Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 564
Filtrar
1.
PLoS One ; 17(2): e0264385, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35202450

RESUMEN

Interactions between interleukin (IL)-8 and its receptors, CXCR1, and CXCR2, serve crucial roles in inflammatory conditions and various types of cancers. Inhibition of this signaling pathway has been exploited as a promising strategy in treating these diseases. However, most studies only focused on the design of allosteric antagonists-bound receptors on the intracellular side of IL-8 receptors. Recently, the first cryo-EM structures of IL-8-CXCR2-Gi complexes have been solved, revealing the unique binding and activation modes of the endogenous chemokine IL-8. Hence, we set to identify small molecule inhibitors for IL-8 using critical protein-protein interaction between IL-8 and CXCR2 at the orthosteric binding site. The pharmacophore models and molecular docking screened compounds from DrugBank and NCI databases. The oral bioavailability of the top 23 ligands from the screening was then predicted by the SwissAMDE tool. Molecular dynamics simulation and free binding energy calculation were performed for the best compounds. The result indicated that DB14770, DB12121, and DB03916 could form strong interactions and stable protein-ligand complexes with IL-8. These three candidates are potential IL-8 inhibitors that can be further evaluated by in vitro experiments in the next stage.


Asunto(s)
Quimiocinas/metabolismo , Interleucina-8/antagonistas & inhibidores , Receptores de Interleucina-8B/metabolismo , Dominio Catalítico/efectos de los fármacos , Biología Computacional , Simulación por Computador , Humanos , Modelos Moleculares , Simulación del Acoplamiento Molecular
2.
BMC Cancer ; 21(1): 1283, 2021 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-34847866

RESUMEN

BACKGROUND: Cancer stem cells (CSCs) drive tumor initiation and progression and participate in tumor chemoresistance. We recently discovered that oral squamous cell carcinoma (OSCC) cells that highly express CD10 (CD10H cells) present cancer stem cells (CSC)-associated characteristics, which, in turn, affect the tumor growth, epithelial-mesenchymal transition (EMT), and resistance to cisplatin. In this study, we further investigated this mechanism in vitro and in vivo. We hypothesized that IL8 might regulate migration, invasion, and cisplatin resistance of CD10-positive oral cancer cells through the ERK pathway. METHODS: CD10 MicroBead Kit was used to select HN6 cells with high and low expression of CD10. The target protein IL8 was screened via protein chip assay. Lentiviral transduction and specific inhibitor were applied to investigate the signaling pathway. Real-time PCR, Western blot, and immunohistochemistry were used to analyze the mRNA and protein expression; transwell assay, spheroid formation assay, and cell viability assay were used to study the cell biological behavior in vitro; xenograft animal model was used to evaluate the tumor formation rate in vivo. RESULTS: Overexpression of CD10 promoted CSC-related genes expression and enhanced migration, invasion, spheroid formation, and chemoresistance in HN6 cells. Moreover, the overexpression of IL8 was detected in OSCC tumor tissue and cell lines (HN6 and CAL27) overexpressing CD10. IL8 secreted by CD10H HN6 promoted migration and invasion and restored tumor chemosensitivity via the p-ERK signaling pathway, while the inhibition of IL8 increased the chemosensitivity to cisplatin. CONCLUSIONS: IL8 secretion by CD10 positive cells promotes migration, invasion, and cisplatin resistance of OSCC via the p-ERK signaling pathway.


Asunto(s)
Resistencia a Antineoplásicos , Interleucina-8/metabolismo , Neoplasias de la Boca/metabolismo , Neprilisina/metabolismo , Carcinoma de Células Escamosas de Cabeza y Cuello/metabolismo , eIF-2 Quinasa/metabolismo , Animales , Antineoplásicos/uso terapéutico , Comunicación Celular , Movimiento Celular , Cisplatino/uso terapéutico , Femenino , Expresión Génica , Xenoinjertos , Humanos , Interleucina-8/antagonistas & inhibidores , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neoplasias de la Boca/tratamiento farmacológico , Neoplasias de la Boca/patología , Invasividad Neoplásica , Trasplante de Neoplasias , Células Madre Neoplásicas , Transducción de Señal , Esferoides Celulares/patología , Carcinoma de Células Escamosas de Cabeza y Cuello/tratamiento farmacológico , Carcinoma de Células Escamosas de Cabeza y Cuello/patología
3.
Curr Issues Mol Biol ; 43(1): 187-196, 2021 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-34071008

RESUMEN

To evaluate the effects of allicin on mediators of pain secreted by oral cancer cells in vitro, single-cell suspensions were prepared by enzymatic method from oral squamous cell carcinoma (OSCC). Cancer stem cells were isolated by the CD133+ selection method with magnetic cell sorting. Stemness markers were checked in both cancer cells and cancer stem cells by RT-PCR. Comparative analysis of pain mediators TNF-alpha, IL-8, and endothelin at both RNA and protein levels for normal epithelial cells, cancer cells, and cancer stem cells was carried out with and without allicin treatment. CD133 and CD44 expression levels were checked in cancer cells and cancer stem cells flow cytometrically. Allicin inhibited both gene and protein expression of TNF-alpha, IL-8, and endothelin in both cancer cells and cancer stem cells. Allicin is more likely to be a promising treatment in alleviating the levels of pain and inflammation in OSCCs.


Asunto(s)
Dolor en Cáncer/tratamiento farmacológico , Disulfuros/farmacología , Endotelinas/antagonistas & inhibidores , Interleucina-8/antagonistas & inhibidores , Neoplasias de la Boca/fisiopatología , Células Madre Neoplásicas/efectos de los fármacos , Ácidos Sulfínicos/farmacología , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Antígeno AC133/metabolismo , Antioxidantes/farmacología , Dolor en Cáncer/etiología , Dolor en Cáncer/metabolismo , Dolor en Cáncer/patología , Humanos , Neoplasias de la Boca/tratamiento farmacológico , Neoplasias de la Boca/patología , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Cultivo Primario de Células
5.
Biol Chem ; 402(4): 461-468, 2021 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-33938176

RESUMEN

The chemoresistance is one of the major challenges for acute myeloid leukemia (AML) treatment. We found that the expression of histone deacetylase 8 (HDAC8) was increased in daunorubicin (DNR) resistant AML cells, while targeted inhibition of HDAC8 by its specific siRNA or inhibitor can restore sensitivity of DNR treatment . Further, targeted inhibition of HDAC8 can suppress expression of interleukin 6 (IL-6) and IL-8. While recombinant IL-6 (rIL-6) and rIL-8 can reverse si-HDAC8-resored DNR sensitivity of AML cells. Mechanistical study revealed that HDAC8 increased the expression of p65, one of key components of NF-κB complex, to promote the expression of IL-6 and IL-8. It might be due to that HDAC8 can directly bind with the promoter of p65 to increase its transcription and expression. Collectively, our data suggested that HDAC8 promotes DNR resistance of human AML cells via regulation of IL-6 and IL-8.


Asunto(s)
Antibióticos Antineoplásicos/farmacología , Daunorrubicina/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Histona Desacetilasas/metabolismo , Leucemia Mieloide Aguda/tratamiento farmacológico , Proteínas Represoras/metabolismo , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Histona Desacetilasas/genética , Humanos , Interleucina-6/antagonistas & inhibidores , Interleucina-6/genética , Interleucina-6/metabolismo , Interleucina-8/antagonistas & inhibidores , Interleucina-8/genética , Interleucina-8/metabolismo , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , ARN Interferente Pequeño/farmacología , Proteínas Represoras/antagonistas & inhibidores , Proteínas Represoras/genética , Células Tumorales Cultivadas
6.
Int J Mol Sci ; 22(7)2021 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-33807391

RESUMEN

Salivary levels of interleukin-8 (IL-8) are elevated in patients with periodontitis. Caffeic acid phenethyl ester (CAPE) improves the periodontal status in subjects. However, whether CAPE can reduce IL-8 expression is unclear. We collected saliva to determine proinflammatory cytokine levels and used subgingival calculus and surrounding tissues from patients with periodontitis for oral microbiota analysis via 16s ribosomal RNA gene sequencing. THP-1 cells were stimulated with sterile-filtered saliva from patients, and target gene/protein expression was assessed. IL-8 mRNA expression was analyzed in saliva-stimulated THP-1 cells treated with CAPE and the heme oxygenase-1 (HO-1) inhibitor tin-protoporphyrin (SnPP). In 72 symptomatic individuals, IL-8 was correlated with periodontal inflammation (bleeding on probing, r = 0.45; p < 0.001) and disease severity (bleeding on probing, r = 0.45; p < 0.001) but not with the four oral microbiota species tested. Reduced salivary IL-8 secretion was correlated with effective periodontitis treatment (r = 0.37, p = 0.0013). In THP-1 cells, saliva treatment induced high IL-8 expression and IKK2 and nuclear factor-κB (NF-κB) phosphorylation. However, the IKK inhibitor BMS-345541, NF-κB inhibitor BAY 11-7082, and CAPE attenuated saliva-induced IL-8 expression. CAPE induced HO-1 expression and inhibited IKK2, IκBα, and NF-κB phosphorylation. Blocking HO-1 decreased the anti-inflammatory activity of CAPE. The targeted suppression of IL-8 production using CAPE reduces inflammation and periodontitis.


Asunto(s)
Ácidos Cafeicos/farmacología , Interleucina-8/metabolismo , Periodontitis/tratamiento farmacológico , Alcohol Feniletílico/análogos & derivados , Antiinflamatorios/farmacología , Ácidos Cafeicos/metabolismo , Citocinas/metabolismo , Hemo-Oxigenasa 1/metabolismo , Humanos , Proteínas I-kappa B/metabolismo , Inflamación/tratamiento farmacológico , Interleucina-8/antagonistas & inhibidores , Lipopolisacáridos/metabolismo , Inhibidor NF-kappaB alfa/metabolismo , FN-kappa B/metabolismo , Periodontitis/inmunología , Periodontitis/metabolismo , Alcohol Feniletílico/metabolismo , Alcohol Feniletílico/farmacología , Fosforilación/efectos de los fármacos , Saliva/química , Células THP-1
7.
Oncol Rep ; 45(3): 1284-1294, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33650662

RESUMEN

The cancer microenvironment exhibits local acidosis compared with the surrounding normal tissue. Many reports have shown that acidosis accelerates the invasiveness and metastasis of cancer, yet the underlying molecular mechanisms remain unclear. In the present study, we focused on acid-induced functional changes through acid receptors in breast cancer cells. Acidic treatment induced interleukin (IL)-8 expression in MDA-MB-231 cells and promoted cell migration and invasion. The acidic microenvironment elevated matrix metalloproteinase (MMP)-2 and MMP-9 activity, and addition of IL-8 had similar effects. However, inhibition of IL-8 suppressed the acid-induced migration and invasion of MDA-MB-231 cells. MDA-MB-231 cells express various acid receptors including ion channels and G protein-coupled receptors. Interestingly, acidic stimulation increased the expression of acid-sensing ion channel 1 (ASIC1), and acid-induced IL-8 was significantly decreased by ASIC1 knockdown. Moreover, phosphorylation of nuclear factor (NF)-κB was induced by acidic treatment, and inhibition of NF-κB activation reduced acid-induced IL-8 expression. These results suggest that IL-8 induction by an acidic microenvironment promotes breast cancer development and that ASIC1 might be a novel therapeutic target for breast cancer metastasis.


Asunto(s)
Canales Iónicos Sensibles al Ácido/metabolismo , Neoplasias de la Mama/patología , Interleucina-8/metabolismo , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Microambiente Tumoral , Canales Iónicos Sensibles al Ácido/genética , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Medios de Cultivo/química , Medios de Cultivo/farmacología , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Concentración de Iones de Hidrógeno , Interleucina-8/antagonistas & inhibidores , Interleucina-8/genética , Fosforilación/efectos de los fármacos , Factor de Transcripción ReIA/metabolismo , Microambiente Tumoral/efectos de los fármacos
8.
PLoS Comput Biol ; 17(2): e1008257, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33596197

RESUMEN

Lung cancer is one of the leading causes of cancer-related deaths worldwide and is characterized by hijacking immune system for active growth and aggressive metastasis. Neutrophils, which in their original form should establish immune activities to the tumor as a first line of defense, are undermined by tumor cells to promote tumor invasion in several ways. In this study, we investigate the mutual interactions between the tumor cells and the neutrophils that facilitate tumor invasion by developing a mathematical model that involves taxis-reaction-diffusion equations for the critical components in the interaction. These include the densities of tumor and neutrophils, and the concentrations of signaling molecules and structure such as neutrophil extracellular traps (NETs). We apply the mathematical model to a Boyden invasion assay used in the experiments to demonstrate that the tumor-associated neutrophils can enhance tumor cell invasion by secreting the neutrophil elastase. We show that the model can both reproduce the major experimental observation on NET-mediated cancer invasion and make several important predictions to guide future experiments with the goal of the development of new anti-tumor strategies. Moreover, using this model, we investigate the fundamental mechanism of NET-mediated invasion of cancer cells and the impact of internal and external heterogeneity on the migration patterning of tumour cells and their response to different treatment schedules.


Asunto(s)
Trampas Extracelulares/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Modelos Biológicos , Biología Computacional , Simulación por Computador , Humanos , Técnicas In Vitro , Interleucina-8/antagonistas & inhibidores , Neoplasias Pulmonares/terapia , Invasividad Neoplásica/patología , Metástasis de la Neoplasia/patología , Neutrófilos/metabolismo , Neutrófilos/patología , Receptores de Factores de Crecimiento Transformadores beta/antagonistas & inhibidores , Transducción de Señal , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Microambiente Tumoral
9.
Anticancer Res ; 41(2): 783-794, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33517283

RESUMEN

BACKGROUND/AIM: The inflammatory cytokine IL-8 and its receptor CXCR2 are key signalling pathway molecules in cancer development. We hypothesized that IL-8/CXCR2 signalling promotes tumour progression in oesophageal squamous cell carcinoma (ESCC) patients. MATERIALS AND METHODS: We examined the relationship between IL-8/CXCR2 expression and clinicopathological factors by immunohistochemistry in samples from 63 patients with resectable ESCC. The effects of IL-8/CXCR2 signalling on cell proliferation and gene expression were examined in vitro and in vivo using ESCC cell lines. RESULTS: Increased IL-8/CXCR2 signalling was associated with shorter overall survival (p<0.05) and recurrence-free survival (p<0.05) in ESCC patients. Multivariate analysis identified IL-8/CXCR2 expression as a prognostic factor for surgically treated ESCC (p<0.05). In vitro, IL-8 exposure or over-expression significantly enhanced ESCC cell proliferation. SB225002, a CXCR2-specific antagonist, and IL-8 siRNA significantly suppressed cell proliferation. CONCLUSION: IL-8/CXCR2 expression is an independent prognostic factor for surgically treated ESCC, and IL-8/CXCR2 signalling contributes to ESCC cell proliferation.


Asunto(s)
Neoplasias Esofágicas/cirugía , Carcinoma de Células Escamosas de Esófago/cirugía , Interleucina-8/metabolismo , Receptores de Interleucina-8B/metabolismo , Regulación hacia Arriba , Anciano , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/patología , Carcinoma de Células Escamosas de Esófago/metabolismo , Carcinoma de Células Escamosas de Esófago/patología , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Interleucina-8/antagonistas & inhibidores , Interleucina-8/genética , Masculino , Ratones , Persona de Mediana Edad , Trasplante de Neoplasias , Compuestos de Fenilurea/farmacología , Pronóstico , ARN Interferente Pequeño/farmacología , Receptores de Interleucina-8B/antagonistas & inhibidores , Receptores de Interleucina-8B/genética , Transducción de Señal/efectos de los fármacos , Análisis de Supervivencia
10.
Front Immunol ; 12: 790925, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34975889

RESUMEN

Extracts from Taiwan's traditional medicinal mushroom, Antrodia cinnamomea, exhibit anti-inflammatory activities in cellular and preclinical studies. However, this paper is the first to report that Antcin K, a triterpenoid isolated from A. cinnamomea, inhibits proinflammatory cytokine production in human rheumatoid synovial fibroblasts (RASFs), which are major players in rheumatoid arthritis (RA) disease. In our analysis of the mechanism of action, Antcin K inhibited the expression of three cytokines (tumor necrosis factor alpha [TNF-α], interleukin 1 beta [IL-1ß] and IL-8) in human RASFs; cytokines that are crucial to RA synovial inflammation. Notably, incubation of RASFs with Antcin K reduced the phosphorylation of the focal adhesion kinase (FAK), phosphoinositide 3-kinase (PI3K), protein kinase B (AKT) and nuclear factor-κB (NF-κB) signaling cascades, all of which promote cytokine production in RA. Intraperitoneal injections of Antcin K (10 mg/kg or 30 mg/kg) attenuated paw swelling, cartilage degradation and bone erosion, and decreased serum levels of TNF-α, IL-1ß, IL-8 in collagen-induced arthritis (CIA) mice; in further experiments, IL-6 levels were similarly reduced. The inhibitory effects of Antcin K upon TNF-α, IL-1ß and IL-8 expression in human RASFs was achieved through the downregulation of the FAK, PI3K, AKT and NF-κB signaling cascades. Our data support clinical investigations using Antcin K in RA disease.


Asunto(s)
Artritis Reumatoide/tratamiento farmacológico , Cartílago/metabolismo , Colestenos/farmacología , Citocinas/antagonistas & inhibidores , Membrana Sinovial/efectos de los fármacos , Animales , Células Cultivadas , Colestenos/uso terapéutico , Citocinas/biosíntesis , Fibroblastos/efectos de los fármacos , Fibroblastos/inmunología , Humanos , Interleucina-1beta/antagonistas & inhibidores , Interleucina-8/antagonistas & inhibidores , Ratones , Ratones Endogámicos C57BL , FN-kappa B/fisiología , Fosfatidilinositol 3-Quinasas/fisiología , Proteínas Proto-Oncogénicas c-akt/fisiología , Membrana Sinovial/inmunología , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores
11.
Mol Carcinog ; 60(1): 51-59, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33264466

RESUMEN

Triple-negative breast cancer (TNBC) is the most aggressive subtype of the disease with lack of recognized molecular targets for therapy. TNBC cells are known to secrete high levels of the proinflammatory cytokines interleukin-6 (IL-6) and IL-8, which promote angiogenesis and favor the growth and spread of the disease. In the present study, we have shown that the humanized anti-IL-6 receptor tocilizumab (Actemra) is also a potent inhibitor of IL-8 in TNBC cells. Similar effect was also obtained by specific IL-6 inhibition either by small interfering RNA or by neutralizing antibody. Likewise, neutralizing IL-8 with specific antibody downregulated IL-8 and inhibited the IL-6/signal transducer and activator of transcription 3 and nuclear factor-κB pathways. Interestingly, simultaneous co-inhibition of IL-6 and IL-8 did not increase the effects of the single inhibitors. Additionally, we present clear evidence that tocilizumab has potent antiangiogenic effect. Indeed, tocilizumab abolished the ability of TNBC cells to induce the differentiation of endothelial cells into network-like tubular structures in vitro and impaired neovascularization in humanized breast orthotopic tumor xenografts. This was associated with tocilizumab-dependent downregulation of the main proangiogenic factor vascular endothelial growth factor A and its coactivator hypoxia-inducible factor 1 both in vitro and in vivo. Therefore, tocilizumab could be of great therapeutic value for TNBC patients through targeting angiogenesis.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Anticuerpos Monoclonales Humanizados/farmacología , Antineoplásicos Inmunológicos/farmacología , Interleucina-8/antagonistas & inhibidores , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Inhibidores de la Angiogénesis/uso terapéutico , Animales , Antineoplásicos Inmunológicos/uso terapéutico , Línea Celular Tumoral , Femenino , Humanos , Interleucina-8/inmunología , Ratones Endogámicos NOD , Ratones SCID , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/inmunología , Neovascularización Patológica/patología , Neoplasias de la Mama Triple Negativas/irrigación sanguínea , Neoplasias de la Mama Triple Negativas/inmunología , Neoplasias de la Mama Triple Negativas/patología
12.
Artículo en Inglés | MEDLINE | ID: mdl-33186640

RESUMEN

Low grade neuroinflammation has been suggested as one of the underlying mechanisms of many psychiatric diseases as well as cognitive disorders. Interleukin 8 (IL-8), a proinflammatory cytokine produced by many cell types including macrophage and microglia, mainly functions as a neutrophil chemoattractant in the bloodstream. IL-8 is also found in the brain, where it is released from microglia in response to proinflammatory stimuli. In this review, we highlight studies focusing on the role of IL-8 in psychiatric diseases such as major depression, bipolar disorder, schizophrenia, sleep disorder, autism spectrum disorder, anxiety disorders and dementia. Increased peripheral IL-8 levels have been reported in these diseases, particularly in schizophrenic disorder, bipolar disorder, obstructive sleep apnea and autism spectrum disorder. The literature on IL-8 and major depression is inconsistent. IL-8 has been found to be a factor associated with schizophrenic prognosis and therapeutic response, and may affect a wide range of symptomatology. Considering that the exact role of immune alterations is still under research, the success of immune-based therapies in psychiatric diseases is limited for the time being.


Asunto(s)
Depresión/inmunología , Depresión/terapia , Mediadores de Inflamación/inmunología , Interleucina-8/inmunología , Animales , Trastornos de Ansiedad/inmunología , Trastornos de Ansiedad/metabolismo , Trastornos de Ansiedad/terapia , Depresión/metabolismo , Humanos , Inmunoterapia/métodos , Inmunoterapia/tendencias , Mediadores de Inflamación/antagonistas & inhibidores , Mediadores de Inflamación/metabolismo , Interleucina-8/antagonistas & inhibidores , Interleucina-8/metabolismo , Trastornos Mentales/inmunología , Trastornos Mentales/metabolismo , Trastornos Mentales/terapia , Enfermedades Neuroinflamatorias/inmunología , Enfermedades Neuroinflamatorias/metabolismo , Enfermedades Neuroinflamatorias/terapia , Esquizofrenia/inmunología , Esquizofrenia/metabolismo , Esquizofrenia/terapia , Trastornos del Sueño-Vigilia/inmunología , Trastornos del Sueño-Vigilia/metabolismo , Trastornos del Sueño-Vigilia/terapia
13.
Vet Microbiol ; 251: 108892, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33126183

RESUMEN

Porcine epidemic diarrhea virus (PEDV) is an enveloped, single-stranded positive-sense RNA virus that belongs to a porcine entero-pathogenic alphacoronavirus, causing lethal watery diarrhea in piglets. Despite existing study reports the sole accessory protein ORF3 identified as NF-κB antagonist, the contribution of PEDV ORF3 to production of the pro-inflammatory cytokines mediated by NF-κB signaling remains largely unknown. Thus in this present study, we showed that PEDV ORF3 protein significantly inhibited the productions of pro-inflammatory cytokines interleukin-6 (IL-6) and IL-8. The phosphorylation of IκBα was inhibited by ORF3 protein, but no degradation of IκBα was induced in ORF3-expressing cells. Furthermore, PEDV ORF3 inhibited NF-κB activation through preventing nuclear factor p65 phosphorylation and down-regulating p65 expression level, as well as interfering nuclear translocation of p65, eventually resulting into the inhibition of IL-6 and IL-8 production. Our study definitely links PEDV ORF3 to inhibition of pro-inflammatory cytokines production, which will provide new insight into the molecular mechanisms of NF-κB activity inhibited by PEDV proteins to facilitate virus evasion of host innate immune.


Asunto(s)
Interleucina-6/antagonistas & inhibidores , Interleucina-8/antagonistas & inhibidores , Virus de la Diarrea Epidémica Porcina/genética , Factor de Transcripción ReIA/genética , Proteínas Virales/genética , Proteínas Virales/inmunología , Animales , Chlorocebus aethiops , Células HEK293 , Células HeLa , Humanos , Interleucina-6/inmunología , Interleucina-8/inmunología , Virus de la Diarrea Epidémica Porcina/inmunología , Transducción de Señal , Porcinos , Factor de Transcripción ReIA/inmunología , Células Vero , Replicación Viral
14.
J Cancer Res Clin Oncol ; 146(12): 3255-3268, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32870388

RESUMEN

PURPOSE: Deoxycholic acid (DCA), a secondary bile acid, is reportedly increased in the serum of patients with nonalcoholic steatohepatitis and animals with experimentally induced hepatocellular carcinoma (HCC), but its contribution to malignant behaviors of HCC has not been precisely clarified. This study aimed to examine the effect of DCA on hepatic stellate cells (HSCs), a major component of nonparenchymal cells in the liver, and its subsequent indirect effect on HCC cells. METHODS: LX2 cells, a human HSC line, were treated with DCA in vitro. Then, HuH7 cells, a human hepatoma cell line, were incubated in conditioned media of DCA-treated LX2 to investigate the subsequent effect focusing on malignant behaviors. RESULTS: DCA resulted in cellular senescence in LX2 with the decreased cell proliferation via cell cycle arrest at G0/1 phase, together with the induction of senescence-associated secretory phenotype (SASP) factors. To investigate the influence of SASP factors secreted by HSCs in response to DCA, HCC cells were treated with conditioned media that promoted cell migration and invasion via induction of epithelial mesenchymal transition. These changes were attenuated in the presence of neutralizing antibody against IL8 or TGFß. Pathological analysis of surgical specimens from HCC patients revealed that senescent HSCs were detected in the stroma surrounding HCC. CONCLUSION: Our data suggest an important role of HSC senescence caused by DCA for the malignant biological behaviors of HCC via induction of SASP factors, particularly IL8 and TGFß.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Ácido Desoxicólico/farmacología , Células Estrelladas Hepáticas/efectos de los fármacos , Neoplasias Hepáticas/metabolismo , Anticuerpos Neutralizantes/farmacología , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Senescencia Celular/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Células Estrelladas Hepáticas/metabolismo , Humanos , Interleucina-8/antagonistas & inhibidores , Interleucina-8/inmunología , Hígado/efectos de los fármacos , Hígado/patología , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Factor de Crecimiento Transformador beta/inmunología
15.
J Am Soc Mass Spectrom ; 31(1): 109-116, 2020 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-32881511

RESUMEN

The polypeptide chemokine Interleukin-8 (IL8) plays a crucial role in inflammatory processes in humans. IL8 is involved in chronic inflammatory lung diseases, rheumatoid arthritis, and cancer. Previous studies have shown that the interaction of IL8 with its natural receptors CXCR1 and CXCR2 is critical in these diseases. Antibodies have been used to study the receptor interaction of IL8; however, the binding epitopes were hitherto unknown. Identification of the antibody epitope(s) could lead to a molecular understanding of the inhibiting mechanism and development of improved inhibitors. Here, we report the epitope identification and the affinity characterization of IL8 to a monoclonal anti-human IL8 antibody inhibiting the receptor binding by a combination of surface plasmon resonance (SPR) biosensor analysis and MALDI-mass spectrometry. SPR determination of IL8 with the immobilized antibody revealed high affinity (KD, 82.2 nM). Epitope identification of IL-8 was obtained by proteolytic epitope-extraction mass spectrometry of the peptide fragments upon high pressure trypsin digestion, using an affinity microcolumn with immobilized anti-IL-8 antibody. MALDI-MS of the affinity-bound peptide elution fraction revealed an assembled (discontinuous) epitope comprising two specific peptides, IL8 [12-20] and IL8 [55-60]. Identical epitope peptides were identified by direct MALDI-MS of the eluted epitope fraction from the immobilized anti-IL8 antibody on the SPR chip. SPR determination of the synthetic epitope peptides provided high affinities confirming their binding specificity. The previously reported finding that the anti-Il8 antibody is inhibiting the IL8-CXCR1 interaction is well consistent with the overlapping region of epitope interactions identified in the present study.


Asunto(s)
Anticuerpos/inmunología , Epítopos/inmunología , Interleucina-8/inmunología , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción/métodos , Anticuerpos/química , Anticuerpos/metabolismo , Técnicas Biosensibles , Cromatografía de Afinidad , Epítopos/química , Humanos , Interleucina-8/antagonistas & inhibidores , Interleucina-8/química , Fragmentos de Péptidos/síntesis química , Fragmentos de Péptidos/química , Fragmentos de Péptidos/inmunología , Mapeo Peptídico/métodos , Resonancia por Plasmón de Superficie
16.
Ann Allergy Asthma Immunol ; 125(5): 503-504, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32585180

Asunto(s)
Corticoesteroides/uso terapéutico , Antiasmáticos/uso terapéutico , Asma/tratamiento farmacológico , Infecciones por Coronavirus/prevención & control , Síndrome de Liberación de Citoquinas/prevención & control , Dexametasona/uso terapéutico , Pandemias/prevención & control , Neumonía Viral/prevención & control , Administración por Inhalación , Enzima Convertidora de Angiotensina 2 , Asma/inmunología , Asma/patología , Asma/virología , Betacoronavirus/efectos de los fármacos , Betacoronavirus/inmunología , Betacoronavirus/patogenicidad , COVID-19 , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/patología , Infecciones por Coronavirus/virología , Síndrome de Liberación de Citoquinas/inmunología , Síndrome de Liberación de Citoquinas/patología , Síndrome de Liberación de Citoquinas/virología , Endorribonucleasas/antagonistas & inhibidores , Endorribonucleasas/genética , Endorribonucleasas/inmunología , Regulación de la Expresión Génica/efectos de los fármacos , Interacciones Huésped-Patógeno/efectos de los fármacos , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Interleucina-6/antagonistas & inhibidores , Interleucina-6/genética , Interleucina-6/inmunología , Interleucina-8/antagonistas & inhibidores , Interleucina-8/genética , Interleucina-8/inmunología , Peptidil-Dipeptidasa A/genética , Peptidil-Dipeptidasa A/inmunología , Neumonía Viral/inmunología , Neumonía Viral/patología , Neumonía Viral/virología , Receptores Virales/antagonistas & inhibidores , Receptores Virales/genética , Receptores Virales/inmunología , SARS-CoV-2 , Serina Endopeptidasas/genética , Serina Endopeptidasas/inmunología , Índice de Severidad de la Enfermedad , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología , Proteínas no Estructurales Virales/antagonistas & inhibidores , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/inmunología
17.
J Physiol Pharmacol ; 71(1)2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32554850

RESUMEN

Ataxia telangiectasia (A-T) is an inherited neurodegenerative disease caused by mutation in the ataxia telangiectasia mutated (ATM) gene, leading to loss of function in the encoded protein ATM. Because ATM functions to reduce oxidative stress by up-regulating antioxidant enzymes, oxidative stress is a prevalent A-T phenotype and a mediator of the inflammation that drives A-T pathology. Reactive oxygen species (ROS) levels and the expression of pro-inflammatory cytokine interleukin-8 (IL-8) were higher in A-T cells than in normal cells. ROS are related to mitochondrial dysfunction and activation of nuclear factor kappa B (NF-κB) to induce IL-8 expression. Alpha-lipoic acid (α-LA), a naturally occurring thiol compound, shows an antioxidant effect in various cells. This study is aimed to determine if α-LA confers protection against NF-κB activation, IL-8 expression, and mitochondrial dysfunction in A-T cells which are exposed to the inflammatory cytokine IL-1ß. A-T fibroblasts were treated with or without α-LA. The levels of intracellular and mitochondrial ROS, mRNA and protein levels of IL-8, mitochondrial membrane potential (MMP), ATP levels, and DNA binding activity of NF-κB were determined. As a result, IL-1ß increased NF-κB activation, IL-8 expression, intracellular and mitochondrial ROS levels, but decreased MMP and ATP level in A-T cells. Pretreatment of A-T cells with α-LA inhibited IL-1ß-induced activation of NF-κB, IL-8 expression, and mitochondrial dysfunction by reducing ROS levels. In conclusion, supplementation with α-LA may be beneficial for reducing the oxidative stress-induced mitochondrial dysfunction and IL-8 production associated with A-T.


Asunto(s)
Ataxia Telangiectasia/metabolismo , Fibroblastos/metabolismo , Interleucina-1beta/farmacología , Interleucina-8/biosíntesis , Mitocondrias/metabolismo , Ácido Tióctico/farmacología , Antioxidantes/farmacología , Ataxia Telangiectasia/genética , Ataxia Telangiectasia/patología , Línea Celular , Fibroblastos/efectos de los fármacos , Fibroblastos/patología , Expresión Génica , Humanos , Interleucina-8/antagonistas & inhibidores , Interleucina-8/genética , Mitocondrias/efectos de los fármacos , Mitocondrias/patología , Especies Reactivas de Oxígeno/metabolismo
18.
ChemMedChem ; 15(15): 1421-1428, 2020 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-32497366

RESUMEN

Infections from antibiotic-resistant Staphylococcus aureus and Pseudomonas aeruginosa are a serious threat because reduced antibiotic efficacy complicates treatment decisions and prolongs the disease state in many patients. To expand the arsenal of treatments against antimicrobial-resistant (AMR) pathogens, 600-Da branched polyethylenimine (BPEI) can overcome antibiotic resistance mechanisms and potentiate ß-lactam antibiotics against Gram-positive bacteria. BPEI binds cell-wall teichoic acids and disables resistance factors from penicillin binding proteins PBP2a and PBP4. This study describes a new mechanism of action for BPEI potentiation of antibiotics generally regarded as agents effective against Gram-positive pathogens but not Gram-negative bacteria. 600-Da BPEI is able to reduce the barriers to drug influx and facilitate the uptake of a non-ß-lactam co-drug, erythromycin, which targets the intracellular machinery. Also, BPEI can suppress production of the cytokine interleukin IL-8 by human epithelial keratinocytes. This enables BPEI to function as a broad-spectrum antibiotic potentiator, and expands the opportunities to improve drug design, antibiotic development, and therapeutic approaches against pathogenic bacteria, especially for wound care.


Asunto(s)
Antibacterianos/farmacología , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Pseudomonas aeruginosa/efectos de los fármacos , Antibacterianos/química , Relación Dosis-Respuesta a Droga , Células HEK293 , Humanos , Interleucina-8/antagonistas & inhibidores , Interleucina-8/biosíntesis , Pruebas de Sensibilidad Microbiana , Estructura Molecular , Relación Estructura-Actividad
19.
Arch Pharm Res ; 43(5): 526-539, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-28861755

RESUMEN

Rhinoviral infection is associated with an increased risk of asthma attacks. The macrolide clarithromycin decreases cytokine production in nasopharyngeal aspirates from patients with wheezing, but the effects of macrolides on cytokine production in nasal epithelial cells obtained from asthmatic subjects remain unclear. Here, human nasal epithelial cells were infected with type-14 rhinovirus (RV14), a major RV group. Titers and RNA of RV14 and cytokine concentrations, including IL-1ß and IL-6, were higher in the supernatants of the cells obtained from subjects with bronchial asthma (asthmatic group) than in those from the non-asthmatic group. Pretreatment with clarithromycin decreased RV14 titers, viral RNA and cytokine concentrations, and susceptibility to RV14 infection. Pretreatment with clarithromycin also decreased IL-33 production, which was detected after infection. Pretreatment with clarithromycin decreased the expression of intercellular adhesion molecule-1, the receptor for RV14, after infection, the number and fluorescence intensity of the acidic endosomes through which RV RNA enters the cytoplasm, and the activation of nuclear factor kappa-B proteins in nuclear extracts. These findings suggested that RV replication and cytokine production may be enhanced in nasal epithelial cells obtained from subjects with bronchial asthma and may be modulated by clarithromycin.


Asunto(s)
Antivirales/farmacología , Asma/tratamiento farmacológico , Claritromicina/farmacología , Citocinas/biosíntesis , Células Epiteliales/efectos de los fármacos , Rhinovirus/efectos de los fármacos , Asma/metabolismo , Células Cultivadas , Células Epiteliales/metabolismo , Femenino , Humanos , Interleucina-33/antagonistas & inhibidores , Interleucina-33/biosíntesis , Interleucina-6/antagonistas & inhibidores , Interleucina-6/biosíntesis , Interleucina-8/antagonistas & inhibidores , Interleucina-8/biosíntesis , Masculino , Persona de Mediana Edad , Replicación Viral/efectos de los fármacos
20.
Am J Physiol Endocrinol Metab ; 317(6): E1205-E1217, 2019 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-31573846

RESUMEN

Interleukin-8 (IL-8, also named CXCL8) binds to its receptors (CXCR1 and CXCR2) with subsequent recruitment of neutrophils and enhancement of their infiltration into inflamed sites, which exaggerates inflammation in many diseases. Recent studies have proposed that metabolic disorders can be attenuated by counteracting certain inflammatory signal pathways. In this study, we examined whether intervention with G31P, an antagonist of CXCL8, could attenuate tissue inflammation and development of metabolic disorders in db/db mice. The db/m and db/db mice were subcutaneously injected with G31P or equivalent normal saline once a day for 6 wk. The physical and metabolic parameters, glucose tolerance, insulin sensitivity, hepatic lipid accumulation, and inflammation markers were measured. G31P improved hepatic insulin sensitivity by modulating expression of genes related to gluconeogenesis and phosphorylated Akt levels. The expressions of several genes encoding proteins involved in de novo lipogenesis were decreased in G31P-treated db/db mice. Meanwhile, immune cell infiltration and cytokine release were attenuated in db/db mice with G31P treatment. G31P also improved the ratio of proinflammatory M1 and anti-inflammatory M2 macrophages. Furthermore, G31P ameliorates metabolic disturbances via inhibition of CXCR1 and CXCR2 pathways in db/db mice. These data suggest that the selective inhibition of CXC chemokines may have therapeutic effects on symptoms associated with obesity and diabetes.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Resistencia a la Insulina , Interleucina-8/antagonistas & inhibidores , Metabolismo de los Lípidos/efectos de los fármacos , Hígado/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Receptores de Interleucina-8A/antagonistas & inhibidores , Receptores de Interleucina-8B/antagonistas & inhibidores , Animales , Citocinas/efectos de los fármacos , Citocinas/metabolismo , Diabetes Mellitus Tipo 2/genética , Modelos Animales de Enfermedad , Ácidos Grasos no Esterificados/metabolismo , Gluconeogénesis/efectos de los fármacos , Gluconeogénesis/genética , Insulina/metabolismo , Interleucina-6/metabolismo , Interleucina-8/genética , Interleucina-8/metabolismo , Hígado/metabolismo , Hígado/patología , Macrófagos/efectos de los fármacos , Ratones , Fosforilación , Proteínas Proto-Oncogénicas c-akt/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Interleucina-8A/genética , Receptores de Interleucina-8A/metabolismo , Receptores de Interleucina-8B/genética , Receptores de Interleucina-8B/metabolismo , Factor de Necrosis Tumoral alfa/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA