Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.448
Filtrar
1.
Prostaglandins Other Lipid Mediat ; 174: 106880, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39121944

RESUMEN

Previous studies have shown prostaglandin E2 (PGE2) produced a marked increase in calcitonin secretion in human C-cells derived from medullary thyroid carcinoma. However, it's unclear whether PGE2 can increase the growth of C cells. In this study, we use TT cells as a C cell model to investigate the effect of PGE2 on the growth of C cells. The results revealed that both PGE2 and arachidonic acid (AA) significantly increased the count of TT cells, whereas indomethacin and Dup697 reduced this count. Notably, an increase in the level of AA was associated with an increase in the number of proliferating TT cells, indicating a dose-response relationship. PGE2 and its receptor agonists (sulprostone and butaprost) enhanced the proliferation of TT cells. By contrast, 17-phenyl-trinor-PGE2 exerted no significant effect on TT cell proliferation, whereas L161982 suppressed it. The positive effect of AA on TT cell proliferation was inhibited by indomethacin, NS398, Dup697 (complete inhibition), and SC560. Both PGE2 and AA increased the level of p-STAT5a. The positive effect of AA on p-STAT5a was completely inhibited by Dup697 but not indomethacin, NS398, or SC560. Treatment with indomethacin or Dup697 alone reduced the level of STAT5a in TT cells. AA increased the level of STAT5a, but this effect was inhibited by indomethacin, NS398, and Dup697. Overall, this study confirms the effect of PGE2 on the proliferation of TT cells. This effect is likely mediated through EP2, EP3, and EP4 receptors and associated with an increase in p-STAT5a level within TT cells.


Asunto(s)
Ácido Araquidónico , Proliferación Celular , Supervivencia Celular , Dinoprostona , Indometacina , Dinoprostona/farmacología , Dinoprostona/metabolismo , Dinoprostona/análogos & derivados , Humanos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Indometacina/farmacología , Ácido Araquidónico/farmacología , Línea Celular Tumoral , División Celular/efectos de los fármacos , Neoplasias de la Tiroides/patología , Neoplasias de la Tiroides/metabolismo , Factor de Transcripción STAT5/metabolismo , Alprostadil/farmacología , Alprostadil/análogos & derivados
2.
Talanta ; 277: 126411, 2024 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-38876034

RESUMEN

Limaprost, an orally administered analogue of prostaglandin E1, possesses potent vasodilatory, antiplatelet, and cytoprotective properties. Due to its extremely low therapeutic doses and exceedingly low plasma concentrations, the pharmacokinetic and bioequivalence studies of limaprost necessitate a highly sensitive quantitative method with a sub-pg/mL level of lower limit of quantification. Moreover, the intensity of endogenous interferences can even exceed the maximum concentration level of limaprost in human plasma, presenting further challenge to the quantification of limaprost. As a result, existing methods have not yet met the necessary level of sensitivity, selectivity, and throughput needed for the quantitative analysis of limaprost in pharmacokinetic and bioequivalence investigations. This study presents a new methodology that combines differential mobility spectrometry (DMS) with liquid chromatography-tandem mass spectrometry (LC-MS/MS) and utilizes a distinctive strategy to achieve more accurate DMS conditions. This integration yields a method that is currently the most sensitive and features the shortest analytical time, making it the sole technique capable of meeting the requirements for limaprost pharmacokinetic and bioequivalence investigations. This method demonstrates robustness and is successfully employed in a pharmacokinetic investigation of limaprost in human subjects, underscoring that the combination of DMS with LC-MS/MS serves as an efficacious strategy for overcoming the challenges inherent in analyzing biological samples afflicted by multiple interferences.


Asunto(s)
Alprostadil , Espectrometría de Masas en Tándem , Espectrometría de Masas en Tándem/métodos , Humanos , Alprostadil/farmacocinética , Alprostadil/análogos & derivados , Alprostadil/sangre , Alprostadil/análisis , Cromatografía Liquida/métodos , Límite de Detección , Cromatografía Líquida con Espectrometría de Masas
3.
World Neurosurg ; 186: e694-e701, 2024 06.
Artículo en Inglés | MEDLINE | ID: mdl-38608818

RESUMEN

BACKGROUND: Patients with Lumbar Spinal Stenosis (LSS) typically complain of back pain and leg pain. These symptoms reduce the quality of life (QoL) and also cause sleep disturbances. This study compares pregabalin and limaprost's efficacy in LSS for pain, disability, QoL, and sleep, aiming to offer insights for medication selection. METHODS: This study was designed as a prospective, randomized, single-center, single-blinded, clinical superiority trial targeting patients with LSS. For 6 weeks, 111 patients per group were administered medication following a standard regimen, after which patient-reported outcomes were measured. The primary outcome was the Visual Analogue Scale (VAS) for back and leg pain, and the secondary outcomes included the Oswestry Disability Index (ODI), European Quality of Life 5 Dimensions (EQ-5D), and sleep quality. RESULTS: After 6 weeks of medication, there were significant improvements over time in the primary outcome, VAS for back pain and leg pain, in both groups, but no significant difference between the 2 groups. Similarly, for the secondary outcomes, ODI and EQ-5D, both groups showed significant improvements, yet there was no significant difference between them. In the subgroup analysis targeting poor sleepers (Pittsburgh sleep quality index, PSQI >5), both groups also exhibited significant improvements in sleep quality, but again, there was no significant difference between the groups. CONCLUSIONS: Efficacy of pregabalin, limaprost in back and leg pain, ODI, EQ-5D, and sleep quality, but there was no significant difference between the 2 groups. Thus, it is advisable to prescribe based on individual drug responses and potential complications.


Asunto(s)
Analgésicos , Vértebras Lumbares , Pregabalina , Calidad de Vida , Estenosis Espinal , Humanos , Pregabalina/uso terapéutico , Estenosis Espinal/tratamiento farmacológico , Estenosis Espinal/complicaciones , Masculino , Femenino , Anciano , Persona de Mediana Edad , Estudios Prospectivos , Resultado del Tratamiento , Método Simple Ciego , Analgésicos/uso terapéutico , Alprostadil/análogos & derivados , Alprostadil/uso terapéutico , Dimensión del Dolor
4.
J Mol Endocrinol ; 73(1)2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38639976

RESUMEN

The prostanoid G protein-coupled receptor (GPCR) EP2 is widely expressed and implicated in endometriosis, osteoporosis, obesity, pre-term labour and cancer. Internalisation and intracellular trafficking are critical for shaping GPCR activity, yet little is known regarding the spatial programming of EP2 signalling and whether this can be exploited pharmacologically. Using three EP2-selective ligands that favour activation of different EP2 pathways, we show that EP2 undergoes limited agonist-driven internalisation but is constitutively internalised via dynamin-dependent, ß-arrestin-independent pathways. EP2 was constitutively trafficked to early and very early endosomes (VEE), which was not altered by ligand activation. APPL1, a key adaptor and regulatory protein of the VEE, did not impact EP2 agonist-mediated cAMP. Internalisation was required for ~70% of the acute butaprost- and AH13205-mediated cAMP signalling, yet PGN9856i, a Gαs-biased agonist, was less dependent on receptor internalisation for its cAMP signalling, particularly in human term pregnant myometrial cells that endogenously express EP2. Inhibition of EP2 internalisation partially reduced calcium signalling activated by butaprost or AH13205 and had no effect on PGE2 secretion. This indicates an agonist-dependent differential spatial requirement for Gαs and Gαq/11 signalling and a role for plasma membrane-initiated Gαq/11-Ca2+-mediated PGE2 secretion. These findings reveal a key role for EP2 constitutive internalisation in its signalling and potential spatial bias in mediating its downstream functions. This, in turn, could highlight important considerations for future selective targeting of EP2 signalling pathways.


Asunto(s)
Proteínas de Unión al GTP , Subtipo EP2 de Receptores de Prostaglandina E , Transducción de Señal , Femenino , Humanos , Embarazo , Alprostadil/análogos & derivados , Alprostadil/farmacología , Alprostadil/metabolismo , AMP Cíclico/metabolismo , Endosomas/metabolismo , Proteínas de Unión al GTP/metabolismo , Células HEK293 , Miometrio/metabolismo , Transporte de Proteínas , Subtipo EP2 de Receptores de Prostaglandina E/metabolismo
5.
J Obstet Gynaecol ; 42(3): 501-504, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-34162287

RESUMEN

Retained products of conception (RPOC) is a complication that occurs in the second trimester of pregnancy. We enrolled 98 women who had a miscarriage or termination with gemeprost in the second trimester of pregnancy. Eighteen cases (18.4%) were RPOC-positive. The gestational week at miscarriage or termination was earlier in the RPOC-positive group than those in the RPOC-negative group (p = .003). The period of the third stage of labour was longer in the RPOC-positive group than in RPOC-negative group (p = .040). The proportion of placental forceps use was higher in the RPOC-positive group than in RPOC-negative group (p = .003). Multivariate logistic regression analysis showed that gestational week (OR: 3.53; p = .04) and use of placental forceps at delivery (OR: 2.21; p = .012) were independent risk factors for RPOC. Earlier gestational weeks at miscarriage or termination and use of placental forceps at delivery were predictive factors for RPOC after second trimester miscarriage or termination with gemeprost.Impact StatementWhat is already known on this subject? There have been some reports on risk factors of RPOC. A previous report showed that the termination of pregnancy with misoprostol at earlier periods was associated with an increased risk of RPOC.What the results of this study add? There have been few studies on the risk factors of RPOC after miscarriage or termination with gemeprost. In this study, we evaluated the risk factors of RPOC after miscarriage or termination of pregnancy with gemeprost in the second trimester. We found that an earlier gestational age (between 12 and 17 weeks) at delivery and using placental forceps to remove placenta were significant risk factors of RPOC after miscarriage or termination of pregnancy with gemeprost in the second trimester.What the implications are of these findings for clinical practice and/or further research? An earlier gestational age and using forceps to remove placenta may be significant risk factors for RPOC. The accurate evaluation and treatment for RPOC is important for maternal life-saving efforts and subsequent pregnancies. Further research is needed to draft a standardised protocol for RPOC.


Asunto(s)
Abortivos no Esteroideos , Aborto Inducido , Aborto Espontáneo , Abortivos no Esteroideos/efectos adversos , Aborto Inducido/efectos adversos , Aborto Espontáneo/etiología , Alprostadil/análogos & derivados , Femenino , Humanos , Lactante , Japón , Placenta , Embarazo , Segundo Trimestre del Embarazo , Estudios Retrospectivos , Factores de Riesgo
6.
Artículo en Inglés | MEDLINE | ID: mdl-34740034

RESUMEN

It is known that prostaglandin E2 (PGE2) induces proliferation of epithelia in bovine endometrial explants, however, the detailed mechanism of regulation of PGE2 in inducing bovine endometrial epithelial cell (bEEC) proliferation is unclear. In this study, we determined whether proliferation of bEECs is promoted by PGE2-prostaglandin E receptor 2 (PTGER2) signaling activation through cell cycle regulation. The results demonstrated that bEECs proliferation was induced by treatment of PGE2 and PTGER2 agonist butaprost. These processes were down-regulated by PTGER2 antagonist AH6809 and CDK inhibitors (LEE011, CDK2 Inhibitor II and Ro 3306). PGE2 and butaprost induced cyclins (A, B1, D1, D3 and E2), cyclin-dependent kinases (CDKs, 1, 2, 4 and 6), and epidermal growth factor (EGF) expression were inhibited by AH6809 treatment in bEECs. Moreover, proliferating cell nuclear antigen (PCNA), cyclooxygenase-1 (COX-1), cyclooxygenase-2 (COX-2), and PTGER2 expression in bEECs were up-regulated by PGE2 and butaprost treatment. Our data demonstrate that PGE2-PTGER2 signaling activation has a direct molecular association with cell cycle regulation and cell proliferation in bEECs. Collectively, these findings will improve our understanding of the roles for PGE2-PTGER2 signaling activation in the physiological and pharmacological processes of bovine endometrium.


Asunto(s)
Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Dinoprostona/metabolismo , Endometrio/citología , Células Epiteliales/metabolismo , Subtipo EP2 de Receptores de Prostaglandina E/metabolismo , Transducción de Señal/efectos de los fármacos , Alprostadil/análogos & derivados , Alprostadil/farmacología , Aminopiridinas/farmacología , Animales , Bovinos , Células Cultivadas , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Quinasas Ciclina-Dependientes/metabolismo , Ciclooxigenasa 1/metabolismo , Ciclooxigenasa 2/metabolismo , Dinoprostona/farmacología , Femenino , Antígeno Nuclear de Célula en Proliferación/metabolismo , Purinas/farmacología , Subtipo EP2 de Receptores de Prostaglandina E/agonistas , Subtipo EP2 de Receptores de Prostaglandina E/antagonistas & inhibidores , Regulación hacia Arriba/efectos de los fármacos , Xantonas
7.
Nutrients ; 13(9)2021 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-34579131

RESUMEN

In our previous study, intravenous (IV) injection of selenium alleviated breast cancer-related lymphedema (BCRL). This secondary analysis aimed to explore the metabolic effects of selenium on patients with BCRL. Serum samples of the selenium-treated (SE, n = 15) or the placebo-controlled (CTRL, n = 14) groups were analyzed by ultra-high-performance liquid chromatography with Q-Exactive Orbitrap tandem mass spectrometry (UHPLC-Q-Exactive Orbitrap/MS). The SE group showed a lower ratio of extracellular water to segmental water (ECW/SW) in the affected arm to ECW/SW in the unaffected arm (arm ECW/SW ratio) than the CTRL group. Metabolomics analysis showed a valid classification at 2-weeks and 107 differential metabolites were identified. Among them, the levels of corticosterone, LTB4-DMA, and PGE3-which are known anti-inflammatory compounds-were elevated in the SE group. Pathway analysis demonstrated that lipid metabolism (glycerophospholipid metabolism, steroid hormone biosynthesis, or arachidonic acid metabolism), nucleotide metabolism (pyrimidine or purine metabolism), and vitamin metabolism (pantothenate and CoA biosynthesis, vitamin B6 metabolism, ascorbate and aldarate metabolism) were altered in the SE group compared to the CTRL group. In addition, xanthurenic acid levels were negatively associated with whole blood selenium level (WBSe) and positively associated with the arm ECW/SW. In conclusion, selenium IV injection improved the arm ECW/SW ratio and altered the serum metabolic profiles in patients with BCRL, and improved the anti-inflammatory process in lipid, nucleotide and vitamin pathways, which might alleviate the symptoms of BCRL.


Asunto(s)
Neoplasias de la Mama/complicaciones , Linfedema/sangre , Linfedema/tratamiento farmacológico , Metabolómica/métodos , Selenito de Sodio/administración & dosificación , Alprostadil/análogos & derivados , Alprostadil/sangre , Cromatografía Líquida de Alta Presión , Corticosterona/sangre , Femenino , Humanos , Inyecciones Intravenosas , Leucotrieno B4/sangre , Linfedema/etiología , Placebos , Espectrometría de Masas en Tándem
8.
Artículo en Inglés | MEDLINE | ID: mdl-34246925

RESUMEN

The additive effects of prostaglandin (PG)-EP2 agonists on a PG-FP agonist toward adipogenesis in two- or three-dimension (2D or 3D) cultures of 3T3-L1 cells was examined by lipid staining, the mRNA expression of adipogenesis related genes, and extracellular matrixes (ECMs) including collagen molecules (Col) -1, -4 and -6, and fibronectin (Fn), and the sizes and physical properties of 3D sphenoids, as measured by a micro-squeezer. The results indicate that adipogenesis induced 1) an enlargement in the sizes of 3D sphenoids, 2) a substantial enhancement in lipid staining, the expression of the PParγ, Ap2 and Leptin genes, and 3) a significant decrease in the stiffness of the 3D sphenoids. These effects were inhibited by bimatoprost acid (BIM-A), but 4) adipogenesis induced significant down-regulation of Col1 and Fn, and the significant up-regulation of the Col4 and Col6 genes were unchanged by BIM-A. On the addition of an EP2 agonist, such as omidenepag (OMD) or butaprost (Buta), to BIM-A, 1) the sizes of the 3D sphenoids were further decreased, 2) lipid staining was decreased (2D; OMD, 3D; Buta) 3) the stiffness of the 3D sphenoids was increased by Buta, 4) the expression of PParγ was up-regulated (2D; Buta) or unchanged (3D), the expression of Ap2 was down-regulated (2D; OMD) or up-regulated (3D; Buta), and the expression of Leptin was increased (2D), 5) the expression of all four (OMD) or all except Col4 (buta) in 2D, and Col1and Col4 (OMD) in 3D were up-regulated. These collective findings indicate that the addition of an EP2 agonist, OMD or Buta significantly modulated the BIM-A induced suppression of adipogenesis as well as physical properties of 2D and 3D cultured 3T3-L1 cells in different manners.


Asunto(s)
Adipogénesis/efectos de los fármacos , Alprostadil/análogos & derivados , Bimatoprost/farmacología , Proteínas de Unión a Ácidos Grasos/efectos de los fármacos , Glicina/análogos & derivados , Leptina/genética , PPAR gamma/efectos de los fármacos , Pirazoles/farmacología , Piridinas/farmacología , Subtipo EP2 de Receptores de Prostaglandina E/agonistas , Receptores de Prostaglandina/agonistas , Células 3T3-L1 , Adipogénesis/genética , Alprostadil/farmacología , Animales , Técnicas de Cultivo Tridimensional de Células , Colágeno Tipo I/efectos de los fármacos , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Colágeno Tipo IV/efectos de los fármacos , Colágeno Tipo IV/genética , Colágeno Tipo IV/metabolismo , Colágeno Tipo VI/efectos de los fármacos , Colágeno Tipo VI/genética , Colágeno Tipo VI/metabolismo , Sinergismo Farmacológico , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/metabolismo , Proteínas de Unión a Ácidos Grasos/genética , Proteínas de Unión a Ácidos Grasos/metabolismo , Fibronectinas/efectos de los fármacos , Fibronectinas/genética , Fibronectinas/metabolismo , Glicina/farmacología , Leptina/metabolismo , Ratones , PPAR gamma/genética , PPAR gamma/metabolismo , Esferoides Celulares/efectos de los fármacos , Esferoides Celulares/metabolismo
9.
Food Funct ; 12(17): 7741-7748, 2021 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-34296713

RESUMEN

The present study aimed to investigate the relationship between the n-3 index, serum metabolites and breast cancer risk. A total of 104 newly diagnosed breast cancer patients and 70 healthy controls were recruited. The erythrocyte phospholipid fatty acid composition was determined by gas-liquid chromatography, and the n-3 index was calculated with the percentage of eicosapentaenoic acid plus docosahexaenoic acid in total fatty acids. Serum metabolomic profiles were analyzed by UHPLC-Q-Exactive Orbitrap/MS. The results showed that the erythrocyte phospholipid n-3 index was significantly lower in breast cancer patients than in healthy controls, and it was inversely associated with breast cancer risk (OR = 0.60; 95% CI: 0.36-0.84). Metabolomics analyses showed that serum 16α-hydroxy dehydroepiandrosterone (DHEA) 3-sulfate, lysophatidylethanolamines (LPE) 22:0/0:0 and hexanoylcarnitine were significantly higher, while thromboxane B3, prostaglandin E3 (PGE3) and 18ß-glycyrrhetinic acid were significantly lower in breast cancer patients than those in healthy controls. In addition, serum 16α-hydroxy DHEA 3-sulfate was inversely correlated with the n-3 index (r = -0.412, p = 0.036). In conclusion, our findings suggest that the lack of n-3 PUFAs might be a potential risk factor for breast cancer, and the serum metabolite 16α-hydroxy DHEA 3-sulfate may play an important role in linking n-3 PUFA deficiency and breast disease etiology.


Asunto(s)
Neoplasias de la Mama/sangre , Ácidos Grasos Omega-3/sangre , Adulto , Alprostadil/análogos & derivados , Alprostadil/sangre , Biomarcadores/sangre , Estudios de Casos y Controles , China , Ácidos Grasos/sangre , Ácidos Grasos/química , Ácidos Grasos Omega-3/química , Femenino , Humanos , Metabolómica , Persona de Mediana Edad , Factores de Riesgo , Tromboxanos/sangre
10.
J Cell Mol Med ; 25(12): 5586-5601, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33982835

RESUMEN

Alternative polarization of macrophages regulates multiple biological processes. While M1-polarized macrophages generally mediate rapid immune responses, M2-polarized macrophages induce chronic and mild immune responses. In either case, polyunsaturated fatty acid (PUFA)-derived lipid mediators act as both products and regulators of macrophages. Prostaglandin E3 (PGE3 ) is an eicosanoid derived from eicosapentaenoic acid, which is converted by cyclooxygenase, followed by prostaglandin E synthase successively. We found that PGE3 played an anti-inflammatory role by inhibiting LPS and interferon-γ-induced M1 polarization and promoting interleukin-4-mediated M2 polarization (M2a). Further, we found that although PGE3 had no direct effect on the growth of prostate cancer cells in vitro, PGE3 could inhibit prostate cancer in vivo in a nude mouse model of neoplasia. Notably, we found that PGE3 significantly inhibited prostate cancer cell growth in a cancer cell-macrophage co-culture system. Experimental results showed that PGE3 inhibited the polarization of tumour-associated M2 macrophages (TAM), consequently producing indirect anti-tumour activity. Mechanistically, we identified that PGE3 regulated the expression and activation of protein kinase A, which is critical for macrophage polarization. In summary, this study indicates that PGE3 can selectively promote M2a polarization, while inhibiting M1 and TAM polarization, thus exerting an anti-inflammatory effect and anti-tumour effect in prostate cancer.


Asunto(s)
Alprostadil/análogos & derivados , Antiinflamatorios/farmacología , Diferenciación Celular , Inflamación/tratamiento farmacológico , Activación de Macrófagos/inmunología , Neoplasias de la Próstata/tratamiento farmacológico , Alprostadil/farmacología , Animales , Polaridad Celular , Humanos , Inflamación/inmunología , Inflamación/patología , Activación de Macrófagos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Neoplasias de la Próstata/inmunología , Neoplasias de la Próstata/patología , Transducción de Señal
11.
Int J Mol Sci ; 22(9)2021 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-33925005

RESUMEN

To elucidate the additive effects of an EP2 agonist, omidenepag (OMD) or butaprost (Buta) on the Rho-associated coiled-coil-containing protein kinase (ROCK) inhibitor, ripasudil (Rip) on adipose tissue, two- or three-dimension (2D or 3D) cultures of 3T3-L1 cells were analyzed by lipid staining, the mRNA expression of adipogenesis-related genes, extracellular matrix (ECM) molecules including collagen (Col) -1, -4 and -6, and fibronectin (Fn), and the sizes and physical properties of 3D organoids, as measured by a micro-squeezer. The results indicate that adipogenesis induced (1) an enlargement of the 3D organoids; (2) a substantial enhancement in lipid staining as well as the expression of the Pparγ, Ap2 and Leptin genes; (3) a significant softening of the 3D organoids, the effects of which were all enhanced by Rip except for Pparγ expression; and (4) a significant downregulation in Col1 and Fn, and a significant upregulation in Col4, Col6, the effects of which were unchanged by Rip. When adding the EP2 agonist to Rip, (1) the sizes of the 3D organoids were reduced substantially; (2) lipid staining was increased (OMD), or decreased (Buta); (3) the stiffness of the 3D organoids was substantially increased in Buta; (4-1) the expression of Pparγ was suppressed (2D, OMD) or increased (2D, Buta), and the expressions of Ap2 were downregulated (2D, 3D) and Leptin was increased (2D) or decreased (3D), (4-2) all the expressions of four ECM molecules were upregulated in 2D (2D), and in 3D, the expression of Col1, Col4 was upregulated. The collective findings reported herein indicate that the addition of an EP2 agonist, OMD or Buta significantly but differently modulate the Rip-induced effects on adipogenesis and the physical properties of 2D and 3D cultured 3T3-L1 cells.


Asunto(s)
Adipogénesis/efectos de los fármacos , Alprostadil/análogos & derivados , Glicina/análogos & derivados , Isoquinolinas/farmacología , Pirazoles/farmacología , Piridinas/farmacología , Sulfonamidas/farmacología , Quinasas Asociadas a rho/antagonistas & inhibidores , Células 3T3-L1 , Alprostadil/farmacología , Animales , Evaluación Preclínica de Medicamentos , Interacciones Farmacológicas , Glicina/farmacología , Ratones , Organoides , Subtipo EP2 de Receptores de Prostaglandina E/agonistas
12.
Cancer Prev Res (Phila) ; 14(6): 619-626, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33707173

RESUMEN

Grape seed procyanidin extract (GSE) has been shown to exert antineoplastic properties in preclinical studies. Recently, we reported findings from a modified phase I, open-label, dose escalation clinical study conducted to evaluate the safety, tolerability, MTD, and potential chemopreventive effects of leucoselect phytosome, a standardized GSE complexed with soy phospholipids to enhance bioavailability, in heavy active and former smokers. Three months of leucoselect phytosome treatment significantly decreased bronchial Ki-67 labeling index (LI), a marker of cell proliferation on the bronchial epithelium. Because GSE is widely used as a supplement to support cardiovascular health, we evaluate the impact of oral leucoselect phytosome on the fasting serum complex lipid metabolomics profiles in our participants. One month of leucoselect phytosome treatment significantly increased eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), the omega-3 polyunsaturated fatty acids (n-3 PUFA) with well-established anticancer properties. Leucoselect phytosome also significantly increased unsaturated phosphatidylcholines (PC), likely from soy phospolipids in the phytosome and functioning as transporters for these PUFAs. Furthermore, 3-month leucoselect phytosome treatment significantly increased serum prostaglandin (PG) E3 (PGE3), a metabolite of EPA with anti-inflammatory and antineoplastic properties. Such increases in PGE3 correlated with reductions of bronchial Ki-67 LI (r = -0.9; P = 0.0374). Moreover, posttreatment plasma samples from trial participants significantly inhibited proliferation of human lung cancer cell lines A549 (adenocarcinoma), H520 (squamous cell carcinoma), DMS114 (small cell carcinoma), and 1198 (preneoplastic cell line). Our findings further support the potential utility of leucoselect phytosome in reducing cardiovascular and neoplastic risks in heavy former and active smokers. PREVENTION RELEVANCE: In this correlative study of leucoselect phytosome for lung cancer chemoprevention in heavy active and former smokers, we demonstrate for the first time, favorable modulations of n-3PUFA and downstream PGE3 in fasting serum, further supporting the chemopreventive potential of leucoselect phytosome against lung cancer.


Asunto(s)
Extracto de Semillas de Uva/administración & dosificación , Neoplasias Pulmonares/prevención & control , Administración Oral , Alprostadil/análogos & derivados , Alprostadil/sangre , Alprostadil/metabolismo , Bronquios/patología , Línea Celular Tumoral , Ácidos Docosahexaenoicos/sangre , Ácidos Docosahexaenoicos/metabolismo , Ácido Eicosapentaenoico/sangre , Ácido Eicosapentaenoico/metabolismo , Extracto de Semillas de Uva/efectos adversos , Humanos , Neoplasias Pulmonares/sangre , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Resultado del Tratamiento
13.
Am J Phys Med Rehabil ; 100(3): 297-302, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33065578

RESUMEN

ABSTRACT: Lumbar spinal stenosis is one of the most commonly diagnosed spinal disorders worldwide and remains a major cause for surgery in older adults. Lumbar spinal stenosis is clinically defined as a progressive degenerative disorder with low back pain and associated neurogenic intermittent claudication. Conservative and surgical management of lumbar spinal stenosis has been shown to be minimally effective on its symptoms. A treatment option that has not been investigated in the United States is the utilization of prostaglandin E1 analogs, which have been used primarily in Japan for the treatment of lumbar spinal stenosis since the 1980s. The vasodilatory and antiplatelet aggregation effects of prostaglandin E1 presumably improve symptoms of lumbar spinal stenosis by increasing blood flow to the spinal nerve roots. This brief report examines the potential vascular pathology of lumbar spinal stenosis, reviews evidence on the use of prostaglandin E1 analog limaprost in Japan for lumbar spinal stenosis, and briefly discusses misoprostol as a possible alternative in the United States. The studies summarized in this report suggest that prostaglandin E1 analogs may provide benefit as a conservative treatment option for patients with lumbar spinal stenosis. However, higher-quality studies conducted in the United States and comparison with other currently used conservative treatments are required before it can be recommended for routine clinical use.


Asunto(s)
Alprostadil/análogos & derivados , Misoprostol/administración & dosificación , Prostaglandinas E Sintéticas/administración & dosificación , Estenosis Espinal/tratamiento farmacológico , Alprostadil/administración & dosificación , Humanos
14.
Molecules ; 25(24)2020 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-33322104

RESUMEN

Prostanit is a novel drug developed for the treatment of peripheral arterial diseases. It consists of a prostaglandin E1 (PGE1) moiety with two nitric oxide (NO) donor fragments, which provide a combined vasodilation effect on smooth muscles and vascular spastic reaction. Prostanit pharmacokinetics, however, remains poorly investigated. Thus, the object of this study was to investigate the pharmacokinetics of Prostanit-related and -affected metabolites in rabbit plasma using the liquid chromatography-mass spectrometry (LC-MS) approach. Besides, NO generation from Prostanit in isolated rat aorta and human smooth muscle cells was studied using the Griess method. In plasma, Prostanit was rapidly metabolized to 1,3-dinitroglycerol (1,3-DNG), PGE1, and 13,14-dihydro-15-keto-PGE1. Simultaneously, the constant growth of amino acid (proline, 4-hydroxyproline, alanine, phenylalanine, etc.), steroid (androsterone and corticosterone), and purine (adenosine, adenosine-5 monophosphate, and guanosine) levels was observed. Glycine, aspartate, cortisol, and testosterone levels were decreased. Ex vivo Prostanit induced both NO synthase-dependent and -independent NO generation. The observed pharmacokinetic properties suggested some novel beneficial activities (i.e., effect prolongation and anti-inflammation). These properties may provide a basis for future research of the effectiveness and safety of Prostanit, as well as for its characterization from a clinical perspective.


Asunto(s)
Alprostadil/análogos & derivados , Alprostadil/farmacocinética , Antiinflamatorios no Esteroideos/farmacocinética , Metabolómica , Óxido Nítrico/antagonistas & inhibidores , Alprostadil/sangre , Animales , Antiinflamatorios no Esteroideos/química , Aorta/efectos de los fármacos , Aorta/metabolismo , Cromatografía Liquida , Humanos , Espectrometría de Masas , Redes y Vías Metabólicas , Metabolómica/métodos , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Óxido Nítrico/biosíntesis , Enfermedad Arterial Periférica/tratamiento farmacológico , Conejos
15.
Invest Ophthalmol Vis Sci ; 61(8): 44, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32725213

RESUMEN

Purpose: Cyclic adenosine monophosphate (cAMP) and peroxisome proliferator-activated receptor alpha (PPARα) levels mediate extracellular matrix (ECM) changes by altering the levels of hypoxia-inducible factor 1-alpha (HIF-1α) in various tissues. We aimed to determine, in the sclera of guinea pigs, whether a prostanoid receptor (EP2)-linked cAMP modulation affects PPARα and HIF-1α signaling during myopia. Methods: Three-week-old guinea pigs (n = 20 in each group), were monocularly injected with either an EP2 agonist (butaprost 1 µmol/L/10 µmol/L), an antagonist (AH6809 10 µmol/L/30 µmol/L) or a vehicle solution for two weeks during normal ocular growth. Separate sets of animals received these injections and underwent form deprivation (FD) simultaneously. Refraction and axial length (AL) were measured at two weeks, followed by scleral tissue isolation for quantitative PCR (qPCR) analysis (n = 10) and cAMP detection (n = 10) using a radioimmunoassay. Results: Butaprost induced myopia development during normal ocular growth, with proportional increases in AL and cAMP levels. FD did not augment the magnitude of myopia or cAMP elevations in these agonist-injected eyes. AH6809 suppressed cAMP increases and myopia progression during FD, but had no effect in a normal visual environment. Of the diverse set of 27 genes related to cAMP, PPARα and HIF-1α signaling and ECM remodeling, butaprost differentially regulated 15 of them during myopia development. AH6809 injections during FD negated such differential gene expressions. Conclusion: EP2 agonism increased cAMP and HIF-1α signaling subsequent to declines in PPARα and RXR mRNA levels, which in turn decreased scleral fibrosis and promoted myopia. EP2 antagonism instead inhibited each of these responses. Our data suggest that EP2 suppression may sustain scleral ECM structure and inhibit myopia development.


Asunto(s)
Alprostadil/análogos & derivados , Matriz Extracelular , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Miopía Degenerativa , PPAR alfa/metabolismo , Subtipo EP2 de Receptores de Prostaglandina E , Xantonas/farmacología , Alprostadil/farmacología , Animales , AMP Cíclico/metabolismo , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/metabolismo , Cobayas , Miopía Degenerativa/etiología , Miopía Degenerativa/metabolismo , Miopía Degenerativa/prevención & control , Antagonistas de Prostaglandina/farmacología , Prostaglandinas E Sintéticas/farmacología , Subtipo EP2 de Receptores de Prostaglandina E/agonistas , Subtipo EP2 de Receptores de Prostaglandina E/antagonistas & inhibidores , Subtipo EP2 de Receptores de Prostaglandina E/metabolismo , Transducción de Señal
16.
Am J Physiol Lung Cell Mol Physiol ; 318(5): L1074-L1083, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-32186399

RESUMEN

Activation of the inflammasome-caspase-1 axis in lung endothelial cells is emerging as a novel arm of the innate immune response to pneumonia and sepsis caused by Pseudomonas aeruginosa. Increased levels of circulating autacoids are hallmarks of pneumonia and sepsis and induce physiological responses via cAMP signaling in targeted cells. However, it is unknown whether cAMP affects other functions, such as P. aeruginosa-induced caspase-1 activation. Herein, we describe the effects of cAMP signaling on caspase-1 activation using a single cell flow cytometry-based assay. P. aeruginosa infection of cultured lung endothelial cells caused caspase-1 activation in a distinct population of cells. Unexpectedly, pharmacological cAMP elevation increased the total number of lung endothelial cells with activated caspase-1. Interestingly, addition of cAMP agonists augmented P. aeruginosa infection of lung endothelial cells as a partial explanation underlying cAMP priming of caspase-1 activation. The cAMP effect(s) appeared to function as a priming signal because addition of cAMP agonists was required either before or early during the onset of infection. However, absolute cAMP levels measured by ELISA were not predictive of cAMP-priming effects. Importantly, inhibition of de novo cAMP synthesis decreased the number of lung endothelial cells with activated caspase-1 during infection. Collectively, our data suggest that lung endothelial cells rely on cAMP signaling to prime caspase-1 activation during P. aeruginosa infection.


Asunto(s)
Caspasa 1/genética , AMP Cíclico/metabolismo , Células Endoteliales/metabolismo , Pseudomonas aeruginosa/metabolismo , Transducción de Señal , 1-Metil-3-Isobutilxantina/farmacología , 8-Bromo Monofosfato de Adenosina Cíclica/farmacología , Adenina/análogos & derivados , Adenina/farmacología , Alprostadil/análogos & derivados , Alprostadil/farmacología , Animales , Caspasa 1/metabolismo , Proliferación Celular/efectos de los fármacos , Colforsina/farmacología , AMP Cíclico/agonistas , AMP Cíclico/antagonistas & inhibidores , GMP Cíclico/análogos & derivados , GMP Cíclico/farmacología , Dinoprostona/farmacología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/microbiología , Células Endoteliales/patología , Regulación de la Expresión Génica , Interacciones Huésped-Patógeno/genética , Inflamasomas/efectos de los fármacos , Inflamasomas/genética , Inflamasomas/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Pulmón/metabolismo , Pulmón/microbiología , Pulmón/patología , Cultivo Primario de Células , Pseudomonas aeruginosa/efectos de los fármacos , Pseudomonas aeruginosa/crecimiento & desarrollo , Ratas , Rolipram/farmacología , Análisis de la Célula Individual
17.
Contraception ; 101(5): 315-320, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32032639

RESUMEN

OBJECTIVES: To evaluate the characteristics, clinical information, and storage instructions contained in package inserts from medical abortion commodities collected in low- and middle-income countries. STUDY DESIGN: From November 2017 to February 2018 mifepristone, misoprostol, and combined mifepristone-misoprostol (combipack) products were collected to populate the Medical Abortion Commodities Database. We extracted stated indications for use, storage instructions, and date of last revision from each package insert obtained. For those inserts listing medical abortion as an indication, we also extracted eligibility criteria, recommended regimens, side effects, and contraindications. RESULTS: We identified 41 package inserts from 20 countries; 19 (46%) listed medical abortion as an indication including all 7 combipacks, all 7 mifepristone products, and 5/27 (19%) misoprostol products. Date of last insert revision ranged from 1991 to 2016. Gestational age limits for early medical abortion ranged from 49 days to "first trimester." Three (43%) mifepristone products recommended a 600 mg oral dose and two (29%) recommended regimens with gemeprost. Eighteen (67%) misoprostol and one (14%) combipack inserts recommended protection from moisture. CONCLUSIONS: The characteristics, clinical information, and storage instructions in medical abortion product package inserts from a variety of field settings in low- and middle-income countries included inadequate storage instructions and outdated gestational age limits and regimens. IMPLICATIONS: There is an urgent need to revisit approved inserts for medical abortion products in low- and middle-income countries to ensure information is accurate and reflects the current evidence base. Simultaneously, providing supplemental instructions targeted at users may fill some gaps. People have a right to accurate information to ensure a safe and effective medical abortion experience.


Asunto(s)
Abortivos/uso terapéutico , Aborto Inducido/métodos , Mifepristona/uso terapéutico , Misoprostol/uso terapéutico , Etiquetado de Productos/métodos , Abortivos/efectos adversos , Aborto Inducido/efectos adversos , Alprostadil/análogos & derivados , Alprostadil/uso terapéutico , Estudios Transversales , Países en Desarrollo , Quimioterapia Combinada , Femenino , Humanos , Mifepristona/efectos adversos , Misoprostol/efectos adversos , Embarazo , Primer Trimestre del Embarazo , Resultado del Tratamiento
18.
Pharmacology ; 104(5-6): 267-275, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31434088

RESUMEN

Epigallocatechin-3-gallate (EGCG), the principal catechin of green tea, modulates different molecular mechanisms underlying hepatocellular carcinoma (HCC). Accumulating studies showed that the activation of prostaglandin (PG) receptor EP1 promotes cell migration and invasion in different cancers, which could be inverted by blocking the EP1 receptor. This study investigated the synergetic effects of EP1-selective antagonist ONO-8711 and EGCG treatment on HCC to better understand the potential strategy to treat HCC. We found that EGCG significantly inhibited PGE2 and EP1-selective agonist induced migration of HCC cells and increased the ratio of Bax/Bcl-2 even in the presence of ONO-DI-004 or PGE2. ONO-8711 significantly inhibited PGE2-induced HCC proliferation while increased the inhibitory effect of EGCG on HCC cell viability and migration ability compared with EGCG alone. These findings suggest that a combination of ONO-8711 and EGCG is a potential treatment for HCC therapy.


Asunto(s)
Alprostadil/análogos & derivados , Carcinoma Hepatocelular/tratamiento farmacológico , Catequina/análogos & derivados , Neoplasias Hepáticas/tratamiento farmacológico , Subtipo EP1 de Receptores de Prostaglandina E/antagonistas & inhibidores , Alprostadil/farmacología , Apoptosis/efectos de los fármacos , Carcinoma Hepatocelular/metabolismo , Catequina/farmacología , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Dinoprostona/metabolismo , Sinergismo Farmacológico , Células Hep G2 , Humanos , Neoplasias Hepáticas/metabolismo , Subtipo EP1 de Receptores de Prostaglandina E/metabolismo , Cicatrización de Heridas/efectos de los fármacos
19.
Acta Physiol (Oxf) ; 227(1): e13291, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31054202

RESUMEN

AIM: Renal fibrosis plays a pivotal role in the development and progression of chronic kidney disease, which affects 10% of the adult population. Previously, it has been demonstrated that the cyclooxygenase-2 (COX-2)/prostaglandin (PG) system influences the progression of renal injury. Here, we evaluated the impact of butaprost, a selective EP2 receptor agonist, on renal fibrosis in several models of kidney injury, including human tissue slices. METHODS: We studied the anti-fibrotic efficacy of butaprost using Madin-Darby Canine Kidney (MDCK) cells, mice that underwent unilateral ureteral obstruction and human precision-cut kidney slices. Fibrogenesis was evaluated on a gene and protein level by qPCR and Western blotting. RESULTS: Butaprost (50 µM) reduced TGF-ß-induced fibronectin (FN) expression, Smad2 phosphorylation and epithelial-mesenchymal transition in MDCK cells. In addition, treatment with 4 mg/kg/day butaprost attenuated the development of fibrosis in mice that underwent unilateral ureteral obstruction surgery, as illustrated by a reduction in the gene and protein expression of α-smooth muscle actin, FN and collagen 1A1. More importantly, a similar anti-fibrotic effect of butaprost was observed in human precision-cut kidney slices exposed to TGF-ß. The mechanism of action of butaprost appeared to be a direct effect on TGF-ß/Smad signalling, which was independent of the cAMP/PKA pathway. CONCLUSION: In conclusion, this study demonstrates that stimulation of the EP2 receptor effectively mitigates renal fibrogenesis in various fibrosis models. These findings warrant further research into the clinical application of butaprost, or other EP2 agonists, for the inhibition of renal fibrosis.


Asunto(s)
Alprostadil/análogos & derivados , Fibrosis/tratamiento farmacológico , Enfermedades Renales/metabolismo , Riñón/efectos de los fármacos , Subtipo EP2 de Receptores de Prostaglandina E/agonistas , Anciano , Alprostadil/farmacología , Animales , Línea Celular , Perros , Transición Epitelial-Mesenquimal/efectos de los fármacos , Femenino , Humanos , Riñón/patología , Enfermedades Renales/patología , Antígeno MART-1 , Masculino , Ratones , Ratones Endogámicos C57BL , Técnicas de Cultivo de Tejidos , Obstrucción Ureteral , Agentes Urológicos/farmacología
20.
J Med Chem ; 62(9): 4731-4741, 2019 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-30964292

RESUMEN

A series of small-molecule full agonists of the prostaglandin E2 type 4 (EP4) receptor have been generated and evaluated for binding affinity and cellular potency. KMN-80 and its gem-difluoro analog KMN-159 possess high selectivity relative to other prostanoid receptors. Difluoro substitution is positioned alpha to the lactam ring carbonyl and results in KMN-159's fivefold increase in potency versus KMN-80. The two analogs exhibit electronic and conformational variations, including altered nitrogen hybridization and lactam ring puckering, that may drive the observed difluoro-associated increased potency within this four-compound series.


Asunto(s)
Alprostadil/análogos & derivados , Alprostadil/farmacología , Ácidos Heptanoicos/farmacología , Lactamas/farmacología , Pirrolidinas/farmacología , Subtipo EP4 de Receptores de Prostaglandina E/agonistas , Alprostadil/metabolismo , Animales , Sitios de Unión , Células CHO , Células CACO-2 , Cricetulus , Humanos , Lactamas/síntesis química , Lactamas/metabolismo , Modelos Químicos , Simulación del Acoplamiento Molecular , Estructura Molecular , Teoría Cuántica , Subtipo EP3 de Receptores de Prostaglandina E/química , Subtipo EP3 de Receptores de Prostaglandina E/metabolismo , Subtipo EP4 de Receptores de Prostaglandina E/química , Subtipo EP4 de Receptores de Prostaglandina E/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...