Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 138
Filtrar
1.
Molecules ; 24(13)2019 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-31277363

RESUMEN

Berberine is reported to have multiple biological effects, including antimicrobial, anti-inflammatory, and antitumor activities, and 13-alkyl-substituted berberines show higher activity than berberine against certain bacterial species and human cancer cell lines. In particular, 13-ethylberberine (13-EBR) was reported to have anti-inflammatory effects in endotoxin-activated macrophage and septic mouse models. Thus, in this study, we aimed to examine the anticancer effects of 13-EBR and its mechanisms in radiotherapy-resistant (RT-R) MDA-MB-231 cells derived from the highly metastatic MDA-MB-231 cells. When we compared the gene expression between MDA-MB-231 and RT-R MDA-MB-231 cells with an RNA microarray, RT-R MDA-MB-231 showed higher levels of anti-apoptotic genes and lower levels of pro-apoptotic genes compared to MDA-MB-231 cells. Accordingly, we examined the effect of 13-EBR on the induction of apoptosis in RT-R MDA-MB-231 and MDA-MB-231 cells. The results showed that 13-EBR reduced the proliferation and colony-forming ability of both MDA-MB-231 and RT-R MDA-MB-231 cells. Moreover, 13-EBR induced apoptosis by promoting both intracellular and mitochondrial reactive oxygen species (ROS) and by regulating the apoptosis-related proteins involved in the intrinsic pathway, not in the extrinsic pathway. These results suggest that 13-EBR has pro-apoptotic effects in RT-R MDA-MB-231 and MDA-MB-231 cells by inducing mitochondrial ROS production and activating the mitochondrial apoptotic pathway, providing useful insights into new potential therapeutic strategies for RT-R breast cancer treatment.


Asunto(s)
Apoptosis/efectos de los fármacos , Berberina/farmacología , Neoplasias de la Mama/patología , Neoplasias de la Mama/radioterapia , Mitocondrias/metabolismo , Transducción de Señal , Berberina/química , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Mitocondrias/efectos de los fármacos , Modelos Biológicos , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
2.
Oncol Rep ; 40(6): 3752-3762, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30272295

RESUMEN

Previous studies suggest that cancer stem cells (CSCs) exist in solid tumors, and contribute to therapeutic resistance and disease recurrence. Therefore, the present study aimed to investigate whether radioresistant (RT­R) breast cancer cells derived from breast cancer cells increase the number of CSCs, and whether these CSCs are responsible to increased invasiveness and therapeutic resistance. MCF­7, T47D and MDA­MB­231 cells were irradiated 25 times (2 Gy each; 50 Gy total) to generate radioresistant breast cancer cells (RT­R­MCF­7, RT­R­T47D and RT­R­MDA­MB­231). RT­R­breast cancer cells demonstrated increased cell viability against irradiation and increased colony forming abilities compared with parental breast cancer cells. Particularly, RT­R­MDA­MB­231 cells derived from highly metastatic MDA­MB­231 cells exhibited most radioresistance and chemoresistance of the three cell lines. In addition, MDA­MB­231 cells exhibited the most increased protein levels of CSCs markers cluster of differentiation 44, Notch­4, octamer­binding transcription factor 3/4 and aldehyde dehydrogenase 1, compared with RT­R­MCF­7 cells, suggesting highly metastatic breast cancer cells MDA­MB­231 produce more CSCs. RT­R­MDA­MB­231 cells increased intercellular adhesion molecule­1 and vascular cell adhesion molecule­1 levels, resulting in enhanced migration and adhesion to endothelial cells (ECs), and enhanced invasiveness through ECs by inducing matrix metalloproteinase­9, Snail­1 and ß­catenin, and by downregulating E­cadherin compared with MDA­MB­231 cells. These results suggest that highly metastatic breast cancer cells may increase the number of CSCs following radiation therapy, and CSCs present in RT­R­MDA­MB­231 cells contribute to the enhanced invasiveness by increasing migration, adhesion to ECs and invasion through ECs by promoting epithelial­mesenchymal transition (EMT) via the upregulation of adhesion molecules and EMT­associated proteins.


Asunto(s)
Neoplasias de la Mama/metabolismo , Resistencia a Antineoplásicos , Células Madre Neoplásicas/metabolismo , Tolerancia a Radiación , Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/radioterapia , Adhesión Celular , Línea Celular Tumoral , Supervivencia Celular , Transición Epitelial-Mesenquimal/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de la radiación , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de la radiación , Humanos , Células MCF-7 , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/efectos de la radiación , Regulación hacia Arriba
3.
Biochem Biophys Res Commun ; 501(1): 73-79, 2018 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-29680657

RESUMEN

Excessive inflammation plays a detrimental role in endotoxemia. A recent study indicated that alarmins such as high mobility group box 1 (HMGB1) have drawn attention as therapeutic targets of sepsis. Post-translational modification (i.e., acetylation of lysine residues) of HMGB1 leads to the release of HMGB1 into the cellular space, operating as a warning signal that induces inflammation. Sirtuin 1 (SIRT1) has been shown to negatively regulate HMGB1 hyperacetylation and its extracellular release in sepsis. Therefore, we hypothesized that the S-nitrosylation (SNO) of SIRT1 may disrupt the ability of SIRT1 to negatively regulate the hyperacetylation of HMGB1. As long as the S-nitrosylation of SIRT1 occurs during septic conditions, it may worsen the situation. We found that the activity of SIRT1 decreased as the SNO-SIRT1 levels increased, resulting in HMGB1 release by LPS in RAW264.7 cells. Both the iNOS inhibitor (1400 W) and silencing iNOS significantly inhibited SNO-SIRT1, allowing increases in SIRT1 activity that decreased the HMGB1 release by LPS. SNAP, a NO donor, significantly increased both SNO-SIRT1 levels and the HMGB1 release that was accompanied by decreased sirt1 activity. However, sirtinol, a Sirt1 inhibitor, by itself decreased Sirt1 activity compared to that of the control, so that it did not affect already increased SNO-SIRT levels by SNAP. Most importantly, in lung tissues of LPS-endotoxic mice, significantly increased levels of SNO-SIRT were found, which was inhibited by 1400 W treatment. Plasma nitrite and HMGB1 levels were significantly higher than those in the sham controls, and the elevated levels were significantly lowered in the presence of 1400 W. We concluded that the S-nitrosylation of Sirt1 under endotoxic conditions may uninhibit the acetylation of HMGB1 and its extracellular release.


Asunto(s)
Endotoxemia/metabolismo , Proteína HMGB1/metabolismo , Sirtuina 2/metabolismo , Acetilación , Amidinas/farmacología , Animales , Bencilaminas/farmacología , Proteína HMGB1/sangre , Proteína HMGB1/química , Lipopolisacáridos/toxicidad , Masculino , Ratones , Ratones Endogámicos BALB C , Donantes de Óxido Nítrico/farmacología , Óxido Nítrico Sintasa de Tipo II/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo II/genética , Nitritos/sangre , Compuestos Nitrosos/química , Compuestos Nitrosos/metabolismo , Células RAW 264.7 , S-Nitroso-N-Acetilpenicilamina/farmacología , Sirtuina 2/química
4.
Exp Mol Med ; 50(4): 1-17, 2018 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-29674665

RESUMEN

Activation of the α7 nicotinic acetylcholine receptor (α7nAChR) has been shown to attenuate excessive inflammation by inhibiting proinflammatory cytokines during ischemia-reperfusion (IR) injury; however, the underlying kidney-specific molecular mechanisms remain unclear. The protective action of α7nAChR against renal IR injury was investigated using a selective α7nAChR agonist and antagonist. α7nAChR activation reduced plasma creatinine levels and tubular cell damage, whereas α7nAChR inhibition aggravated the IR-induced phenotype. α7nAChR activation decreased neutrophil infiltration and proinflammatory cytokine expression, increased heme oxygenase-1 (HO-1) expression, and reduced proximal tubular apoptosis after IR as shown by terminal deoxynucleotidyl transferase dUTP nick-end labeling staining and caspase-3 cleavage. In this study, we first showed that α7nAChR activation in the proximal tubules induced HO-1 expression through the phosphoinositide 3-kinase (PI3K)/Akt and protein kinase C (PKC) signaling pathway in vivo in renal IR mice and in vitro in proximal tubular cells. Chemical inhibitors of PKC or PI3K/Akt and small interfering RNA-mediated PKC silencing confirmed the signal specificity of α7nAChR-mediated HO-1 induction in the proximal tubular cells. α7nAChR activation inhibited high-mobility group box 1 release by inducing HO-1 expression and reduced proinflammatory cytokine gene expression and apoptotic cell death in tumor necrosis factor α-stimulated proximal tubular cells. Taken together, we conclude that α7nAChR activation in proximal tubular cells directly protects cells against renal IR injury by inducing HO-1 expression through PI3K/Akt and PKC signaling.


Asunto(s)
Lesión Renal Aguda , Regulación Enzimológica de la Expresión Génica , Hemo-Oxigenasa 1/biosíntesis , Isquemia , Túbulos Renales Proximales , Proteínas de la Membrana/biosíntesis , Proteína Quinasa C/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Receptor Nicotínico de Acetilcolina alfa 7/metabolismo , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/patología , Lesión Renal Aguda/prevención & control , Animales , Isquemia/metabolismo , Isquemia/patología , Isquemia/prevención & control , Túbulos Renales Proximales/irrigación sanguínea , Túbulos Renales Proximales/metabolismo , Túbulos Renales Proximales/patología , Masculino , Ratones
5.
Inflamm Res ; 67(5): 445-453, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29497773

RESUMEN

OBJECTIVE: Although luteolin has shown to have anti-inflammatory action, no report is available whether luteolin inhibits HMGB1 and protects acute lung injury (ALI) in endotoxin rodents. We hypothesized that HO-1 induction by luteolin might play a crucial role for inhibition of pro-inflammatory mediators including HMGB1 through MAPK signaling in LPS-induced RAW264.7 cells, and it ameliorates ALI of endotoxin mice. METHODS: The effects of luteolin on the production of pro-inflammatory mediators in LPS-activated RAW264.7 cells and LPS-injected mice were evaluated. The mechanisms were investigated using various signal inhibitors. RESULTS: Luteolin significantly increased HO-1 expression through ERK1/2 signaling in a time- and concentration-dependent manner. Indeed, luteolin inhibited pro-inflammatory mediators (HMGB1, iNOS/NO, COX-2, and NF-κB activity) in LPS-activated RAW264.7 cells. In addition, PD98059, an ERK1/2 inhibitor, treatment failed to inhibit production of these pro-inflammatory mediators by luteolin. Interestingly, luteolin augmented HO-1 induction through Ca2+ influx in RAW264.7 cells. Administration of luteolin significantly inhibited plasma HMGB1 level, and iNOS expression in the lung that resulted in a significant reduction of ALI in endotoxin mice that was reversed by a HO-1 inhibitor, ZnPPIX. CONCLUSION: Therefore, we conclude that luteolin has a great potential for treatment of ALI and related diseases, where HMGB1 is a therapeutic target.


Asunto(s)
Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/prevención & control , Ciclooxigenasa 2/efectos de los fármacos , Proteína HMGB1/antagonistas & inhibidores , Hemo-Oxigenasa 1/biosíntesis , Lipopolisacáridos/antagonistas & inhibidores , Luteolina/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo II/antagonistas & inhibidores , Animales , Señalización del Calcio/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Ciclooxigenasa 2/biosíntesis , Endotoxinas , Proteína HMGB1/biosíntesis , Lipopolisacáridos/farmacología , Masculino , Ratones , Ratones Endogámicos BALB C , Óxido Nítrico Sintasa de Tipo II/biosíntesis , Células RAW 264.7
6.
Arch Med Res ; 48(5): 423-431, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29089150

RESUMEN

BACKGROUND AND AIMS: High mobility group box 1 (HMGB1) plays an important role as a pro-inflammatory cytokine that regulates inflammation in various diseases. We hypothesized that hemin might reduce HMGB1 release through the induction of HO-1 in UVB-induced HaCaTs. METHODS: The effects of hemin on the release of HMGB1 in UVB exposure were evaluated. The mechanisms were investigated using various signal inhibitors and small interfering RNA techniques. RESULTS: Treatment with hemin inhibited reactive oxygen species (ROS) in UVB-induced HaCaTs in a dose-dependent manner. HMGB1 release by UVB was significantly reduced by hemin, N-acetyl-cysteine and DPI (NADPH oxidase inhibitor). Hemin increased HO-1 induction followed by phosphorylation of AMPK in a time- and dose-dependent manner. Additionally, hemin significantly increased the NAD+/NADH ratio in HaCaTs. The inhibitory effects of UVB-induced HMGB1 release by hemin were significantly reversed not only with pharmacological inhibitors of AMPK (compound c) or HO-1 (ZnPPIX) but also through transfection of small interfering RNAs (siRNAs) for AMPK or HO-1. Interestingly, hemin decreased phosphor-AMPK expression by HO-1 siRNA transfection, but it failed to induce HO-1 in AMPK siRNA-transfected cells, which suggested that HO-1 was involved in AMPK activation by hemin in HaCaT. Moreover, recombinant HMGB1 induced Snail and inhibited E-Cadherin in HaCaTs, whereas hemin reversed those effects through rHMGB1. CONCLUSIONS: It is concluded that the increased activity of HO-1/AMPK and scavenging ROS are, at least in part, responsible for the inhibition of UVB-induced HMGB1 release in keratinocyte HaCaTs. Therefore, hemin may be a useful agent for preventing UVB-induced skin cancer.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Proteína HMGB1/metabolismo , Hemo-Oxigenasa 1/biosíntesis , Hemina/farmacología , Queratinocitos/efectos de los fármacos , Queratinocitos/efectos de la radiación , Rayos Ultravioleta/efectos adversos , Antígenos CD , Cadherinas/metabolismo , Línea Celular , Citocinas/metabolismo , Inducción Enzimática , Humanos , Queratinocitos/metabolismo , Protoporfirinas/farmacología , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal
7.
Int Immunopharmacol ; 52: 297-304, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28982049

RESUMEN

Tetrahydroisoquinoline alkaloids (THIs) have shown to increase survival and beneficial effect on animal model of sepsis, partly due to heme oxygenase-1 (HO-1) induction. Here, we aimed to compare a limited series of synthesized THIs on HO-1 induction and inhibitory effect of iNOS and COX-2 expression in lipopolysaccharide (LPS)-activated RAW264.7 cells. To the end, most promising compound (THI-61) was tested whether this compound reduces iNOS protein expression and inflammatory markers (HMGB1, TNF-α) in LPS-treated mice lung tissue. The results indicated that N-carbonyl substituted THI seem to affect HO-1 induction depending on which functional group is attached to C1 position. All compounds that reduce LPS-activated NF-κB-luciferase activity showed to preferential inhibition of iNOS/NO but not COX-2/PGE2 that was partly related to inhibition of STAT-1 phosphorylation. In particular, THI-61 induced translocation of Nrf2 from cytosol into the nucleus by an increased Nrf2-ARE binding activity, and reduced IL-1ß production in LPS-activated RAW264.7 cells. The reduced expression of iNOS/NO by THI-61 was reversed by siHO-1RNA-transfection. In LPS-treated mice, THI-61 significantly reduced iNOS protein in lung tissues, and HMGB1 and TNF-α levels in the BALF. We concluded that 1) lipophilic moiety of 1C substituent is much more important in N-carbonyl substituted THI for induction of HO-1, 2) newly synthesized THI-61 may be beneficial for treatment of lung injury.


Asunto(s)
Lesión Pulmonar Aguda/tratamiento farmacológico , Alcaloides/farmacología , Hemo-Oxigenasa 1/metabolismo , Inflamación/tratamiento farmacológico , Pulmón/metabolismo , Macrófagos/inmunología , Proteínas de la Membrana/metabolismo , Tetrahidroisoquinolinas/farmacología , Alcaloides/síntesis química , Animales , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Modelos Animales de Enfermedad , Proteína HMGB1/metabolismo , Hemo-Oxigenasa 1/genética , Humanos , Lipopolisacáridos/inmunología , Pulmón/efectos de los fármacos , Pulmón/patología , Macrófagos/efectos de los fármacos , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Células RAW 264.7 , Factor de Transcripción STAT1/metabolismo , Tetrahidroisoquinolinas/síntesis química , Factor de Necrosis Tumoral alfa/metabolismo , Regulación hacia Arriba
8.
Biomed Pharmacother ; 86: 48-56, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27939519

RESUMEN

High mobility group box 1 (HMGB1), a highly conserved non-histone DNA-binding protein, plays an important role in the pathogenesis of sepsis. Previously, the authors reported 13-ethylberberine (13-EBR) has anti-inflammatory and antibacterial effects. However, the effect of 13-EBR on HMGB1 release was not investigated. In the present study, it was hypothesized 13-EBR might reduce HMGB1 release by activating AMPK under septic conditions. The results obtained showed 13-EBR significantly reduced HMGB1 release from LPS-activated RAW264.7 cells, and that this reduction was reversed by silencing p38, or AMPK, or by co-treating cells with p38 MAPKinase inhibitor. 13-EBR increased the phosphorylations of p38 and AMPK, and the phosphorylation of p38 by 13-EBR was inhibited by AMPK-siRNA, indicating AMPK acted upstream of p38. In the lung tissues of LPS-treated mice, 13-EBR administration significantly increased p-AMPK but reduced inducible nitric oxide synthase (iNOS) protein levels. Hematoxylin and eosin staining revealed 13-EBR significantly reduced LPS-induced lung and liver damage. In addition, 13-EBR inhibited NF-kB in LPS-activated RAW264.7 cells, and in LPS-treated mice, 13-EBR administration significantly increased survival. Furthermore, co-administration of 13-EBR plus LPS prevented LPS-induced aortic rings hypocontractile response to phenylephrine in vitro. Taken together, these results indicate 13-EBR might offer a means of treating sepsis through AMPK activation.


Asunto(s)
Berberina/uso terapéutico , Endotoxemia/metabolismo , Endotoxemia/prevención & control , Proteína HMGB1/antagonistas & inhibidores , Lipopolisacáridos/toxicidad , Proteínas Quinasas/metabolismo , Quinasas de la Proteína-Quinasa Activada por el AMP , Animales , Berberina/farmacología , Endotoxemia/inducido químicamente , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Proteína HMGB1/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Insuficiencia Multiorgánica/inducido químicamente , Insuficiencia Multiorgánica/metabolismo , Insuficiencia Multiorgánica/prevención & control , Células RAW 264.7 , Ratas , Ratas Sprague-Dawley
9.
Int Immunopharmacol ; 41: 98-105, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27865166

RESUMEN

High mobility group box 1 (HMGB1), a cytokine present in the late phase of sepsis, may be a potential target for the treatment of sepsis. For HMGB1 to be actively secreted from macrophages during infections, it must be post-translationally modified. Although ethyl pyruvate (EP), a simple aliphatic ester derived from pyruvic acid, has been shown to inhibit the release of HMGB1 in lipopolysaccharide (LPS)-treated RAW 264.7 cells, the underlying mechanism(s) are not yet clear. We investigated the hypothesis that the upregulation of SIRT1 by EP might promote the deacetylation of HMGB1, which reduces HMGB1 release in LPS-activated macrophages. Our results show that EP induced the expression of the SIRT1 protein in RAW264.7 cells and that it significantly inhibited the LPS-induced acetylation of HMGB1. Transfection with a SIRT1-overexpressing vector resulted in a significant decrease in the acetylation of HMGB1 in LPS-activated RAW264.7 cells relative to control cells. The genetic ablation or the pharmacological inhibition of SIRT1 by sirtinol increased LPS-induced HMGB1 acetylation. Moreover, EP inhibited the acetylation of HMGB1 in peritoneal macrophages treated with LPS. Interestingly, EP significantly reduced the LPS-induced phosphorylation of STAT1, which was significantly reversed by siSIRT1 transfection in RAW264.7 cells, indicating that SIRT1 negatively regulates the phosphorylation of STAT1. Overall, the results show that EP promotes the deacetylation of HMGB1 via the inhibition of STAT1 phosphorylation through the upregulation of SIRT1, which reduces HMGB1 release in LPS-activated RAW264.7 cells. In conclusion, EP might be useful in the treatment of diseases that target HMGB1, such as sepsis.


Asunto(s)
Proteína HMGB1/antagonistas & inhibidores , Macrófagos Peritoneales/efectos de los fármacos , Piruvatos/farmacología , Factor de Transcripción STAT1/antagonistas & inhibidores , Sirtuina 1/metabolismo , Acetilación/efectos de los fármacos , Animales , Células Cultivadas , Proteína HMGB1/metabolismo , Inflamación/metabolismo , Lipopolisacáridos , Macrófagos Peritoneales/metabolismo , Ratones , Ratones Endogámicos BALB C , Fosforilación/efectos de los fármacos , Células RAW 264.7 , Factor de Transcripción STAT1/metabolismo , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
10.
Int Immunopharmacol ; 40: 269-276, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27632705

RESUMEN

High mobility group box 1 (HMGB1), a late phase cytokine of sepsis, is viewed as a potential target for the treatment of sepsis. The authors considered that 13-methylberberine (13-MB) might reduce circulating HMGB1 levels and increase survival in a mouse model of sepsis by activating AMP-activated protein kinase (AMPK). Western blot analysis and vascular contraction testing were performed using RAW264.7 cells and rat thoracic aorta, respectively. The mechanisms responsible were investigated using various signal inhibitors and small interfering RNA techniques. 13-MB significantly reduced HMGB1 release by LPS-activated RAW264.7 cells, and this was prevented by silencing AMPK or p38, or by pretreating cells with p38 MAPKinase inhibitor, suggesting that the activations of p38 and AMPK were responsible for the observed reduction in HMGB1 release. As was expected, 13-MB increased the phosphorylations of p38 and AMPK. Interestingly, phosphorylations of p38 by 13-MB were inhibited by AMPKsiRNA, indicating that AMPK lies upstream of p38. Furthermore, 13-MB concentration-dependently inhibited IκB phosphorylation in LPS-activated RAW264.7 cells, and in aortic rings, co-treatment with 13-MB and LPS for 8h, in vitro, significantly restored the isometric contraction induced by phenylephrine. Importantly, 13-MB significantly increased the survival rate of LPS-induced endotoxemic mice. These results suggest 13-MB may be useful for treating diseases in which HMGB1 is viewed as a target.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Berberina , Proteína HMGB1/antagonistas & inhibidores , Sepsis/tratamiento farmacológico , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Aorta Torácica/efectos de los fármacos , Aorta Torácica/fisiología , Berberina/análogos & derivados , Berberina/farmacología , Berberina/uso terapéutico , Proteína HMGB1/metabolismo , Técnicas In Vitro , Lipopolisacáridos , Luciferasas/genética , Luciferasas/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , FN-kappa B/metabolismo , Células RAW 264.7 , ARN Interferente Pequeño/genética , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Vasoconstricción/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/genética
11.
Eur J Pharmacol ; 788: 200-209, 2016 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-27343380

RESUMEN

Obesity-associated non-alcoholic fatty liver disease (NAFLD) increases coagulation and inflammation. We hypothesized that (S)YS-51, an agent found to be beneficial in animal models of sepsis, may reduce NAFLD in high-fat diet (HFD) mice by reducing coagulation and inflammation. C57BL/6 mice were fed either a chow diet or HFD and each was supplemented with or without (S)YS-51 (10mg/kg, daily, i.p.) for 16 weeks. The results showed that HFD caused significant increases in lipogenesis [CD36, fatty acid synthase (FAS) and sterol response element binding protein (SREBP)-1c mRNA and protein], inflammation [monocyte chemotactic protein (MCP)-1, tumor necrosis factor (TNF)-α, intercellular cell adhesion molecule-1 (ICAM-1), TGF-ß, and procollagen type 1 mRNA, macrophage infiltration] and coagulation [tissue factor (TF) and plasminogen activator inhibitor-1 (PAI-1) mRNA and thrombin antithrombin complex (TAT)] in the liver, adipose tissue and serum, which were significantly reduced by (S)YS-51. These results of (S)YS-51 were accompanied by significant reduction of weight gain, liver size, hepatic steatosis and fibrosis, blood cholesterol, hepatic triglyceride, and macrophage infiltration and inflammatory cytokines in adipose tissue without affecting food intake in HFD mice. Interestingly, (S)YS-51 increased SIRT1 mRNA and protein and AMPK expression in the liver of HFD mice by increasing both NAD(+)/NADH ratio and LKB1 phosphorylation. In HepG2 cells, (S)YS-51 activated SIRT1 followed by AMPK. Finally, (S)YS-51 improved glucose tolerance and insulin resistance in HFD mice. We concluded that (S)YS-51 attenuates NAFLD and insulin resistance in HFD mice by, at least, activation of SIRT1/AMPK signals. Thus, (S)YS-51 may be beneficial in NAFLD treatment.


Asunto(s)
Coagulación Sanguínea/efectos de los fármacos , Lipogénesis/efectos de los fármacos , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Obesidad/complicaciones , Tetrahidroisoquinolinas/farmacología , Proteínas Quinasas Activadas por AMP/metabolismo , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/patología , Animales , Dieta Alta en Grasa/efectos adversos , Fibrosis , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Células Hep G2 , Humanos , Inflamación/complicaciones , Resistencia a la Insulina , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Hígado/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico/complicaciones , Enfermedad del Hígado Graso no Alcohólico/fisiopatología , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal/efectos de los fármacos , Sirtuina 1/metabolismo , Tetrahidroisoquinolinas/uso terapéutico
12.
Thromb Res ; 136(2): 456-64, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26116490

RESUMEN

INTRODUCTION: Inflammation and coagulation play important roles in the pathogenesis of sepsis. Anticoagulants with anti-inflammatory action draw attention as therapeutic agent in sepsis. OBJECTIVE: Whether cilostazol (6-[4-(1-cyclohexyl-1H-tetrazol-5-yl) butoxy]-3,4-dihydro-2-(1H)-quinolinone), anticoagulant, protects mice against sepsis and underlying mechanism(s) were investigated. METHODS: Induction of heme oxygenase (HO)-1 protein, phosphorylation of 5' adenosine monophosphate-activated protein kinase (AMPK), nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) luciferase activity, and release of high mobility group box 1 (HMGB1) were analyzed using signal inhibitors and transfection techniques. Survival and organ damage were compared in septic mice with and without cilostazol. RESULTS: In RAW264.7 cells, cilostazol increased phosphorylation of AMPK which was followed by HO-1 induction. Lipopolysaccharide (LPS)-activated HMGB1 release was reduced by cilostazol which was reversed by both SB203580 and silencing of HO-1 or AMPK RNA. Interestingly, silencing AMPK reduced HO-1 expression, whereas silencing HO-1 did not affect p-AMPK by cilostazol. Both compound C and zinc protoporphyrin IX (ZnPPIX) antagonized inhibitory effect of HMGB1 by cilostazol. Cilostazol inhibited NF-κB luciferase activity which was antagonized by SB203580. Finally, the administration of cilostazol increased the survival of endotoxemic mice but failed to do so when co-treated with rHMGB1. Cilostazol reduced circulating HMGB1, plasminogen activator inhibitor-1 (PAI-1) levels, organ damages and protein expression of PAI-1 in lung tissues of CLP-septic mice, which were antagonized by ZnPPIX. CONCLUSION: These findings suggest that HMGB1 can be a target molecule of cilostazol by 1) AMPK activation, and 2) induction of HO-1 by p38 MAPK and AMPK. Therefore, cilostazol may be useful for treatment of sepsis.


Asunto(s)
Proteína HMGB1/inmunología , Hemo-Oxigenasa 1/inmunología , Proteínas de la Membrana/inmunología , Sepsis/tratamiento farmacológico , Sepsis/inmunología , Serpina E2/inmunología , Tetrazoles/administración & dosificación , Animales , Anticoagulantes/administración & dosificación , Cilostazol , Relación Dosis-Respuesta a Droga , Lipopolisacáridos , Ratones , Células RAW 264.7 , Sepsis/inducido químicamente , Tasa de Supervivencia , Resultado del Tratamiento
13.
Atherosclerosis ; 241(2): 615-23, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26116962

RESUMEN

OBJECTIVE: Retinoic acid (RA) is the active vitamin A derivative and has diverse immunomodulatory actions. We hypothesized that RA reduces prothrombotic mediators such as tissue factor (TF) in endothelial cells during inflammatory conditions via an AMPK-dependent pathway, which attenuates cardiovascular complications. RESULTS: RA significantly increased AMPK and Akt phosphorylation in a time- and concentration-dependent manner in endothelial cells (EC). RA downregulated TF expression at the transcriptional and translational levels in TNF-α activated ECs, which was reversed by the silencing of AMPK and transfection of DN-AMPK. Interestingly, the PI3-kinase inhibitor LY294002 reversed the RA effect on TF expression. Increased AMPK phosphorylation by RA was inhibited by LY294002. However, increased Akt phosphorylation was not reduced by compound C, indicating that PI3K/Akt signaling modulates AMPK activity. In addition, RA reduced HMGB1 release in TNF-α activated ECs, which was reversed by both LY294001 and siAMPK. Importantly, administration of RA (1 mg/kg) significantly reduced blood TF activity, circulating HMGB1 and PAI-1 levels and expression of hepatic TF mRNA as well as fibrin deposition in LPS (5 mg/kg)-injected mice. CONCLUSIONS: Taken together, the activation of PI3K/Akt by RA modulates AMPK activity in ECs and plays a crucial role in the inhibition of coagulatory factors such as TF, PAI-1, and HMGB1 in inflammatory conditions.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Proteína HMGB1/antagonistas & inhibidores , Inhibidor 1 de Activador Plasminogénico/metabolismo , Serpina E2/metabolismo , Tromboplastina/antagonistas & inhibidores , Tretinoina/química , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Cromonas/química , Medios de Cultivo , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Fibrina/química , Células Endoteliales de la Vena Umbilical Humana , Humanos , Inflamación , Lipopolisacáridos/metabolismo , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Morfolinas/química , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación
14.
Oncol Rep ; 34(1): 195-202, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26063340

RESUMEN

Tumor metastasis is considered the main cause of mortality in cancer patients, thus it is important to investigate the differences between high- and low-metastatic cancer cells. Our previous study showed that the highly metastatic breast cancer cell line MDA-MB-231 released higher levels of ATP and exhibited higher P2Y2R activity compared with the low-metastatic breast cancer cell line MCF-7. In addition, P2Y2R activation by ATP released from MDA-MB-231 cells induced hypoxia-inducible factor-1α expression, lysyl oxidase secretion and collagen crosslinking, generating a receptive microenvironment for pre-metastatic niche formation. Thus, in the present study, we investigated which P2Y2R-related signaling pathways are involved in the invasion of breast cancer cells. The highly metastatic breast cancer cells MDA-MB-231 and SK-BR-3 showed higher invasion than MCF-7 and T47D cells at a basal level, which was abolished through P2Y2R knockdown or in the presence of apyrase, an enzyme that hydrolyzes extracellular nucleotides. MDA-MB-231 cells also showed high levels of mesenchymal markers, such as Snail, Vimentin and N-cadherin, but not the epithelial marker E-cadherin and this expression was inhibited through ATP degradation or P2Y2R knockdown. Moreover, SK-BR-3 and MDA-MB231 cells exhibited higher ERK and PKC phosphorylation levels than T47D and MCF-7 cells and upregulated phospho-ERK and -PKC levels in MDA-MB-231 cells were significantly downregulated by apyrase or P2Y2R knockdown. Specific inhibitors of ERK, PKC and PLC markedly reduced the invasion and levels of mesenchymal marker expression in MDA-MB-231 cells. These results suggest that over-activated ERK and PKC pathways are involved in the P2Y2R-mediated invasion of breast cancer cells.


Asunto(s)
Neoplasias de la Mama/genética , Proliferación Celular/genética , Proteína Quinasa C/biosíntesis , Receptores Purinérgicos P2Y2/biosíntesis , Neoplasias de la Mama/patología , Transición Epitelial-Mesenquimal/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Células MCF-7 , Metástasis de la Neoplasia , Proteína Quinasa C/genética , Proteína-Lisina 6-Oxidasa/biosíntesis , Receptores Purinérgicos P2Y2/genética , Transducción de Señal , Microambiente Tumoral/genética
15.
Vascul Pharmacol ; 72: 108-17, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25956731

RESUMEN

Vascular smooth muscle cells (VSMCs) are the major cell type in blood vessel walls, and their proliferation and migration play important roles in the development of atherosclerosis. Recently, it has been reported that IL-1ß mediates the inflammatory response through the upregulation of the P2Y2 receptor (P2Y2R). Thus, we examined the role of P2Y2R in IL-1ß-mediated proliferation and migration of VSMCs and the underlying molecular mechanisms. VSMCs were pretreated with IL-1ß for 24h to upregulate P2Y2R expression. The cells were then stimulated with UTP or ATP for the indicated times, and cell proliferation and migration and the related signaling pathways were examined. The equipotent P2Y2R agonists ATP and UTP enhanced proliferation, RAGE expression and HMGB1 secretion in IL-1ß-pretreated VSMCs. Additionally, pretreatment with IL-1ß enhanced UTP-mediated VSMC migration and MMP-2 release, but these effects were not observed in the P2Y2R-siRNA- or RAGE-siRNA-transfected VSMCs. Next, the signaling molecules involved in P2Y2R-mediated cell proliferation and migration were determined. The ERK, AKT, PKC, Rac-1 and ROCK2 pathways were involved in UTP-induced cell proliferation and migration, MMP-2 and HMGB1 secretion and RAGE expression in the IL-1ß-pretreated VSMCs. UTP induced the phosphorylation of ERK, AKT and PKC and the translocation of Rac-1 and ROCK2 from cytosol to membrane as well as stress fiber formation, which were markedly increased in the IL-1ß-pretreated VSMCs but not in the P2Y2R-siRNA-transfected VSMCs. These results demonstrate that pro-inflammatory cytokines associated with atherosclerosis, such as IL-1ß, can accelerate the process of atherosclerosis through the upregulation of P2Y2R.


Asunto(s)
Movimiento Celular/fisiología , Proliferación Celular/fisiología , Proteína HMGB1/metabolismo , Interleucina-1beta/metabolismo , Músculo Liso Vascular/metabolismo , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Receptores Purinérgicos P2Y2/metabolismo , Animales , Aterosclerosis/metabolismo , Aterosclerosis/patología , Células Cultivadas , Músculo Liso Vascular/fisiología , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/fisiología , Fosforilación/fisiología , Ratas , Transducción de Señal/fisiología , Regulación hacia Arriba/fisiología
16.
Biochem Pharmacol ; 95(4): 279-89, 2015 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-25896849

RESUMEN

High mobility group box 1 (HMGB1) is now recognized as a late mediator of sepsis. We tested hypothesis that ascorbic acid (AscA) induces heme oxygenase (HO)-1 which inhibits HMGB1 release in lipopolysaccharide (LPS)-stimulated cells and increases survival of septic mice. AscA increased HO-1 protein expression in a concentration- and time-dependent manner via Nrf2 activation in RAW 264.7 cells. HO-1 induction by AscA was significantly reduced by Nrf2 siRNA-transfected cells. Mutation of cysteine to serine of keap-1 proteins (C151S, C273S, and C288S) lost the ability of HO-1 induction by AscA, due to failure of translocation of Nrf-2 to nucleus. The PI3 kinase inhibitor, LY294002, inhibited HO-1 induction by AscA. Oxyhemoglobin (HbO2), LY294002, and ZnPPIX (HO-1 enzyme inhibitor) reversed effect of AscA on HMGB1 release. Most importantly, administration of AscA (200mg/kg, i.p.) significantly increased survival in LPS-induced endotoxemic mice. In cecal ligation and puncture (CLP)-induced septic mice, AscA reduced hepatic injury and serum HMGB1 and plasminogen activator inhibitor (PAI)-1 in a ZnPPIX-sensitive manner. In addition, AscA failed to increase survival in Nrf2 knockout mice by LPS. Thus, we concluded that high dose of AscA may be useful in the treatment of sepsis, at least, by activation of Nrf2/HO-1 signals.


Asunto(s)
Ácido Ascórbico/farmacología , Proteína HMGB1/metabolismo , Hemo-Oxigenasa 1/metabolismo , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Proteínas de la Membrana/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Sepsis/tratamiento farmacológico , Transporte Activo de Núcleo Celular , Animales , Ácido Ascórbico/uso terapéutico , Monóxido de Carbono/metabolismo , Línea Celular , Núcleo Celular/metabolismo , Chlorocebus aethiops , Activación Enzimática , Hígado/efectos de los fármacos , Hígado/patología , Activación de Macrófagos , Macrófagos/metabolismo , Masculino , Ratones Endogámicos ICR , FN-kappa B/metabolismo , Compuestos Organometálicos/metabolismo , Compuestos Organometálicos/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Sepsis/mortalidad , Sepsis/fisiopatología
17.
Int Immunopharmacol ; 26(1): 112-8, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25812767

RESUMEN

High mobility group box 1 (HMGB1) is now recognized as a late mediator of sepsis. Although glycyrrhizin was known as inhibitor of HMGB1, it is not yet clear underlying mechanism(s). We found that glycyrrhizin activates translocation of Nrf2 from cytosol to nucleus and induces heme oxygenase (HO)-1 expression in RAW 264.7 cells. In addition, deletion of Nrf2 by siRNA significantly reduced mRNA expression of NQO1 and HO-1 suggesting that glycyrrhizin targets Nrf2 gene. The expression of iNOS protein and release of HMGB1 in LPS activated cells were significantly reduced by glycyrrhizin and cells transfected with mouse HO-1 expression vector. The p38MAPK inhibitor (SB203580) but not JNK inhibitor (SP600125) or ERK inhibitor (PD98059) significantly inhibited HO-1 induction by glycyrrhizin, which was confirmed by showing that siP38 transfected cells significantly reduced HO-1 induction. Pretreatment with SB203580 significantly reversed the expression of iNOS and release of NO and HMGB1 in LPS-activated cells. Most importantly, administration of glycyrrhizin (200mg/kg, i.p) significantly reduced hepatic injury and serum HMGB1 in a ZnPPIX-sensitive manner. Thus, it is concluded that glycyrrhizin reduces HMGB1 secretion in lipopolysaccharide-activated RAW 264.7 cells and endotoxemic mice by p38/Nrf2-dependent induction of HO-1.


Asunto(s)
Endotoxemia/tratamiento farmacológico , Ácido Glicirrínico/farmacología , Proteína HMGB1/metabolismo , Hemo-Oxigenasa 1/biosíntesis , Activación de Macrófagos/efectos de los fármacos , Macrófagos/efectos de los fármacos , Proteínas de la Membrana/biosíntesis , Factor 2 Relacionado con NF-E2/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Western Blotting , Técnicas de Cultivo de Célula , Línea Celular , Supervivencia Celular/efectos de los fármacos , Endotoxemia/inmunología , Endotoxemia/metabolismo , Ácido Glicirrínico/administración & dosificación , Ácido Glicirrínico/uso terapéutico , Ácido Glicirrínico/toxicidad , Proteína HMGB1/antagonistas & inhibidores , Hemo-Oxigenasa 1/genética , Lipopolisacáridos/farmacología , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos ICR , Factor 2 Relacionado con NF-E2/genética , ARN Interferente Pequeño/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Transfección , Proteínas Quinasas p38 Activadas por Mitógenos/genética
18.
Toxicol Appl Pharmacol ; 284(2): 113-24, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25737164

RESUMEN

Honokiol is a bioactive neolignan compound isolated from the species of Magnolia. This study was designed to elucidate the cellular mechanism by which honokiol alleviates the development of non-alcoholic steatosis. HepG2 cells were treated with honokiol for 1h, and then exposed to 1mM free fatty acid (FFA) for 24h to simulate non-alcoholic steatosis in vitro. C57BL/6 mice were fed with a high-fat diet for 28days, and honokiol (10mg/kg/day) was daily treated. Honokiol concentration-dependently attenuated intracellular fat overloading and triglyceride (TG) accumulation in FFA-exposed HepG2 cells. These effects were blocked by pretreatment with an AMP-activated protein kinase (AMPK) inhibitor. Honokiol significantly inhibited sterol regulatory element-binding protein-1c (SREBP-1c) maturation and the induction of lipogenic proteins, stearoyl-CoA desaturase-1 (SCD-1) and fatty acid synthase (FAS) in FFA-exposed HepG2 cells, but these effects were blocked by pretreatment of an AMPK inhibitor. Honokiol induced AMPK phosphorylation and subsequent acetyl-CoA carboxylase (ACC) phosphorylation, which were inhibited by genetic deletion of liver kinase B1 (LKB1). Honokiol stimulated LKB1 phosphorylation, and genetic deletion of LKB1 blocked the effect of honokiol on SREBP-1c maturation and the induction of SCD-1 and FAS proteins in FFA-exposed HepG2 cells. Honokiol attenuated the increases in hepatic TG and lipogenic protein levels and fat accumulation in the mice fed with high-fat diet, while significantly induced LKB1 and AMPK phosphorylation. Taken together, our findings suggest that honokiol has an anti-lipogenic effect in hepatocytes, and this effect may be mediated by the LKB1-AMPK signaling pathway, which induces ACC phosphorylation and inhibits SREBP-1c maturation in hepatocytes.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Compuestos de Bifenilo/farmacología , Hepatocitos/efectos de los fármacos , Lignanos/farmacología , Metabolismo de los Lípidos/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/metabolismo , Acetil-CoA Carboxilasa/metabolismo , Animales , Línea Celular Tumoral , Dieta Alta en Grasa/métodos , Ácido Graso Sintasas/metabolismo , Ácidos Grasos no Esterificados/metabolismo , Células Hep G2 , Hepatocitos/metabolismo , Humanos , Lipogénesis/efectos de los fármacos , Hígado/citología , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Fosforilación/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Estearoil-CoA Desaturasa/metabolismo , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Triglicéridos/metabolismo
19.
Oncol Rep ; 33(3): 1141-6, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25482101

RESUMEN

CKD-712, a newly synthesized tetrahydroisoquinoline (THI) and an enantiomer (S form) of YS 49 (a derivative of higenamine) has been reported to suppress nuclear factor-κB (NF-κB) activity in normal cells. In the present study, we investigated the anticancer effects of THI at a low concentration where CKD-712 did not induce cell death in normal cells. At the range of concentrations used, CKD-712 induced cell growth arrest, and inhibited the invasion and motility of A549 cells as determined by cell cycle analysis, a Matrigel-coated chamber assay, and a wound-healing assay, respectively. CKD-712 suppressed MMP-9, but not MMP-2 and other NF-κB-regulated proteins involved in cancer metastasis such as VEGF. Moreover, CKD-712 induced cell cycle arrest at G2M phase by suppressing cyclin A, cyclin B and CDK-1 expression. Taken together, these data suggested that CKD-712 may exert anticancer effects by suppressing NF-κB pathways and inducing cell cycle arrest at G2M phase.


Asunto(s)
Antineoplásicos/farmacología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Neoplasias Pulmonares/metabolismo , FN-kappa B/efectos de los fármacos , Tetrahidroisoquinolinas/farmacología , Western Blotting , Línea Celular Tumoral , Citometría de Flujo , Humanos , FN-kappa B/metabolismo
20.
Oncotarget ; 5(19): 9322-34, 2014 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-25238333

RESUMEN

Tumor microenvironmental hypoxia induces hypoxia inducible factor-1α (HIF-1α) overexpression, leading to the release of lysyl oxidase (LOX), which crosslinks collagen at distant sites to facilitate environmental changes that allow cancer cells to easily metastasize. Our previous study showed that activation of the P2Y2 receptor (P2Y2R) by ATP released from MDA-MB-231 cells increased MDA-MB-231 cell invasion through endothelial cells. Therefore, in this study, we investigated the role of P2Y2R in breast cancer cell metastasis to distant sites. ATP or UTP released from hypoxia-treated MDA-MB-231 cells induced HIF-1α expression and LOX secretion by the activation of P2Y2R, and this phenomenon was significantly reduced in P2Y2R-depleted MDA-MB-231 cells. Furthermore, P2Y2R-mediated LOX release induced collagen crosslinking in an in vitro model. Finally, nude mice injected with MDA-MB-231 cells showed high levels of LOX secretion, crosslinked collagen and CD11b+ BMDC recruitment in the lung; however, mice that were injected with P2Y2R-depleted MDA-MB-231 cells did not exhibit these changes. These results demonstrate that P2Y2R plays an important role in activation of the HIF-1α-LOX axis, the induction of collagen crosslinking and the recruitment of CD11b+ BMDCs. Furthermore, P2Y2R activation by nucleotides recruits THP-1 monocytes, resulting in primary tumor progression and pre-metastatic niche formation.


Asunto(s)
Monocitos/inmunología , Metástasis de la Neoplasia/patología , Nucleótidos/metabolismo , Proteína-Lisina 6-Oxidasa/metabolismo , Receptores Purinérgicos P2Y2/metabolismo , Animales , Neoplasias de la Mama , Antígeno CD11b/metabolismo , Hipoxia de la Célula , Línea Celular Tumoral , Movimiento Celular , Colágeno/metabolismo , Progresión de la Enfermedad , Células Endoteliales/patología , Activación Enzimática , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/biosíntesis , Células MCF-7 , Macrófagos/inmunología , Ratones , Ratones Desnudos , Invasividad Neoplásica/patología , Trasplante de Neoplasias , Proteína-Lisina 6-Oxidasa/antagonistas & inhibidores , Interferencia de ARN , ARN Interferente Pequeño , Receptores Purinérgicos P2Y2/genética , Serina Endopeptidasas/genética , Trasplante Heterólogo , Microambiente Tumoral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...