-derived natural products such as ginsenosides can be considered as an alternative to treat arthritis. Our previous in vitro data displayed a strong anti-inflammatory action of red ginseng saponin fraction-A (RGSF-A). We, herein, report a marked antiarthritic property of RGSF-A rich in ginsenoside Rb1, Rc, and Rb2. Collagen-induced arthritic (CIA) mice were treated with RGSF-A or methotrexate (MTX) for 5 weeks. Joint pathology, serum antibody production and leukocye activation, cytokine production in the circulation, lymph nodes, and joints were examined. RGSF-A markedly reduced severity of arthritis, cellular infiltration, and cartilage damage. It suppressed CD3(+)/CD69(+), CD4(+)/CD25(+), CD8(+) T-cell, CD19(+), B220/CD23(+) B-cell, MHCII(+)/CD11c(+), and Gr-1(+)/CD11b(+) cell activations. It further suppressed anti-CII- or anti-RF-IgG/IgM, TNF-α, IL-1ß, IL-17, and IL-6 secretions but stimulated IL-10 levels in the serum, joint, or splenocyte. RGSF-A attenuated arthritis severity, modified leukocyte activations, and restored cytokine imbalances, suggesting that it can be considered as an antiarthritic agent with the capacity to ameliorate the immune and inflammatory responses in CIA mice.


Asunto(s)
Artritis Experimental/tratamiento farmacológico , Ginsenósidos/química , Panax/química , Extractos Vegetales/uso terapéutico , Saponinas/uso terapéutico , Animales , Artritis Experimental/sangre , Cromatografía Líquida de Alta Presión , Interleucina-10/sangre , Interleucina-17/sangre , Interleucina-6/sangre , Masculino , Ratones , Factor de Necrosis Tumoral alfa/sangre
14.
Immunobiology ; 218(12): 1452-67, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23735482

RESUMEN

Quercetin is a major bioflavonoid widely present in fruits and vegetables. It exhibits anti-inflammatory, anti-tumor, antioxidant properties and reduces cardiovascular disease risks. However, the molecular mechanism of action against inflammation in RAW 264.7 cells is only partially explored. Quercetin effect on LPS-induced gene and protein expressions of inflammatory mediators and cytokines were determined. Moreover, involvement of heme-oxygenase-1, protein kinases, adaptor proteins and transcription factors in molecular mechanism of quercetin action against inflammation were examined. Quercetin inhibited LPS-induced NO, PGE2, iNOS, COX-2, TNF-α, IL-1ß, IL-6 and GM-CSF mRNA and protein expressions while it promoted HO-1 induction in a dose- and time-dependent manner. It also suppressed I-κB-phosphorylation, NF-κB translocation, AP-1 and NF-κB-DNA-binding and reporter gene transcription. Quercetin attenuated p38(MAPK) and JNK1/2 but not ERK1/2 activations and this effect was further confirmed by SB203580 and SP600125-mediated suppressions of HO-1, iNOS, and COX-2 protein expressions. Moreover, quercetin arrested Src, PI3K, PDK1 and Akt activation in a time- and dose-dependent manner, which was comparable to PP2 and LY294002 inhibition of Src, PI3K/Akt and iNOS expressions. Quercetin further arrested Src and Syk tyrosine phosphorylations and their kinase activities followed by inhibition of PI3K tyrosine phosphorylation. Moreover, quercetin disrupted LPS-induced p85 association to TLR4/MyD88 complex and it then limited activation of IRAK1, TRAF6 and TAK1 with a subsequent reduction in p38 and JNK activations, and suppression in IKKα/ß-mediated I-κB phosphorylation. Quercetin limits LPS-induced inflammation via inhibition of Src- and Syk-mediated PI3K-(p85) tyrosine phosphorylation and subsequent TLR4/MyD88/PI3K complex formation that limits activation of downstream signaling pathways.


Asunto(s)
Antiinflamatorios/farmacología , Mediadores de Inflamación/metabolismo , Inflamación/tratamiento farmacológico , Macrófagos/efectos de los fármacos , Quercetina/farmacología , Animales , Línea Celular , Citocinas/metabolismo , Quinasa I-kappa B/metabolismo , Inflamación/inmunología , Lipopolisacáridos/inmunología , Activación de Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Ratones , Factor 88 de Diferenciación Mieloide/metabolismo , FN-kappa B/metabolismo , Estrés Oxidativo/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Factor de Transcripción AP-1/metabolismo
15.
Int Immunopharmacol ; 16(2): 232-42, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23623942

RESUMEN

Advancements in rheumatoid-arthritis-(RA) therapies have shown considerable progresses in the comprehension of disease. However, the development of new potential agents with relative safety and efficacy continues and natural compounds have been considered as alternatives to identify new entities. Since previous in-vivo data and our in-vitro findings showed that torilin has a strong anti-inflammatory property, we further investigated its effect against collagen-induced-arthritis-(CIA) in mice. CIA-induced DBA/1J mice were treated with torilin or methotrexate (MTX) for 5-weeks. Arthritis severity was evaluated by arthritic score and joint histopathology. Draining lymph node (dLN), joint and peripheral-blood mononuclear-cell (PBMC) counts, and activation/localization of T-/B-lymphocytes, dendritic cells (DCs) and neutrophils were examined by FACS analysis. Serum anti-type-II-collagen-(CII) antibody levels and cultured-splenocyte and serum cytokines were also evaluated. Torilin markedly reduced CIA-induced arthritic score, histopathology and leukocyte counts. Besides, torilin suppressed CIA-activated T-cells including CD3+, CD3+/CD69+, CD8+, CD4+ and CD4+/CD25+ in dLNs or joints. It also modified CD19+ or CD20+/CD23+ (B-cells), MHCII+/CD11c+ (DCs) and Gr-1+/CD11b+ (neutrophil) subpopulations. It further depressed total anti-CII-IgG, anti-CII-IgG1 and anti-CII-IgG2a antibody productions. Moreover, while IFN-γ and IL-10 were not affected, torilin suppressed CIA-induced serum TNF-α, IL-1ß and IL-6 levels. Interestingly, torilin also blocked IFN-γ, IL-17 and IL-6 cytokines while it did not affect IL-10 but enhanced IL-4 in splenocytes. These results show that torilin attenuated arthritis severity, modified leukocyte activations in dLNs or joints, and restored serum and splenocyte cytokine imbalances. Torilin may have immunomodulatory and anti-inflammatory properties with the capacity to ameliorate the inflammatory response in CIA-mice.


Asunto(s)
Antiinflamatorios/uso terapéutico , Artritis Experimental/tratamiento farmacológico , Animales , Antiinflamatorios/farmacología , Artritis Experimental/sangre , Artritis Experimental/inmunología , Artritis Experimental/patología , Colágeno Tipo II/inmunología , Citocinas/sangre , Citocinas/inmunología , Pie/patología , Inmunoglobulina G/sangre , Articulaciones/citología , Recuento de Leucocitos , Leucocitos/inmunología , Ganglios Linfáticos/citología , Masculino , Ratones , Ratones Endogámicos DBA , Sesquiterpenos de Guayano/farmacología , Sesquiterpenos de Guayano/uso terapéutico , Bazo/citología
16.
Artículo en Inglés | MEDLINE | ID: mdl-23304202

RESUMEN

Background. Progressive diseases including cancer, metabolic, and cardiovascular disorders are marked by platelet activation and chronic inflammation. Studies suggest that dietary flavonoids such as quercetin possess antioxidant, anti-inflammatory, and antiplatelet properties, which could prevent various chronic diseases including atherosclerosis and thrombosis. However, the mechanism and the signaling pathway that links quercetin's antiplatelet activity with its anti-inflammatory property is limited and thus further exploration is required. The aim of this paper was to examine the link between antiplatelet and anti-inflammatory roles of quercetin in agonist-induced platelet activation. Methods. Quercetin effects on agonist-activated platelet-aggregation, granule-secretion, [Ca(2+)](i), and glycoprotein-IIb/IIIa activation were examined. Its effects on PI3K/Akt, VASP, and MAPK phosphorylations were also studied on collaged-activated platelets. Results. Quercetin dose dependently suppressed collagen, thrombin, or ADP-induced platelet aggregation. It significantly inhibited collagen-induced ATP release, P-selectin expression, [Ca(2+)](i) mobilization, integrin-α(IIb)ß(3) activation, and augmented cAMP and VASP levels. Moreover, quercetin attenuated PI3K, Akt, ERK2, JNK1, and p38 MAPK activations, which were supported by platelet-aggregation inhibition with the respective kinase inhibitors. Conclusion. Quercetin-mediated antiplatelet activity involves PI3K/Akt inactivation, cAMP elevation, and VASP stimulation that, in turn, suppresses MAPK phosphorylations. This result suggests quercetin may have a potential to treat cardiovascular diseases involving aberrant platelet activation and inflammation.

17.
J Infect Dis ; 205(3): 445-52, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-22158566

RESUMEN

BACKGROUND: Brucella abortus can proliferate within professional and nonprofessional phagocytic host cells and thereby successfully bypass the bacteriocidal effects of phagocytes. However, the intracellular survival mechanism and factors of virulence are not fully understood. METHODS: We have investigated the role of the regulator of G protein signaling 2 (RGS2), an intracellular calcium ([Ca(2+)](i)) regulator of the host cell, in the intracellular survival of B. abortus within phagocytes. RESULTS: B. abortus infection markedly induced RGS2 messenger RNA expression in early phase and increased the [Ca(2+)](i) level up to 24 hours postinfection within macrophages from wild-type mice. The [Ca(2+)](i) level, however, was not influenced by B. abortus infection within macrophages from RGS2-deficient mice. Furthermore, B. abortus survival was reduced within RGS2-deficient macrophages, and hence bacterial proliferation was inhibited in RGS2-deficient mice. Moreover, treatment with the Ca(2+) chelator ethylenediaminetetraacetic acid (EDTA) or 1,2-bis-(2-amino-phenoxy)ethane-N,N,N',N'-tetraacetic acid acetoxymethyl ester (BAPTA-AM) and the L-type Ca(2+) channel-blocking agent nifedipine or genistein also showed a reduced intracellular replication of B. abortus within macrophages. CONCLUSION: These results indicate that B. abortus infection induces host RGS2 expression and that up-regulation of [Ca(2+)](i) levels is an essential factor for the intracellular survival of B. abortus within phagocytes.


Asunto(s)
Brucella abortus/crecimiento & desarrollo , Calcio/metabolismo , Cationes Bivalentes/metabolismo , Citosol/microbiología , Fagocitos/microbiología , Proteínas RGS/metabolismo , Animales , Brucella abortus/fisiología , Supervivencia Celular , Recuento de Colonia Microbiana , Citosol/química , Eliminación de Gen , Perfilación de la Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , Fagocitos/química , Fagocitos/metabolismo , Proteínas RGS/genética , Bazo/microbiología , Bazo/patología
18.
Phytother Res ; 25(11): 1596-603, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21394810

RESUMEN

Phellinus baumii is a mushroom that has been used as folk medicine against various diseases and is reported to have antidiabetic, anticancer, antioxidant, antiinflammatory and antihypertensive activities. However, information on the effects of P. baumii extract in platelet function is limited. Therefore, the aim of this study was to examine the impact of a P. baumii methanol extract (PBME) on platelet activation and to investigate the mechanism behind its antiplatelet activity. PBME effects on agonist-induced platelet aggregation, granule secretion, [Ca²âº](i) mobilization, α(IIb) ß3 activation, cyclic AMP release and mitogen-activated protein kinase (MAPK) phosphorylations were studied using rat platelets. PBME dose-dependently inhibited collagen, thrombin and ADP-induced platelet aggregation with an IC50 of 51.0 ± 2.4, 54.0 ± 2.1 and 53.0 ± 4.3 µg/mL, respectively. Likewise, thrombin-induced [Ca²âº](i) and collagen-activated ATP secretions were suppressed in PBME treated platelets. Aggregation and ATP secretion were also markedly attenuated by PBME alone or in combination with PP2 (Src inhibitor) and U-73122 (PLC inhibitor) in collagen-stimulated platelets. Besides, PBME treatment elevated basal cyclic AMP levels and inhibited collagen-induced integrin-α(IIb) ß3 activation. Moreover, PBME attenuated extracellular-signal-regulated protein kinase 2 (ERK2) and c-Jun N-terminal kinase 1 (JNK1) phosphorylations. Further PD98059 (ERK inhibitor) and SP60025 (JNK inhibitor) reduced collagen-induced platelet aggregation and ATP secretion. In conclusion, the observed PBME antiplatelet activity may be mediated by activation of cyclic AMP and inhibition of ERK2 and JNK1 phosphorylations. Finally, these data suggest that PBME may have therapeutic potential for the treatment of cardiovascular diseases that involve aberrant platelet function.


Asunto(s)
Basidiomycota/química , Productos Biológicos/farmacología , Plaquetas/efectos de los fármacos , AMP Cíclico/metabolismo , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Inhibidores de Agregación Plaquetaria/farmacología , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/antagonistas & inhibidores , Agaricales/química , Animales , Colágeno/antagonistas & inhibidores , Masculino , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 8 Activada por Mitógenos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosforilación , Activación Plaquetaria/efectos de los fármacos , Agregación Plaquetaria/efectos de los fármacos , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Ratas , Ratas Sprague-Dawley , Trombina/antagonistas & inhibidores
19.
J Vet Med Sci ; 73(2): 149-54, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20834197

RESUMEN

Erysipelothrix rhusiopathiae is pathogenic for humans, many domestic animals and wild birds, but infectious cases with clinical symptoms in cats have not been reported. E. rhusiopathiae was recovered from a 4-month Russian blue breed cat with a very poor body condition score of 1 (BCS: 1/5). The isolate was typed as serotype 2b. Mice experimentally infected with the clinical isolate of E. rhusiopathiae through subcutaneous or intraperitoneal routes survived, and the organism was recovered from the spleen and synovial and pericardial fluids. Cats experimentally inoculated with the isolate either orally or subcutaneously survived but commonly exhibited depression and emaciation together with localized erythemal lesion of the skin accompanied by purulent ocular discharge. On hematological analysis, the number of total white blood cells was high compared with that in normal cats. Histological examination revealed congestion and moderate inflammation with focal necrosis. This observation may provide insight on E. rhusiopathiae infection in cats with the possible epidemiological significance and implications as a potential source of infection to other animals and humans.


Asunto(s)
Enfermedades de los Gatos/microbiología , Depresión/microbiología , Infecciones por Erysipelothrix/microbiología , Erysipelothrix/aislamiento & purificación , Enfermedades Cutáneas Bacterianas/veterinaria , Animales , Bioensayo , Enfermedades de los Gatos/psicología , Gatos , ADN Bacteriano/química , ADN Bacteriano/genética , Depresión/psicología , Erysipelothrix/genética , Infecciones por Erysipelothrix/psicología , Femenino , Ratones , Ratones Endogámicos BALB C , Pruebas de Sensibilidad Microbiana/veterinaria , Reacción en Cadena de la Polimerasa/veterinaria , Serotipificación/veterinaria , Enfermedades Cutáneas Bacterianas/microbiología , Enfermedades Cutáneas Bacterianas/psicología , Organismos Libres de Patógenos Específicos
20.
Lab Anim Res ; 27(4): 317-25, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22232640

RESUMEN

Suaeda asparagoides (Miq.) has long been used as a Korean folk herbal medicine for the treatment of functional gastrointestinal disorders. However, reports on its pharmacological activity on gastrointestinal motility are scarce. The present study investigated the effects of Suaeda asparagoides water fraction of the extract (SAWF) on antral motility in vitro. Muscle strips from rat gastric antrum were set up in an organ bath in a circular orientation. SAWF (100 µg/mL) inhibited the spontaneous contraction of antral circular muscle strips. These inhibitory effects were not significantly affected by tetrodotoxin (1 µM), N(ω)-Nitro-L-arginine methyl ester hydrochloride (100 µM), 1H-(1,2,4)oxadiazolo(4,3-a)quinoxalin-1-one (10 µM), ryanodine (10 µM) and phentolamine (10 µM). SAWF-induced inhibition was mostly restored by cyclopiazonic acid (10 µM). Furthermore, the ß-adrenergic receptor antagonist, propranolol (10 µM), abolished SAWF-induced inhibition. These results suggest that SAWF may exert its activity on gastrointestinal smooth muscle via â-adrenergic receptors and sarcoplasmic reticulum Ca(2+) ATPase.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA