Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
1.
J Biol Chem ; 299(11): 105308, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37778730

RESUMEN

Nuclear factor kappa B (NF-κB) activity is regulated by various posttranslational modifications, of which Ser276 phosphorylation of RelA/p65 is particularly impacted by reactive oxygen species (ROS). This modification is responsible for selective upregulation of a subset of NF-κB targets; however, the precise mechanism remains elusive. ROS have the ability to modify cellular molecules including DNA. One of the most common oxidation products is 8-oxo-7,8-dihydroguanine (8-oxoGua), which is repaired by the 8-oxoguanine DNA glycosylase1 (OGG1)-initiated base excision repair pathway. Recently, a new function of OGG1 has been uncovered. OGG1 binds to 8-oxoGua, facilitating the occupancy of NF-κB at promoters and enhancing transcription of pro-inflammatory cytokines and chemokines. In the present study, we demonstrated that an interaction between DNA-bound OGG1 and mitogen-and stress-activated kinase 1 is crucial for RelA/p65 Ser276 phosphorylation. ROS scavenging or OGG1 depletion/inhibition hindered the interaction between mitogen-and stress-activated kinase 1 and RelA/p65, thereby decreasing the level of phospho-Ser276 and leading to significantly lowered expression of ROS-responsive cytokine/chemokine genes, but not that of Nfkbis. Blockade of OGG1 binding to DNA also prevented promoter recruitment of RelA/p65, Pol II, and p-RNAP II in a gene-specific manner. Collectively, the data presented offer new insights into how ROS signaling dictates NF-κB phosphorylation codes and how the promoter-situated substrate-bound OGG1 is exploited by aerobic mammalian cells for timely transcriptional activation of ROS-responsive genes.


Asunto(s)
ADN Glicosilasas , FN-kappa B , Animales , ADN/metabolismo , ADN Glicosilasas/genética , ADN Glicosilasas/metabolismo , Mamíferos/metabolismo , Mitógenos , FN-kappa B/metabolismo , Fosforilación , Especies Reactivas de Oxígeno/metabolismo , Factor de Transcripción ReIA/genética , Factor de Transcripción ReIA/metabolismo , Humanos , Ratones , Línea Celular , Ratones Noqueados
2.
J Biol Chem ; 299(10): 105230, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37689116

RESUMEN

Macrophages must respond appropriately to pathogens and other pro-inflammatory stimuli in order to perform their roles in fighting infection. One way in which inflammatory stimuli can vary is in their dynamics-that is, the amplitude and duration of stimulus experienced by the cell. In this study, we performed long-term live cell imaging in a microfluidic device to investigate how the pro-inflammatory genes IRF1, CXCL10, and CXCL9 respond to dynamic interferon-gamma (IFNγ) stimulation. We found that IRF1 responds to low concentration or short duration IFNγ stimulation, whereas CXCL10 and CXCL9 require longer or higherconcentration stimulation to be expressed. We also investigated the heterogeneity in the expression of each gene and found that CXCL10 and CXCL9 have substantial cell-to-cell variability. In particular, the expression of CXCL10 appears to be largely stochastic with a subpopulation of nonresponding cells across all the stimulation conditions tested. We developed both deterministic and stochastic models for the expression of each gene. Our modeling analysis revealed that the heterogeneity in CXCL10 can be attributed to a slow chromatin-opening step that is on a similar timescale to that of adaptation of the upstream signal. In this way, CXCL10 expression in individual cells can remain stochastic in response to each pulse of repeated stimulation, which we also validated by experiments. Together, we conclude that pro-inflammatory genes in the same signaling pathway can respond to dynamic IFNγ stimulus with very different response features and that upstream signal adaptation can contribute to shaping heterogeneous gene expression.


Asunto(s)
Quimiocina CXCL10 , Quimiocina CXCL9 , Regulación de la Expresión Génica , Factor 1 Regulador del Interferón , Macrófagos , Quimiocina CXCL10/genética , Quimiocina CXCL10/metabolismo , Quimiocina CXCL9/genética , Quimiocina CXCL9/metabolismo , Interferón gamma/farmacología , Macrófagos/metabolismo , Transducción de Señal/genética , Células RAW 264.7 , Animales , Ratones , Factor 1 Regulador del Interferón/genética , Factor 1 Regulador del Interferón/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/inmunología , Simulación por Computador , Análisis de la Célula Individual , Adyuvantes Inmunológicos/farmacología
3.
J Biol Chem ; 297(5): 101274, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34619149

RESUMEN

The ubiquitously expressed transcription factor interferon (IFN) regulatory factor 3 (IRF3) is critical for the induction of antiviral genes, e.g., type-I IFN. In addition to its transcriptional function, IRF3 also activates a nontranscriptional, proapoptotic signaling pathway. While the proapoptotic function of IRF3 protects against viral infections, it is also involved in harmful immune responses that trigger hepatocyte cell death and promote liver disease. Thus, we hypothesized that a small-molecule inhibitor of the proapoptotic activity of IRF3 could alleviate fatty-acid-induced hepatocyte cell death. We conducted a high-throughput screen, which identified auranofin as a small-molecule inhibitor of the proapoptotic activity of IRF3. In addition to the nontranscriptional apoptotic pathway, auranofin also inhibited the transcriptional activity of IRF3. Using biochemical and genetic tools in human and mouse cells, we uncovered a novel mechanism of action for auranofin, in which it induces cellular autophagy to degrade IRF3 protein, thereby suppressing IRF3 functions. Autophagy-deficient cells were unable to degrade IRF3 upon auranofin treatment, suggesting that the autophagic degradation of IRF3 is a novel approach to regulate IRF3 activities. Using a physiologically relevant in vitro model, we demonstrated that auranofin inhibited fatty-acid-induced apoptotic cell death of hepatocytes. In summary, auranofin is a novel inhibitor of IRF3 functions and may represent a potential therapeutic option in diseases where IRF3 is deleterious.


Asunto(s)
Apoptosis/efectos de los fármacos , Auranofina/farmacología , Autofagia/efectos de los fármacos , Factor 3 Regulador del Interferón/metabolismo , Proteolisis/efectos de los fármacos , Transcripción Genética/efectos de los fármacos , Animales , Humanos , Factor 3 Regulador del Interferón/genética , Ratones , Células RAW 264.7
4.
World J Surg Oncol ; 20(1): 263, 2022 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-35982458

RESUMEN

Cutaneous melanoma (CM) is a skin cancer that is highly metastatic and aggressive, with a dismal prognosis. This is the first study to use inflammatory response-related genes to build a model and evaluate their predictive significance in CM. This study used public databases to download CM patients' mRNA expression profiles and clinical data to create multigene prognostic markers in the UCSC cohort. We compared overall survival (OS) between high- and low-risk groups using the Kaplan-Meier curve and determined independent predictors using Cox analysis. We also used enrichment analysis to assess immune cell infiltration fraction and immune pathway-related activity using KEGG enrichment analysis. Furthermore, we detected prognostic genes' mRNA and protein expression in CM and normal skin tissues using qRT-PCR and immunohistochemistry. Finally, we developed a 5-gene predictive model that showed that patients in the high-risk group had a considerably shorter OS than those in the low-risk group. The analysis of the receiver operating characteristic (ROC) curve proved the model's predictive ability. We also conducted a drug sensitivity analysis and discovered that the expression levels of prognostic genes were substantially linked with cancer cell sensitivity to antitumor medicines. The findings show that the model we developed, which consists of five inflammatory response-related genes, can be used to forecast the prognosis and immunological state of CM, giving personalized and precision medicine a new goal and direction.


Asunto(s)
Antineoplásicos , Melanoma , Neoplasias Cutáneas , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Humanos , Estimación de Kaplan-Meier , Melanoma/tratamiento farmacológico , Melanoma/genética , Melanoma/patología , Pronóstico , ARN Mensajero/metabolismo , Neoplasias Cutáneas/tratamiento farmacológico , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología , Melanoma Cutáneo Maligno
5.
Inflammopharmacology ; 30(4): 1459-1474, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35608743

RESUMEN

Uncontrolled inflammation plays a central role in the pathogenesis of various diseases. Currently available anti-inflammatory agents on prolonged use may lead to ulcers or thrombus formation. The present study was designed to evaluate the anti-inflammatory, anti-arthritic and anti-angiogenic potentials of methanol extract of Viola betonicifolia using battery of in vivo models. Methanol extract of Viola betonicifolia (Vb.Me) was prepared through maceration. High performance liquid chromatography (HPLC) and gas chromatography mass spectrometery (GC-MS) were performed to identify bioactive compounds present in Vb.Me. In vivo safety profile of Vb.Me was evaluated following OECD 425 acute toxicity guidelines. Anti-inflammatory potential of Vb.Me at three different dose levels was evaluated in in vivo acute (carrageenan and, histamine-induced paw oedema), sub-chronic (cotton pellet-induced granuloma) and chronic (Complete Freund's adjuvant-induced arthritis) models. Blood and paws samples were collected to study effects of Vb.Me treatment on the expression of various pro- and anti-inflammatory genes (RT-PCR) and to study the histopathological changes at tissue levels. Effects of Vb.Me on neovasculature development were studied in ex-ovo chicken chorioallantoic membrane (CAM) assay. Quercetin and n-hexadecanoic were identified as one of the major bioactive molecules in HPLC and GC-MS analysis of Vb.Me. Toxicity data revealed that Vb.Me was safe for administration up to the dose of 2000 mg/kg. Findings of inflammatory models showed that Vb.Me produced time and dose-dependent effects. 500 mg/kg Vb.Me showed significantly (p < 0.05) better effects as compared with 125 and 250 mg/kg. 500 mg/kg Vb.Me also showed comparable anti-inflammatory effects with indomethacin in both acute and chronic models respectively. RT-PCR data exhibited significant (p < 0.05) down-regulation of IL-6, IL-1ß, NF-kß, TNF-α and COX-2 genes with simultaneous up-regulation of IL-4 and IL-10 genes in the blood samples of animals treated with 500 mg/kg of Vb.Me and 10 mg/kg of indomethacin respectively. CAM assay data revealed arrest of microvessel outgrowth in Vb.Me-treated eggs. Altogether, findings of the current study indicate that Vb.Me exerts in vivo anti-inflammatory and anti-angiogenic effects through regulation of expression of various pro- and anti-inflammatory genes. Synergist actions of various bioactive molecules in Vb.Me are proposed to be responsible for these attributes. However, further studies to standardize the extract and evaluation of its potential in various inflammation-induced diseases are warranted.


Asunto(s)
Viola , Animales , Antiinflamatorios/uso terapéutico , Edema/inducido químicamente , Edema/tratamiento farmacológico , Indometacina/uso terapéutico , Inflamación/inducido químicamente , Inflamación/tratamiento farmacológico , Metanol , Extractos Vegetales/uso terapéutico
6.
BMC Genomics ; 22(1): 200, 2021 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-33752587

RESUMEN

BACKGROUND: Tilapia (Oreochromis niloticus) cultures are frequently infected by Vibrio vulnificus, causing major economic losses to production units. Previously, tilapia expressing recombinant delta-5 desaturase and delta-6 desaturase (D56) were found to be resistant to V. vulnificus infection. In this report, we profile the D56-mediated molecular changes underlying this resistance in tilapia. A comparative transcriptome analysis was performed on V. vulnificus-infected wild-type and D56-transgenic tilapia using Illumina's sequencing-by-synthesis approach. Gene enrichment analysis on differentially expressed unigenes was performed, and the expression patterns were validated by real-time PCR. RESULTS: Comparative transcriptome analysis was performed on RNA-sequence profiles obtained from wild-type and D56-transgenic tilapia at 0, 6 and 24 h post-infection with V. vulnificaus. GO and KEGG gene enrichment analyses showed that D56 regulates several pathways and genes, including fatty acid (FA) metabolism associated, and inflammatory and immune response. Expression of selected FA metabolism-associated, inflammatory and immune responsive genes was validated by qPCR. The inflammatory and immune responsive genes that are modulated by FA-associated D56 likely contribute to the enhanced resistance against V. vulnificus infection in Tilapia. CONCLUSIONS: Transcriptome profiling and filtering for two-fold change variation showed that 3795 genes were upregulated and 1839 genes were downregulated in D56-transgenic tilapia. These genes were grouped into pathways, such as FA metabolism, FA elongation, FA biosynthesis, biosynthesis of unsaturated FA, FA degradation, inflammation, immune response, and chemokines. FA-associated genes and immune-related genes were modulated by D56 at 6 h and 24 h post infection with V. vulnificus. The expression patterns of FA-related genes, inflammatory genes, antimicrobial peptide genes and immune responsive genes at 0, 3, 6, 12, 24 and 48 h post-infection suggests these genes are involved in the enhanced resistance of D56 transgenic tilapia to V. vulnificus.


Asunto(s)
Cíclidos , Enfermedades de los Peces , Tilapia , Vibriosis , Vibrio vulnificus , Animales , Cíclidos/genética , Enfermedades de los Peces/genética , Perfilación de la Expresión Génica , Tilapia/genética , Transcriptoma , Vibriosis/genética , Vibriosis/veterinaria , Vibrio vulnificus/genética
7.
Mol Cancer ; 20(1): 5, 2021 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-33397398

RESUMEN

BACKGROUND: Cytokine release syndrome (CRS) is a systemic inflammatory response characterized by the overexpression of inflammatory genes. Controlling CRS is essential for improving the therapeutic effects of chimeric antigen receptor (CAR) engineered T cells. However, current treatment options are limited given the complexity of cytokine interactions so it is important to seek a mild strategy with broad-spectrum inhibition to overcome this challenge. METHODS: Using THZ1, a covalent inhibitor of cyclin-dependent kinase 7 (CDK7), we demonstrated the transcriptional suppression of inflammatory genes in activated macrophages. RNA sequencing and ChIP sequencing were conducted to identify the key target genes of the inflammatory response. Pathogen- and CAR T cell-induced CRS models were also established to assess the efficacy and safety of targeting CDK7. RESULTS: CDK7 blockade attenuated cytokine release, mitigated hyperinflammatory states and rescued mice from lethal CRS. Targeting CDK7 preferentially suppressed a set of inflammatory genes, of which STAT1 and IL1 were the key targets associated with super enhancers. Furthermore, we confirmed the potent efficacy of THZ1 in alleviating the CRS induced by CAR T cell infusion without causing tissue injury or impairing antitumor effects. CONCLUSIONS: Our work indicates the CDK7-dependent transcription addiction of inflammatory genes. Targeting CDK7 is a promising strategy for treating CRS by inhibiting multiple cytokines.


Asunto(s)
Quinasas Ciclina-Dependientes/metabolismo , Síndrome de Liberación de Citoquinas/etiología , Elementos de Facilitación Genéticos/genética , Inmunoterapia Adoptiva/efectos adversos , Inflamación/genética , Animales , Antineoplásicos/farmacología , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Femenino , Perfilación de la Expresión Génica , Humanos , Inflamación/patología , Lipopolisacáridos , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones Endogámicos C57BL , Modelos Biológicos , Fenilendiaminas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Pirimidinas/farmacología , ARN Polimerasa II/metabolismo , Transcripción Genética/efectos de los fármacos , Quinasa Activadora de Quinasas Ciclina-Dependientes
8.
Brain Behav Immun ; 98: 251-256, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34400237

RESUMEN

The vagus nerve mediates parasympathetic nervous system control of peripheral physiological processes including cardiovascular activity and immune response. In mice, tonic vagal activation down-regulates inflammation via nicotinic acetylcholine receptor-mediated inhibition of the pro-inflammatory transcription factor NF-κB in monocyte/macrophages. Because Type I interferon and pro-inflammatory genes are regulated reciprocally at the level of transcription factor activation and cell differentiation, we hypothesized that vagal activity would up-regulate Type I interferon response genes concurrently with inflammatory downregulation in human immune cells. We mapped empirical individual differences in the circulating leukocyte transcriptome and vagal activity indexed by high frequency (0.15-0.40 Hz) heart rate variability (HF-HRV) in 380 participants in the Midlife in the US study. Here we show that promoter-based bioinformatics analyses linked greater HF-HRV to reduced NF-κB activity and increased activity of IRF transcription factors involved in Type I interferon response (independent of ß-antagonists, BMI, smoking, heavy alcohol consumption, and demographic factors). Transcript origin analyses implicated myeloid lineage immune cells as targets, representing per-cell alterations in gene transcription as HF-HRV was not associated with differential prevalence of leukocyte subsets. These findings support the concept of parasympathetic inhibition of pro-inflammatory gene expression in humans and up-regulation of Type I interferons that could augment host defense against viral infections.


Asunto(s)
Antivirales , Sistema Nervioso Parasimpático , Animales , Humanos , Factores Reguladores del Interferón , Leucocitos/metabolismo , Ratones , FN-kappa B/metabolismo
9.
Fish Shellfish Immunol ; 114: 238-252, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33989765

RESUMEN

The effect of Agaricus bisporus polysaccharides (ABPs) supplemented diet on growth rate, antioxidant capacity, innate-adaptive immune response, proinflammatory and antiinflammatory genes expression in Ctenopharyngodon idella against Aeromonas hydrophila is reported. In both normal and challenged groups fed with 1.0 and 1.5 mg kg-1 ABPs diets resulted in a significant weight gain and feed intake. The survival was 100% in normal fish fed without or with any ABPs diet; the challenged fish fed with 1.0 mg kg-1 ABPs diet had 98.6% survival. The RBC and WBC counts, Hb, and Hct levels were significant in both normal and challenged groups fed with 1.0 and 1.5 mg kg-1 ABPs diets. A significant increase in total protein and albumin level was observed in both groups fed with 1.0 and 1.5 mg kg-1 ABPs diets. Significant increase in GPx, ROS, GR, GSH, PC, and MnSOD activity was observed in HK of both groups fed with 1.0 and 1.5 mg kg-1 ABPs diets; similarly both groups when fed with the same ABPs diets showed significant Lz, C3, and C4 activity. However, both groups fed with 1.0 mg kg-1 ABPs diet showed significant ß-defensin, LEAP-2A, IL-6, and NF-κB P65 mRNA expression. Similarly, IFN-γ2, IL-10, and TNFα mRNA expressions were significant in both groups fed with 1.0 mg kg-1 ABPs diet. The results indicate that both normal and challenged C. idella fed with a 1.0 mg kg-1 ABPs diet had better growth, antioxidant status, immune response, and pro-anti-inflammatory gene modulation against A. hydrophila.


Asunto(s)
Agaricus/química , Alimentación Animal/análisis , Carpas/metabolismo , Suplementos Dietéticos , Polisacáridos/farmacología , Inmunidad Adaptativa/efectos de los fármacos , Fenómenos Fisiológicos Nutricionales de los Animales , Animales , Antiinflamatorios/metabolismo , Antioxidantes/metabolismo , Dieta/veterinaria , Regulación de la Expresión Génica/efectos de los fármacos , Inmunidad Innata/efectos de los fármacos , Polisacáridos/química
10.
J Stroke Cerebrovasc Dis ; 30(12): 106105, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34547676

RESUMEN

OBJECTIVES: Post-ischemic inflammation leads to apoptosis as an indirect cause of functional disabilities after the stroke. Melatonin may be a good candidate for the stroke recovery because of its anti-inflammatory effects. Therefore, we investigated the effect of melatonin on inflammation in the functional recovery of brain by evaluating ipsilesional and contralesional alterations. MATERIALS AND METHODS: Melatonin (4 mg/kg/day) was intraperitoneally administered into the mice from the 3rd to the 55th day of the post-ischemia after 30 min of middle cerebral artery occlusion. RESULTS: Melatonin produced a functional recovery by reducing the emigration of the circulatory leukocytes and the local microglial activation within the ischemic brain. Overall, the expression of the inflammation-related genes reduced upon melatonin treatment in the ischemic hemisphere. On the other hand, the expression level of the inflammatory cytokine genes raised in the contralateral hemisphere at the 55th day of the post-ischemia. Furthermore, melatonin triggers an increase in the iNOS expression and a decrease in the nNOS expression in the ipsilateral hemisphere at the earlier times in the post-ischemic recovery. At the 55th day of the post-ischemic recovery, melatonin administration enhanced the eNOS and nNOS protein expressions. CONCLUSIONS: The present molecular, biological, and histological data have revealed broad anti-inflammatory effects of melatonin in both hemispheres with distinct temporal and spatial patterns at different phases of post-stroke recovery. These outcomes also established that melatonin act recruitment of contralesional rather than of ipsilesional.


Asunto(s)
Isquemia Encefálica , Citocinas , Inflamación , Melatonina , Plasticidad Neuronal , Animales , Antiinflamatorios/administración & dosificación , Isquemia Encefálica/fisiopatología , Citocinas/metabolismo , Inflamación/tratamiento farmacológico , Melatonina/administración & dosificación , Ratones , Plasticidad Neuronal/fisiología , Tiempo de Tratamiento
11.
Pharm Biol ; 59(1): 799-810, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34190667

RESUMEN

CONTEXT: Among the plants in the genus Barringtonia (Lecythidaceae) used as traditional medicines to treat arthralgia, chest pain, and haemorrhoids in Indonesia, Barringtonia racemosa L. and Barringtonia acutangula (L.) Gaertn. have demonstrated anti-inflammatory activity in systemic inflammatory models. OBJECTIVE: The anti-inflammatory activity of Barringtonia angusta Kurz has not been investigated. We prepared a methanol extract of the leaves and stems of B. angusta (Ba-ME) and systemically evaluated its anti-inflammatory effects in vitro and in vivo. MATERIALS AND METHODS: RAW264.7 cells stimulated with LPS or Pam3CSK4 for 24 h were treated with Ba-ME (12.5, 25, 50, 100, and 150 µg/mL), and NO production and mRNA levels of inflammatory genes were evaluated. Luciferase reporter gene assay, western blot analysis, overexpression experiments, and cellular thermal shift assay were conducted to explore the mechanism of Ba-ME. In addition, the anti-gastritis activity of Ba-ME (50 and 100 mg/kg, administered twice per day for two days) was evaluated using an HCl/EtOH-induced gastritis mouse model. RESULTS: Ba-ME dose-dependently suppressed NO production [IC50 = 123.33 µg/mL (LPS) and 46.89 µg/mL (Pam3CSK4)] without affecting cell viability. Transcriptional expression of iNOS, IL-1ß, COX-2, IL-6, and TNF-α and phosphorylation of Src, IκBα, p50/105, and p65 were inhibited by Ba-ME. The extract specifically targeted the Src protein by binding to its SH2 domain. Moreover, Ba-ME significantly ameliorated inflammatory lesions in the HCl/EtOH-induced gastritis model. DISCUSSION AND CONCLUSIONS: The anti-inflammatory activity of Ba-ME is mediated by targeting of the Src/NF-κB signalling pathway, and B. angusta has potential as an anti-inflammatory drug.


Asunto(s)
Antiinflamatorios/administración & dosificación , Barringtonia , Sistemas de Liberación de Medicamentos/métodos , Gastritis/tratamiento farmacológico , Extractos Vegetales/administración & dosificación , Familia-src Quinasas/antagonistas & inhibidores , Animales , Antiinflamatorios/aislamiento & purificación , Antiinflamatorios/metabolismo , Relación Dosis-Respuesta a Droga , Gastritis/inducido químicamente , Gastritis/metabolismo , Células HEK293 , Humanos , Masculino , Metanol/administración & dosificación , Metanol/metabolismo , Ratones , Ratones Endogámicos ICR , FN-kappa B , Extractos Vegetales/aislamiento & purificación , Extractos Vegetales/metabolismo , Hojas de la Planta , Tallos de la Planta , Células RAW 264.7 , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Familia-src Quinasas/metabolismo
12.
J Transl Med ; 18(1): 433, 2020 11 12.
Artículo en Inglés | MEDLINE | ID: mdl-33183332

RESUMEN

BACKGROUND: Obesity is a chronic low-grade inflammatory disease that is generally characterized by enhanced inflammation in obese adipose tissue (AT). Here, we investigated alterations in gene expression between lean and obese conditions using mRNA-Seq data derived from human purified adipocytes (ACs) and preadipocytes (preACs). RESULTS: Total mRNA-seq data were generated with 27 AC and 21 preAC samples purified from human visceral AT collected during resection surgery in cancer patients, where the samples were classified into lean and obese categories by BMI > 25 kg/m2. We defined four classes of differentially expressed genes (DEGs) by comparing gene expression between (1) lean and obese ACs, (2) lean and obese preACs, (3) lean ACs and lean preACs, and 4) obese ACs and obese preACs. Based on an analysis of comparison 1, numerous canonical obesity-related genes, particularly inflammatory genes including IL-6, TNF-α and IL-1ß, i.e., the genes that are expected to be upregulated in obesity conditions, were found to be expressed at significantly lower levels in obese ACs than in lean ACs. In contrast, some inflammatory genes were found to be expressed at higher levels in obese preACs than lean preACs in the analysis of comparison 2. The analysis of comparisons 3 and 4 showed that inflammatory gene classes were expressed at higher levels in differentiated ACs than undifferentiated preACs under both lean and obese conditions; however, the degree of upregulation was significantly greater for lean than for obese conditions. We validated our observations using previously published microarray transcriptome data deposited in the GEO database (GSE80654). CONCLUSIONS: Taken together, our analyses suggest that inflammatory genes are expressed at lower levels in obese ACs than in lean ACs because lean adipogenesis involves even greater enhancement of inflammatory responses than does obese adipogenesis.


Asunto(s)
Adipocitos , Tejido Adiposo , Adipogénesis/genética , Expresión Génica , Humanos , Obesidad/genética
13.
BMC Vet Res ; 16(1): 9, 2020 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-31918707

RESUMEN

BACKGROUND: Bacterial infections are common in postpartum dairy cows. Cortisol level has been observed to increase in dairy cows during peripartum period, and is associated with the endometrial innate immunity against pathogens like E.coli. However, the mechanism underlying how cortisol regulates E.coli-induced inflammatory response in bovine endometrial epithelial cells (BEEC) remains elusive. RESULTS: Cortisol decreased the expressions of IL1ß, IL6, TNF-α, IL8, and TLR4 mRNA in BEEC treated with LPS or heat-killed E.coli, but up-regulated these gene expressions in BEEC stimulated by live E.coli. CONCLUSION: Cortisol exerted the anti-inflammatory action on LPS- or heat-killed E.coli-stimulated BEEC, but the pro-inflammatory action on live E.coli-induced BEEC.


Asunto(s)
Citocinas/genética , Escherichia coli , Hidrocortisona/farmacología , Animales , Bovinos , Enfermedades de los Bovinos/inmunología , Enfermedades de los Bovinos/microbiología , Células Cultivadas , Citocinas/metabolismo , Endometritis/veterinaria , Endometrio/citología , Endometrio/efectos de los fármacos , Endometrio/inmunología , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/inmunología , Femenino , Regulación de la Expresión Génica , Inmunidad Innata , Lipopolisacáridos/toxicidad , ARN Mensajero/metabolismo
14.
Am J Physiol Regul Integr Comp Physiol ; 316(5): R525-R534, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30811246

RESUMEN

Double-stranded (ds) RNA, both synthetic and produced during virus replication, rapidly stimulates MAPK and NF-κB signaling that results in expression of the inflammatory genes inducible nitric oxide synthase, cyclooxygenase 2, and IL-1ß by macrophages. Using biochemical and genetic approaches, we have identified the chemokine ligand-binding C-C chemokine receptor type 5 (CCR5) as a cell surface signaling receptor required for macrophage expression of inflammatory genes in response to dsRNA. Activation of macrophages by synthetic dsRNA does not require known dsRNA receptors, as poly(inosinic:cytidylic) acid [poly(I:C)] activates signaling pathways leading to expression of inflammatory genes to similar levels in wild-type and Toll-like receptor 3- or melanoma differentiation antigen 5-deficient macrophages. In contrast, macrophage activation in response to poly(I:C) is attenuated in macrophages isolated from mice lacking CCR5. These findings support a role for CCR5 as a cell surface signaling receptor that participates in activation of inflammatory genes in macrophages in response to the viral dsRNA mimetic poly(inosinic:cytidylic) acid by pathways that are distinct from classical dsRNA receptor-mediated responses.


Asunto(s)
Inflamación/metabolismo , Activación de Macrófagos/efectos de los fármacos , Macrófagos Peritoneales/efectos de los fármacos , Poli I-C/farmacología , Receptores CCR5/agonistas , Transducción de Señal/efectos de los fármacos , Animales , Citocinas/genética , Citocinas/metabolismo , Regulación de la Expresión Génica , Inflamación/genética , Inflamación/inmunología , Helicasa Inducida por Interferón IFIH1/deficiencia , Helicasa Inducida por Interferón IFIH1/genética , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células RAW 264.7 , Receptores CCR5/genética , Receptores CCR5/metabolismo , Receptor Toll-Like 3/genética , Receptor Toll-Like 3/metabolismo
15.
Can J Physiol Pharmacol ; 97(4): 320-327, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30388381

RESUMEN

Regulation of cardiac fatty acid metabolism is central to the development of cardiac hypertrophy and heart failure. We investigated the effects of select fatty acids on the expression of genes involved in immediate early as well as inflammatory and hypertrophic responses in adult rat cardiomyocytes. Cardiac remodeling begins with upregulation of immediate early genes for c-fos and c-jun, followed by upregulation of inflammatory genes for nuclear factor kappa B (NF-κB) and nuclear factor of activated T-cells (NFAT). At later stages, genes involved in hypertrophic responses, such as atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP), are upregulated. Adult rat cardiomyocytes were treated with palmitic acid, a saturated fatty acid; oleic acid, a monounsaturated fatty acid; linoleic acid, a polyunsaturated fatty acid belonging to the n-6 class; and docosahexaenoic acid, a polyunsaturated fatty acid belonging to the n-3 class. Linoleic acid produced a greater increase in the mRNA expression of c-fos, c-jun, NF-κB, NFAT3, ANP, and BNP relative to palmitic acid and oleic acid. In contrast, docosahexaenoic acid caused a decrease in the expression of genes involved in cardiac hypertrophy. Our findings suggest that linoleic acid may be a potent inducer of genes involved in cardiac hypertrophy, whereas docosahexaenoic acid may be protective against the cardiomyocyte hypertrophic response.


Asunto(s)
Ácidos Docosahexaenoicos/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Ácido Linoleico/farmacología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Animales , Biomarcadores/metabolismo , Inflamación/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/genética , Masculino , Proteínas Proto-Oncogénicas c-fos/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley
16.
Pediatr Hematol Oncol ; 36(1): 28-39, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30888230

RESUMEN

A major problem of hemophilia A (HA) treatment is the development of factor VIII (FVIII) inhibitor, which usually occurs shortly after initiating replacement therapy. Several studies showed the correlation between inhibitor development and polymorphisms in inflammatory and immune response genes of HA patients; however, literature data are not available to prove this association in Iranian population. The aim of this study was to investigate a possible association between FVIII inhibitor formation and the polymorphisms of 16 inflammatory and immune response genes in Iranian severe HA patients (FVIII activity < 1%). This case-control study was performed on 55 patients with severe HA inhibitors and 45 samples without inhibitors from Iranian Comprehensive Hemophilia Care center. After extraction of whole genomic DNA from blood samples and design of primers for 16 genes, the genotyping was performed by Tetra primer ARMS PCR, and the validation of single nucleotide polymorphisms was determined by DNA sequencing. The data indicated that there was a significant association between inhibitor development, and F13A1 (TT), DOCK2 (CC& CT), and MAPK9 (TT) genotypes. Moreover, a considerably increased inhibitor risk carrying T, C, and T allele for F13A1, DOCK2, and MAPK9 genes was observed in patients with inhibitors, respectively. In contrast, there was no statistically significant difference between the genotypic and allelic frequencies for other genes in patients with inhibitors compared to patients without inhibitors. These results demonstrate that only polymorphisms in F13A1, DOCK2, and MAPK9 genes are associated with the risk of developing FVIII inhibitors in Iranian HA patients.


Asunto(s)
Alelos , Inhibidores de Factor de Coagulación Sanguínea/genética , Frecuencia de los Genes , Factores de Intercambio de Guanina Nucleótido/genética , Hemofilia A/genética , Proteína Quinasa 9 Activada por Mitógenos/genética , Polimorfismo Genético , Adulto , Factor VIII/genética , Proteínas Activadoras de GTPasa , Humanos , Irán , Masculino
17.
J Neurochem ; 140(1): 151-169, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27787886

RESUMEN

Oxidative stress, resulting from accumulation of reactive oxygen species, plays a critical role in astroglial cell death occurring in diverse neuropathological conditions. Numerous studies indicate that neuroglobin (Ngb) promotes neuron survival, but nothing is known regarding the action of Ngb in astroglial cell survival. Thus, the purpose of this study was to investigate the potential glioprotective effect of Ngb on hydrogen peroxide (H2 O2 )-induced oxidative stress and apoptosis in cultured mouse astrocytes. Incubation of cells with subnanomolar concentrations of Ngb (10-14 -10-10  M) was found to prevent both H2 O2 -evoked reduction in surviving cells number and accumulation of reactive oxygen species in a concentration-dependent manner. Furthermore, Ngb treatment abolishes H2 O2 -induced increase in mitochondrial oxygen consumption rates. Concomitantly, Ngb treatment rescues H2 O2 -associated reduced expression of endogenous antioxidant enzymes (superoxide dismutases and catalase) and prevents the stimulation of the expression of pro-inflammatory genes (inducible nitric oxide synthase, cyclooxygenase-2, and interleukin (IL) IL-6 and IL-33). Moreover, Ngb blocks the stimulation of Bax (pro-apoptotic) and the inhibition of Bcl-2 (anti-apoptotic) gene expression induced by H2 O2 , which in turn abolishes caspase 3 activation. The protective effect of Ngb upon H2 O2 induced activation of caspase 3 activity and cell death can be accounted for by activation of protein kinase A and mitogen-activated protein kinase transduction cascade. Finally, we demonstrate that Ngb increases Akt phosphorylation and prevents H2 O2 -provoked inhibition of ERK and Akt phosphorylation. Taken together, these data demonstrate for the first time that Ngb is a glioprotective agent that prevents H2 O2 -induced oxidative stress and apoptotic astroglial cell death. Protection of astrocytes from oxidative insult may thus contribute to the neuroprotective effect of Ngb.


Asunto(s)
Apoptosis/efectos de los fármacos , Astrocitos/efectos de los fármacos , Globinas/farmacología , Peróxido de Hidrógeno/toxicidad , Proteínas del Tejido Nervioso/farmacología , Fármacos Neuroprotectores/farmacología , Estrés Oxidativo/efectos de los fármacos , Animales , Apoptosis/fisiología , Astrocitos/metabolismo , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Células Cultivadas , Relación Dosis-Respuesta a Droga , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Neuroglobina , Estrés Oxidativo/fisiología
18.
Toxicol Appl Pharmacol ; 316: 36-47, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-28025110

RESUMEN

Early-life exposure to arsenic increases risk of developing a variety of non-malignant and malignant diseases. Arsenic-induced carcinogenesis may be mediated through epigenetic mechanisms and pathways leading to inflammation. Our previous study reported that prenatal arsenic exposure leads to increased mRNA expression of several genes related to inflammation, including COX2, EGR1, and SOCS3. This study aimed to investigate the effects of arsenic exposure on promoter DNA methylation and mRNA expression of these inflammatory genes (COX2, EGR1, and SOCS3), as well as the generation of 8-nitroguanine, which is a mutagenic DNA lesion involved in inflammation-related carcinogenesis. Prenatally arsenic-exposed newborns had promoter hypomethylation of COX2, EGR1, and SOCS3 in cord blood lymphocytes (p<0.01). A follow-up study in these prenatally arsenic-exposed children showed a significant hypomethylation of these genes in salivary DNA (p<0.01). In vitro experiments confirmed that arsenite treatment at short-term high doses (10-100µM) and long-term low doses (0.5-1µM) in human lymphoblasts (RPMI 1788) caused promoter hypomethylation of these genes, which was in concordance with an increase in their mRNA expression. Additionally, the level of urinary 8-nitroguanine was significantly higher (p<0.01) in exposed newborns and children, by 1.4- and 1.8-fold, respectively. Arsenic accumulation in toenails was negatively correlated with hypomethylation of these genes and positively correlated with levels of 8-nitroguanine. These results indicated that early-life exposure to arsenic causes hypomethylation of COX2, EGR1, and SOCS3, increases mRNA expression of these genes, and increases 8-nitroguanine formation. These effects may be linked to mechanisms of arsenic-induced inflammation and cancer development later in life.


Asunto(s)
Arsénico/toxicidad , Ciclooxigenasa 2/metabolismo , Metilación de ADN/fisiología , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Guanina/análogos & derivados , Proteína 3 Supresora de la Señalización de Citocinas/metabolismo , Biomarcadores/metabolismo , Biomarcadores/orina , Niño , Ciclooxigenasa 2/genética , Metilación de ADN/efectos de los fármacos , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Exposición a Riesgos Ambientales , Femenino , Sangre Fetal/efectos de los fármacos , Sangre Fetal/metabolismo , Estudios de Seguimiento , Guanina/orina , Humanos , Recién Nacido , Mediadores de Inflamación/metabolismo , Masculino , Uñas/química , Uñas/efectos de los fármacos , Uñas/metabolismo , Embarazo , Proteína 3 Supresora de la Señalización de Citocinas/genética , Tailandia
19.
J Clin Periodontol ; 44(9): 905-914, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28736819

RESUMEN

OBJECTIVE: To evaluate the influence of periodontal therapy on DNA methylation in patients with chronic periodontitis as compared to healthy individuals. MATERIAL AND METHODS: Twenty patients were enrolled into two groups: (i) 10 diagnosed as clinically healthy; and (ii) 10 diagnosed with chronic periodontitis. Clinical measures were recorded and gingival biopsies were harvested at baseline (both patient groups) and at 2 and 8 weeks post-baseline for diseased individuals. Molecular DNA methylation analysis was performed by pyrosequencing for the putative inflammation-associated genes LINE-1, COX-2, IFN-γ and TNF-α. Random-intercept linear regression models were applied to evaluate methylation levels across groups at baseline and the methylation changes over time in the diseased and normal tissues. RESULTS: Periodontal therapy did not influence gene expression methylation of TNF-α, IFN-γ and LINE-1 levels at normal and periodontitis sites over time. However, it significantly reduced COX-2 methylation levels comparable to healthy individuals at both 2 and 8 weeks post-treatment (p < .05). CONCLUSIONS: Periodontal therapy resets the DNA methylation status of inflammatory gene for COX-2 in patients with periodontal disease. DNA methylation levels of TNF-α, IFN-γ and LINE-1 were sustained in periodontitis sites despite therapy. Future studies should consider an expanded panel of inflammatory genes over time. (ClinicalTrials.gov NCT02835898).


Asunto(s)
Periodontitis Crónica/genética , Periodontitis Crónica/terapia , Metilación de ADN , Adulto , Anciano , Estudios de Casos y Controles , Ciclooxigenasa 2/genética , Femenino , Perfilación de la Expresión Génica , Humanos , Interferón gamma/genética , Elementos de Nucleótido Esparcido Largo/genética , Masculino , Persona de Mediana Edad , Proyectos Piloto , Estudios Prospectivos , Factor de Necrosis Tumoral alfa/genética
20.
J Neuroinflammation ; 13(1): 289, 2016 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-27832807

RESUMEN

BACKGROUND: While exercise effects on the immune system have received increasing attention in recent years, it remains unclear to what extent gender and fluctuations in sex hormones during menstrual cycle influence immunological responses to exercise. METHODS: We investigated mRNA changes induced through exhaustive exercise (half-marathon; pre-exercise and post-exercise [30 min, 3 h, 24 h] on whole blood cultures ± lipopolysaccharide [LPS] [1 h]) with a specific focus on sex differences (men vs women in luteal phase) as an extension of our previous study. RESULTS: Inflammation related signaling pathways, TLRs, cytosolic DNA sensing and RIG-I like receptors were differentially activated between sexes in LPS-stimulated cultures. Genes differentially regulated between sexes included TNIP-1, TNIP-3, IL-6, HIVEP1, CXCL3, CCR3, IL-8, and CD69, revealing a bias towards less anti-inflammatory gene regulation in women compared to men. In addition, several genes relevant to brain function (KMO, DDIT4, VEGFA, IGF1R, IGF2R, and FGD4) showed differential activation between sexes. Some of these genes (e.g., KMO in women, DDIT4 in both sexes) potentially constitute neuroprotective mechanisms. CONCLUSIONS: These data reveal that the exercise-induced change in gene expression might be gender and menstrual cycle phase dependent.


Asunto(s)
Citocinas/metabolismo , Endotoxinas/farmacología , Ejercicio Físico , Expresión Génica/efectos de los fármacos , Leucocitos/efectos de los fármacos , Leucocitos/metabolismo , Transducción de Señal/efectos de los fármacos , Adulto , Antropometría , Atletas , Células Cultivadas , Citocinas/genética , Femenino , Perfilación de la Expresión Génica , Hormonas , Humanos , Lipopolisacáridos/farmacología , Masculino , Ciclo Menstrual/fisiología , Factores Sexuales , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA