Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Intervalo de año de publicación
1.
PLoS One ; 16(3): e0248298, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33705488

RESUMEN

BACKGROUND: The combination of JAK/STAT and HDAC inhibitors exerted beneficial effects in haematological malignancies, presenting promising therapeutic CTCL targets. We aim to investigate the efficacy of JAK1/2i ruxolitinib in combination with HDACi resminostat in CTCL in vitro. MATERIAL & METHODS: Non-toxic concentrations of ruxolitinib and/or resminostat were administered to MyLa (MF) and SeAx (SS) cells for 24h. Cytotoxicity, cell proliferation and apoptosis were estimated through MTT, BrdU/7AAD and Annexin V/PI assay. Multi-pathway analysis was performed to investigate the effect of JAK1/2i and/or HDACi on JAK/STAT, Akt/mTOR and MAPK signalling pathways. RESULTS: Both drugs and their combination were cytotoxic in MyLa (p<0.05) and in SeAx cell line (p<0.001), inhibited proliferation of MyLa (p<0.001) and SeAx (p<0.001) at 24h, compared to untreated cells. Moreover, combined drug treatment induced apoptosis after 24h (p<0.001) in MyLa, and SeAx (p<0.001). The combination of drugs had a strong synergistic effect with a CI<1. Importantly, the drugs' combination inhibited phosphorylation of STAT3 (p<0.001), Akt (p<0.05), ERK1/2 (p<0.001) and JNK (p<0.001) in MyLa, while it reduced activation of Akt (p<0.05) and JNK (p<0.001) in SeAx. CONCLUSION: The JAKi/HDACi combination exhibited substantial anti-tumor effects in CTCL cell lines, and may represent a promising novel therapeutic modality for CTCL patients.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Linfoma Cutáneo de Células T/tratamiento farmacológico , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteínas de Neoplasias/metabolismo , Línea Celular Tumoral , Sinergismo Farmacológico , Humanos , Ácidos Hidroxámicos/agonistas , Ácidos Hidroxámicos/farmacología , Linfoma Cutáneo de Células T/metabolismo , Linfoma Cutáneo de Células T/patología , Nitrilos , Pirazoles/agonistas , Pirazoles/farmacología , Pirimidinas , Sulfonamidas/agonistas , Sulfonamidas/farmacología
2.
Mol Cell Biochem ; 473(1-2): 247-262, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32661772

RESUMEN

Reverse cholesterol transport (RCT) and transintestinal cholesterol efflux (TICE) are two important pathways for body cholesterol elimination. We studied these pathways in an animal model of diabetes and obesity (ob/ob) where HDL function is compromised as a result of hyperglycemia, low-grade inflammation and oxidative stress. Co-treatment of ob/ob mice with PPAR-α (fenofibrate) and LXR (T0901317) agonists increased fecal cholesterol by 12-fold; PPAR-α and LXR agonists individually showed 2.6- and 4.0-fold fecal cholesterol excretion, respectively. We investigated the mechanism of synergistic efficacy of PPAR-α and LXR agonists in fecal cholesterol excretion. LXR agonist and the combination of PPAR-α and LXR agonists had greater HDL-C elevation. Ex vivo cholesterol efflux showed correlation with the fecal cholesterol excretion but was not sufficient to explain 12-fold increases in the fecal cholesterol in the co-treated mice. Therefore, we examined TICE to explain the 12-fold increases in the fecal cholesterol. A strong positive correlation of fecal cholesterol with ATP binding cassette transporter G5 (ABCG5) and G8 and a negative correlation with NPC1L1 was observed. ABCG5, G8 and NPC1L1 are involved in intestinal cholesterol absorption. The extent of influence of PPAR-α and LXR agonists on RCT and TICE was distinctly different. PPAR-α agonist increased fecal cholesterol primarily by influencing TICE, while LXR agonist influenced fecal cholesterol excretion via both RCT and TICE mechanisms. Synergistic efficacy on fecal cholesterol excretion following co-treatment with PPAR-α and LXR agonists occurred through a combination of RCT, TICE, and the key enzyme in bile synthesis, cholesterol 7-α hydroxylase (cyp7a1). These results suggest that cholesterol efflux, biliary cholesterol excretion, and TICE collectively contributed to the 12-fold increases in the fecal cholesterol excretion in ob/ob mice co-treated with PPAR-α and LXR agonists.


Asunto(s)
Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 5/metabolismo , Transportador de Casete de Unión a ATP, Subfamilia G, Miembro 8/metabolismo , Colesterol/metabolismo , Heces , Fenofibrato/farmacología , Hidrocarburos Fluorados/farmacología , Lipoproteínas/metabolismo , Receptores X del Hígado , Proteínas de Transporte de Membrana/metabolismo , PPAR alfa , Sulfonamidas/farmacología , Animales , Sinergismo Farmacológico , Fenofibrato/agonistas , Hidrocarburos Fluorados/agonistas , Receptores X del Hígado/agonistas , Receptores X del Hígado/metabolismo , Masculino , Ratones , Ratones Obesos , PPAR alfa/agonistas , PPAR alfa/metabolismo , Sulfonamidas/agonistas
5.
Plant Mol Biol ; 100(3): 319-333, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30941543

RESUMEN

KEY MESSAGE: We determined the structure of OsPYL/RCAR3:OsPP2C50 complex with pyrabactin. Our results suggest that a less-conserved phenylalanine of OsPYL/RCAR subfamily I is one of considerations of ABA agonist development for Oryza sativa. Pyrabactin is a synthetic chemical mimicking abscisic acid (ABA), a naturally occurring phytohormone orchestrating abiotic stress responses. ABA and pyrabactin share the same pocket in the ABA receptors but pyrabactin modulates ABA signaling differently, exhibiting both agonistic and antagonistic effects. To explore structural determinants of differential functionality of pyrabactin, we determined the crystal structure of OsPYL/RCAR3:pyrabactin:OsPP2C50, the first rice ABA receptor:co-receptor complex structure with a synthetic ABA mimicry. The water-mediated interaction between the wedging Trp-259 of OsPP2C50 and pyrabactin is lost, undermining the structural integrity of the ABA receptor:co-receptor. The loss of the interaction of the wedging tryptophan of OsPP2C with pyrabactin appears to contribute to the weaker functionality of pyrabactin. Pyrabactin in the OsPYL/RCAR3:OsPP2C50 complex adopts a conformation different from that in ABA receptors from Arabidopsis. Phe125, specific to the subfamily I of OsPYL/RCARs in the ABA binding pocket, appears to be the culprit for the differential conformation of pyrabactin. Although the gate closure essential for the integrity of ABA receptor:co-receptor is preserved in the presence of pyrabactin, Phe125 apparently restricts accessibility of pyrabactin, leading to decreased affinity for OsPYL/RCAR3 evidenced by phosphatase assay. However, Phe125 does not affect conformation and accessibility of ABA. Yeast two-hybrid, germination and gene transcription analyses in rice also support that pyrabactin imposes a weak effect on the control of ABA signaling. Taken together, our results suggest that phenylalanine substitution of OsPYL/RCARs subfamily I may be one of considerations for ABA synthetic agonist development.


Asunto(s)
Ácido Abscísico/metabolismo , Naftalenos/agonistas , Naftalenos/química , Oryza/metabolismo , Proteínas de Plantas/metabolismo , Sulfonamidas/agonistas , Sulfonamidas/química , Arabidopsis , Proteínas de Arabidopsis/química , Proteínas de Arabidopsis/metabolismo , Cristalografía por Rayos X , Germinación , Modelos Moleculares , Fosfoproteínas Fosfatasas/química , Reguladores del Crecimiento de las Plantas/metabolismo , Conformación Proteica , Semillas/metabolismo , Transducción de Señal , Estrés Fisiológico
6.
Med Chem ; 15(6): 676-684, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30799793

RESUMEN

BACKGROUND: Despite a massive industry endeavor to develop RORγ-modulators for autoimmune disorders, there has been no indication of efforts to target the close family member RORα for similar indications. This may be due to the misconception that RORα is redundant to RORγ, or the inherent difficulty in cultivating tractable starting points for RORα. RORα-selective modulators would be useful tools to interrogate the biology of this understudied orphan nuclear receptor. OBJECTIVE: The goal of this research effort was to identify and optimize synthetic ligands for RORα starting from the known LXR agonist T0901317. METHODS: Fourty-five analogs of the sulfonamide lead (1) were synthesized and evaluated for their ability to suppress the transcriptional activity of RORα, RORγ, and LXRα in cell-based assays. Analogs were characterized by 1H-NMR, 13C-NMR, and LC-MS analysis. The pharmacokinetic profile of the most selective RORα inverse agonist was evaluated in rats with intraperitoneal (i.p.) and per oral (p.o.)dosing. RESULTS: Structure-activity relationship studies led to potent dual RORα/RORγ inverse agonists as well as RORα-selective inverse agonists (20, 28). LXR activity could be reduced by removing the sulfonamide nitrogen substituent. Attempts to improve the potency of these selective leads by varying substitution patterns throughout the molecule proved challenging. CONCLUSION: The synthetic RORα-selective inverse agonists identified (20, 28) can be utilized as chemical tools to probe the function of RORα in vitro and in vivo.


Asunto(s)
Agonismo Inverso de Drogas , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares/agonistas , Sulfonamidas/farmacología , Animales , Humanos , Hidrocarburos Fluorados/química , Ligandos , Receptores X del Hígado/agonistas , Ratones , Estructura Molecular , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/agonistas , Ratas , Relación Estructura-Actividad , Sulfonamidas/agonistas , Sulfonamidas/síntesis química , Sulfonamidas/química , Sulfonamidas/farmacocinética , Células Th17
7.
Braz. J. Pharm. Sci. (Online) ; 55: e17032, 2019. tab, graf
Artículo en Inglés | LILACS | ID: biblio-1019533

RESUMEN

The aim of the present research work was to investigate the enzyme inhibitory potential of some new sulfonamides having benzodioxane and acetamide moieties. The synthesis was started by the reaction of N-2,3-dihydrobenzo[1,4]-dioxin-6-amine (1) with 4-methylbenzenesulfonyl chloride (2) in the presence of 10% aqueous Na2CO3 to yield N-(2,3-dihydrobenzo[1,4]-dioxin-6-yl)-4-methylbenzenesulfonamide (3), which was then reacted with 2-bromo-N-(un/substituted-phenyl)acetamides (6a-l) in DMF and lithium hydride as a base to afford various 2-{2,3-dihydro-1,4-benzodioxin-6-yl[(4-methylphenyl)sulfonyl]amino}-N-(un/substituted-phenyl)acetamides (7a-l). All the synthesized compounds were characterized by their IR and 1H-NMR spectral data along with CHN analysis data. The enzyme inhibitory activities of these compounds were tested against a-glucosidase and acetylcholinesterase (AChE). Most of the compounds exhibited substantial inhibitory activity against yeast a-glucosidase and weak against AChE. The in silico molecular docking results were also consistent with in vitro enzyme inhibition data.


Asunto(s)
Sulfonamidas/agonistas , Inhibidores de la Colinesterasa , Inhibidores de Glicósido Hidrolasas , Análisis Espectral/instrumentación , Acetamidas/análisis
8.
Haematologica ; 102(9): 1587-1593, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28619845

RESUMEN

Fluorizoline is a new synthetic molecule that induces apoptosis by selectively targeting prohibitins. In the study herein, the pro-apoptotic effect of fluorizoline was assessed in 34 primary samples from patients with chronic lymphocytic leukemia. Fluorizoline induced apoptosis in chronic lymphocytic leukemia cells at concentrations in the low micromolar range. All primary samples were sensitive to fluorizoline irrespective of patients' clinical or genetic features, whereas normal T lymphocytes were less sensitive. Fluorizoline increased the protein levels of the pro-apoptotic B-cell lymphoma 2 family member NOXA in chronic lymphocytic leukemia cells. Furthermore, fluorizoline synergized with ibrutinib, 5-aminoimidazole-4-carboxamide riboside or venetoclax to induce apoptosis. These results suggest that targeting prohibitins could be a new therapeutic strategy for chronic lymphocytic leukemia.


Asunto(s)
Aminoimidazol Carboxamida/análogos & derivados , Apoptosis/efectos de los fármacos , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Hidrocarburos Fluorados/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Pirazoles/farmacología , Pirimidinas/farmacología , Proteínas Represoras/metabolismo , Ribonucleósidos/farmacología , Sulfonamidas/farmacología , Tiazolidinas/farmacología , Regulación hacia Arriba/efectos de los fármacos , Adenina/análogos & derivados , Aminoimidazol Carboxamida/agonistas , Aminoimidazol Carboxamida/farmacología , Compuestos Bicíclicos Heterocíclicos con Puentes/agonistas , Sinergismo Farmacológico , Femenino , Humanos , Hidrocarburos Fluorados/agonistas , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Leucemia Linfocítica Crónica de Células B/metabolismo , Leucemia Linfocítica Crónica de Células B/patología , Masculino , Piperidinas , Prohibitinas , Pirazoles/agonistas , Pirimidinas/agonistas , Ribonucleósidos/agonistas , Sulfonamidas/agonistas , Tiazolidinas/agonistas , Células Tumorales Cultivadas
9.
Leukemia ; 31(10): 2075-2084, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28111464

RESUMEN

Although the BTK inhibitor ibrutinib has transformed the management of patients with chronic lymphocytic leukemia (CLL), it does not induce substantial apoptosis in vitro, and as such the mechanisms underlying its ability to kill CLL cells are not well understood. Acalabrutinib, a more specific BTK inhibitor now in development, also appears to be highly effective in CLL, but the connection of its mechanism with CLL cell death is also unclear. Using dynamic BH3 profiling, we analyzed alterations in the function of the mitochondrial apoptotic pathway induced by ibrutinib and acalabrutinib. We studied CLL patient samples treated ex vivo with both drugs, as well as primary samples from CLL patients on clinical trials of both drugs. We found that BTK inhibition enhances mitochondrial BCL-2 dependence without significantly altering overall mitochondrial priming. Enhancement of BCL-2 dependence was accompanied by an increase in the pro-apoptotic protein BIM. In contrast, treatment with the selective BCL-2 inhibitor venetoclax enhanced overall mitochondrial priming without increasing BCL-2 dependence. Pre-treatment of CLL cells with either BTK inhibitor, whether ex vivo or in vivo in patients, enhanced killing by venetoclax. Our data suggest that BTK inhibition enhances mitochondrial BCL-2 dependence, supporting the ongoing development of clinical trials combining BTK and BCL-2 inhibition.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Benzamidas/farmacología , Compuestos Bicíclicos Heterocíclicos con Puentes/agonistas , Resistencia a Antineoplásicos/efectos de los fármacos , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Terapia Molecular Dirigida , Proteínas de Neoplasias/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-bcl-2/fisiología , Pirazinas/farmacología , Pirazoles/farmacología , Pirimidinas/farmacología , Sulfonamidas/agonistas , Adenina/análogos & derivados , Agammaglobulinemia Tirosina Quinasa , Apoptosis/efectos de los fármacos , Proteína 11 Similar a Bcl2/biosíntesis , Proteína 11 Similar a Bcl2/genética , Benzamidas/administración & dosificación , Benzamidas/uso terapéutico , Compuestos Bicíclicos Heterocíclicos con Puentes/administración & dosificación , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Compuestos Bicíclicos Heterocíclicos con Puentes/uso terapéutico , Ensayos Clínicos como Asunto , Ensayos de Selección de Medicamentos Antitumorales , Sinergismo Farmacológico , Humanos , Leucemia Linfocítica Crónica de Células B/patología , Mitocondrias/efectos de los fármacos , Proteínas de Neoplasias/fisiología , Fragmentos de Péptidos , Piperidinas , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Proto-Oncogénicas , Pirazinas/administración & dosificación , Pirazinas/uso terapéutico , Pirazoles/administración & dosificación , Pirazoles/uso terapéutico , Pirimidinas/administración & dosificación , Pirimidinas/uso terapéutico , Sulfonamidas/administración & dosificación , Sulfonamidas/farmacología , Sulfonamidas/uso terapéutico
10.
J Biol Chem ; 291(51): 26502-26514, 2016 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-27793992

RESUMEN

Macrophage migration inhibitory factor (MIF) is a pleiotropic cytokine that has been implicated in a broad range of inflammatory and oncologic diseases. MIF is unique among cytokines in terms of its release profile and inflammatory role, notably as an endogenous counter-regulator of the anti-inflammatory effects of glucocorticoids. In addition, it exhibits a catalytic tautomerase activity amenable to the design of high affinity small molecule inhibitors. Although several classes of these compounds have been identified, biologic characterization of these molecules remains a topic of active investigation. In this study, we used in vitro LPS-driven assays to characterize representative molecules from several classes of MIF inhibitors. We determined that MIF inhibitors exhibit distinct profiles of anti-inflammatory activity, especially with regard to TNFα. We further investigated a molecule with relatively low anti-inflammatory activity, compound T-614 (also known as the anti-rheumatic drug iguratimod), and found that, in addition to exhibiting selective MIF inhibition in vitro and in vivo, iguratimod also has additive effects with glucocorticoids. Furthermore, we found that iguratimod synergizes with glucocorticoids in attenuating experimental autoimmune encephalitis, a model of multiple sclerosis. Our work identifies iguratimod as a valuable new candidate for drug repurposing to MIF-relevant diseases, including multiple sclerosis.


Asunto(s)
Cromonas/farmacología , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Glucocorticoides/farmacología , Oxidorreductasas Intramoleculares/antagonistas & inhibidores , Factores Inhibidores de la Migración de Macrófagos/antagonistas & inhibidores , Esclerosis Múltiple/tratamiento farmacológico , Sulfonamidas/farmacología , Animales , Línea Celular Tumoral , Cromonas/agonistas , Encefalomielitis Autoinmune Experimental/inducido químicamente , Encefalomielitis Autoinmune Experimental/metabolismo , Glucocorticoides/agonistas , Humanos , Oxidorreductasas Intramoleculares/metabolismo , Lipopolisacáridos/toxicidad , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Esclerosis Múltiple/metabolismo , Esclerosis Múltiple/patología , Sulfonamidas/agonistas , Factor de Necrosis Tumoral alfa/metabolismo
11.
PLoS One ; 10(3): e0120531, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25781619

RESUMEN

Positron emission tomography (PET) with 18F-fluorodeoxyglucose (FDG) is frequently used for visualizing gastrointestinal stromal tumors (GIST), which are highly glucose-avid tumors. Dramatic metabolic responses following imatinib treatment indicate a high, KIT-dependent glucose turnover which has been particularly helpful for predicting tumor response to imatinib. The glucose analogue 2-deoxyglucose (2DG) inhibits glucose metabolism in cancer cells that depend on aerobic glycolysis for ATP production. We show that 2DG inhibits proliferation in both imatinib-sensitive and imatinib-resistant GIST cell lines at levels that can be achieved clinically. KIT-negative GIST48B have 3-14-fold higher IC50 levels than KIT-positive GIST cells indicating that oncogenic KIT may sensitize cells to 2DG. GIST sensitivity to 2DG is increased in low-glucose media (110 mg/dl). 2DG leads to dose- and glucose dependent inhibition of KIT glycosylation with resultant reduction of membrane-bound KIT, inhibition of KIT-phosphorylation and inactivation of KIT-dependent signaling intermediates. In contrast to imatinib, 2DG caused ER-stress and elicited the unfolded protein response (UPR). Mannose but not pyruvate rescued GIST cells from 2DG-induced growth arrest, suggesting that loss of KIT integrity is the predominant effect of 2DG in GIST. Additive anti-tumoral effects were seen with imatinib and BH3-mimetics. Our data provide the first evidence that modulation of the glucose-metabolism by 2DG may have a disease-specific effect and may be therapeutically useful in GIST.


Asunto(s)
Compuestos de Anilina/farmacología , Apoptosis/efectos de los fármacos , Desoxiglucosa/farmacología , Tumores del Estroma Gastrointestinal/metabolismo , Proteínas Proto-Oncogénicas c-kit/metabolismo , Transducción de Señal/efectos de los fármacos , Sulfonamidas/farmacología , Compuestos de Anilina/agonistas , Animales , Apoptosis/genética , Línea Celular Tumoral , Desoxiglucosa/agonistas , Sinergismo Farmacológico , Tumores del Estroma Gastrointestinal/genética , Tumores del Estroma Gastrointestinal/patología , Glicosilación/efectos de los fármacos , Humanos , Ratones , Ratones Desnudos , Proteínas Proto-Oncogénicas c-kit/genética , Transducción de Señal/genética , Sulfonamidas/agonistas
12.
PLoS One ; 8(12): e81763, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24339963

RESUMEN

Combined targeting of the MAPK and PI3K signalling pathways in cancer may be necessary for optimal therapeutic activity. To support clinical studies of combination therapy, 3'-deoxy-3'-[(18)F]-fluorothymidine ([(18)F]-FLT) uptake measured by Positron Emission Tomography (PET) was evaluated as a non-invasive surrogate response biomarker in pre-clinical models. The in vivo anti-tumour efficacy and PK-PD properties of the MEK inhibitor PD 0325901 and the PI3K inhibitor GDC-0941, alone and in combination, were evaluated in HCT116 and HT29 human colorectal cancer xenograft tumour-bearing mice, and [(18)F]-FLT PET investigated in mice bearing HCT116 xenografts. Dual targeting of PI3K and MEK induced marked tumour growth inhibition in vivo, and enhanced anti-tumour activity was predicted by [(18)F]-FLT PET scanning after 2 days of treatment. Pharmacodynamic analyses using the combination of the PI3K inhibitor GDC-0941 and the MEK inhibitor PD 0325901 revealed that increased efficacy is associated with an enhanced inhibition of the phosphorylation of ERK1/2, S6 and 4EBP1, compared to that observed with either single agent, and maintained inhibition of AKT phosphorylation. Pharmacokinetic studies indicated that there was no marked PK interaction between the two drugs. Together these results indicate that the combination of PI3K and MEK inhibitors can result in significant efficacy, and demonstrate for the first time that [(18)F]-FLT PET can be correlated to the improved efficacy of combined PI3K and MEK inhibitor treatment.


Asunto(s)
Antivirales/farmacología , Benzamidas/farmacología , Neoplasias Colorrectales , Didesoxinucleósidos/farmacología , Difenilamina/análogos & derivados , Indazoles/farmacología , Quinasas Quinasa Quinasa PAM/antagonistas & inhibidores , Inhibidores de las Quinasa Fosfoinosítidos-3 , Tomografía de Emisión de Positrones , Sulfonamidas/farmacología , Animales , Benzamidas/agonistas , Línea Celular Tumoral , Neoplasias Colorrectales/diagnóstico por imagen , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/enzimología , Difenilamina/agonistas , Difenilamina/farmacología , Sinergismo Farmacológico , Femenino , Xenoinjertos , Humanos , Indazoles/agonistas , Quinasas Quinasa Quinasa PAM/metabolismo , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Radiografía , Sulfonamidas/agonistas
13.
Immunopharmacol Immunotoxicol ; 34(1): 79-83, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21854184

RESUMEN

CONTEXT: Interleukin-6 (IL-6) plays an important role in immune and inflammatory responses. Midazolam has been reported to modulate IL-6 response. Cyclooxygenase (COX) inhibitors, which are used together with midazolam in some patients undergoing surgery, also modulate it. We hypothesized that their combination results in eliciting the synergistical effect on the IL-6 response. OBJECTIVE: The aim of the present study was to evaluate the effect of the combination of midazolam and a COX inhibitor on IL-6 production. MATERIALS AND METHODS: Peripheral blood mononuclear cells (PBMCs) were isolated from healthy volunteers and incubated with lipopolysaccharide (LPS), midazolam, and/or COX inhibitors, including indomethacin, SC-560, a COX-1 selective inhibitor, and NS-398, a COX-2 selective inhibitor. The supernatant concentrations of IL-6 and prostaglandins (PGs), including PGE2, PGF2α, PGD2, and 15-deoxy-Δ¹²,¹4-prostaglandin J2 (15dPGJ2) were measured. RESULTS: Midazolam had no effect on IL-6 production in the cells incubated for 12 h, and any COX inhibitors also had no effect. However, the combination of midazolam and NS-398 significantly inhibited it. Midazolam raised the concentration of 15dPGJ2 in the supernatant of the cells, but not the concentration of other PGs. DISSCUSSION AND CONCLUSION: The results in the present study demonstrated that the combination of midazolam and a COX-2 inhibitor inhibited LPS-induced IL-6 production in human PBMCs even if each drug separately did not have any effect on it. The finding suggests that their combination is effective against excessive IL-6 production such as severe inflammatory response and that the effect of midazolam on IL-6 production is possibly elicited via 15dPGJ2.


Asunto(s)
Ansiolíticos/farmacología , Inhibidores de la Ciclooxigenasa 2/farmacología , Interleucina-6/biosíntesis , Leucocitos Mononucleares/metabolismo , Lipopolisacáridos/farmacología , Midazolam/farmacología , Nitrobencenos/farmacología , Sulfonamidas/farmacología , Adulto , Ansiolíticos/agonistas , Sinergismo Farmacológico , Femenino , Humanos , Leucocitos Mononucleares/citología , Masculino , Midazolam/agonistas , Nitrobencenos/agonistas , Prostaglandina D2/análogos & derivados , Prostaglandina D2/biosíntesis , Sulfonamidas/agonistas
14.
J Nutr Biochem ; 23(9): 1155-62, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22137263

RESUMEN

In our previous study, we demonstrated that lycopene can inhibit the proliferation of androgen-dependent prostate LNCaP cancer cells through the activation of the peroxisome proliferator-activated receptor gamma (PPARγ)-liver X receptor alpha (LXRα)-ATP-binding cassette transporter 1 (ABCA1) pathway. However, it is still unclear whether lycopene possesses similar effects in androgen-independent prostate cancer cells DU145 and PC-3. As lycopene inhibited the proliferation of both cell types to a similar extent, we chose DU145 cells for most of the subsequent studies. We show that lycopene significantly increased protein and mRNA expression of PPARγ, LXRα and ABCA1 and cholesterol efflux (i.e., decreased cellular cholesterol and increased cholesterol in culture medium). Lycopene (10 µM) in the presence of a specific antagonist of PPARγ (GW9662) or of LXRα (GGPP) restored the proliferation of DU145 cells and significantly suppressed lycopene-induced protein and mRNA expression of PPARγ and LXRα and cholesterol efflux. Liver X receptor α knockdown by siRNA against LXRα significantly promoted the proliferation of DU145 cells, whereas si-LXRα knockdown followed by incubation with lycopene (10 µM) restored the proliferation to the control level. Furthermore, lycopene in combination with the LXRα agonist T0901317 exhibited synergistic effects on cell proliferation and protein expression of PPARγ, LXRα and ABCA1. These results demonstrate that lycopene can inhibit DU145 cell proliferation via PPARγ-LXRα-ABCA1 pathway and that lycopene and T0901317 exhibit synergistic effects.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Antineoplásicos/farmacología , Carotenoides/metabolismo , Hidrocarburos Fluorados/farmacología , Receptores Nucleares Huérfanos/agonistas , Receptores Nucleares Huérfanos/metabolismo , Neoplasias de la Próstata/tratamiento farmacológico , Sulfonamidas/farmacología , Transportador 1 de Casete de Unión a ATP , Transportadoras de Casetes de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/metabolismo , Adenocarcinoma/dietoterapia , Adenocarcinoma/metabolismo , Antineoplásicos/agonistas , Antineoplásicos Fitogénicos/agonistas , Antineoplásicos Fitogénicos/metabolismo , Carotenoides/agonistas , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Colesterol/metabolismo , Suplementos Dietéticos , Interacciones Alimento-Droga , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Hidrocarburos Fluorados/agonistas , Receptores X del Hígado , Licopeno , Masculino , Proteínas de Neoplasias/agonistas , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Receptores Nucleares Huérfanos/antagonistas & inhibidores , Receptores Nucleares Huérfanos/genética , Concentración Osmolar , PPAR gamma/antagonistas & inhibidores , PPAR gamma/genética , PPAR gamma/metabolismo , Neoplasias de la Próstata/dietoterapia , Neoplasias de la Próstata/metabolismo , Interferencia de ARN , ARN Mensajero/metabolismo , ARN Interferente Pequeño , Sulfonamidas/agonistas
15.
J Immunol ; 162(1): 595-602, 1999 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-9886437

RESUMEN

Drugs like sulfamethoxazole (SMX) or lidocaine can be presented to specific human alphabeta+ T cell clones (TCC) by undergoing a noncovalent association with MHC-peptide complexes on HLA-matched APCs. For a better understanding of the molecular basis of the recognition of such drugs by specific TCC, we investigated 1) the fine specificity of the recognizing TCR, 2) the dose-response relationship for the induction of proliferation or cytokine production, and 3) the mechanism of TCR triggering. For that purpose, we tested the reactivity of 11 SMX-specific CD4+ TCC and 2 SMX-specific CD8+ TCC to a panel of 13 different sulfonamide derivatives bearing the same core structure. Five of 13 clones recognized only SMX, while all other clones were responding to as many as 6 different compounds. Some of the compounds needed up to two orders of magnitude higher concentrations than SMX to stimulate TCC, thereby displaying features of weak agonists. Different clones showed clear differences in the minimal drug concentration required for the induction of a proliferative response. Therefore, weaker or stronger agonistic properties were not a characteristic of a given sulfonamide derivative but rather an intrinsic property of the reacting TCR. Finally, the number of down-regulated TCRs was a logarithmic function of the ligand concentration, implicating that specific T cells were activated by serial TCR engagement. Our data demonstrate that, despite the special way of presentation, nonpeptide Ag like drugs appear to interact with the TCR of specific T cells in a similar way as peptide Ags.


Asunto(s)
Receptores de Antígenos de Linfocitos T alfa-beta/biosíntesis , Receptores de Antígenos de Linfocitos T/metabolismo , Sulfametoxazol/inmunología , Sulfonamidas/inmunología , Subgrupos de Linfocitos T/metabolismo , Células Clonales , Reacciones Cruzadas , Relación Dosis-Respuesta Inmunológica , Hipersensibilidad a las Drogas/inmunología , Epítopos de Linfocito T/metabolismo , Humanos , Activación de Linfocitos/efectos de los fármacos , Sulfonamidas/agonistas , Sulfonamidas/química , Subgrupos de Linfocitos T/efectos de los fármacos , Subgrupos de Linfocitos T/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA