Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 79
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Eur J Pharmacol ; 972: 176572, 2024 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-38614381

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is the severe form of interstitial pneumonias. Acute exacerbation (AE) of IPF is characterized by progressive lung fibrosis with the irreversible lung function decline and inflammation, and is often fatal with poor prognosis. However, the physiological and molecular mechanisms in AE of IPF are still not fully understood. In this study, we investigated the mechanism underlying AE of IPF, using bleomycin (BLM) and lipopolysaccharide (LPS) (BLM + LPS)-treated mice. The mice were treated with a single dose of 1.5 mg/kg BLM (on day 0) and/or 0.5 mg/kg LPS (on day 14), and maintained for another 7 days (total 21 days). Administration of BLM + LPS more severely aggravated the respiratory function, fibrosis, and inflammation in the lungs, together with the elevated interleukin-6 level in bronchoalveolar lavage fluid, than the control or BLM alone-treated mice. Moreover, the terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) assay demonstrated that subsequent treatment with LPS elevated cell death in the lungs of BLM-administered mice. Furthermore, the expression levels of mixed lineage kinase domain-like protein (MLKL), a marker of necroptotic cell death, and CD68-positive macrophages were increased, and most of them were co-stained in the lungs of BLM + LPS-treated mice. These results, taken together, indicate that BLM + LPS treatment showed more exacerbated the respiratory function with extensive fibrosis and inflammation than treatment with BLM alone in mice. Fibrosis and inflammation in AE of IPF seen in BLM + LPS-administered mice included an increase in macrophages and their necroptotic cell death.


Asunto(s)
Bleomicina , Fibrosis Pulmonar Idiopática , Lipopolisacáridos , Macrófagos , Animales , Bleomicina/toxicidad , Fibrosis Pulmonar Idiopática/patología , Fibrosis Pulmonar Idiopática/inducido químicamente , Fibrosis Pulmonar Idiopática/metabolismo , Ratones , Masculino , Macrófagos/efectos de los fármacos , Macrófagos/patología , Macrófagos/metabolismo , Progresión de la Enfermedad , Ratones Endogámicos C57BL , Pulmón/patología , Pulmón/efectos de los fármacos , Necroptosis/efectos de los fármacos , Interleucina-6/metabolismo , Líquido del Lavado Bronquioalveolar/citología
2.
Bioorg Med Chem ; 85: 117238, 2023 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-37028120

RESUMEN

Farnesoid X receptor (FXR) and peroxisome proliferator-activated receptor (PPAR)γ are nuclear receptor 1 superfamily of transcription factors. FXR and PPARγ agonists have been individually investigated in clinical trial of anti-diabetic agents in the patients with nonalcoholic fatty liver disease (NAFLD). Regarding recent agonist development, the partial agonists for FXR and PPARγ are drawing attention from the standpoint of avoiding overactive responses caused by full agonists. In this article, we report that 18 with a benzimidazole scaffold possesses FXR/PPARγ dual partial agonistic activity. In addition, 18 shares the ability to reduce cyclin-dependent kinase 5-mediated phosphorylation of PPARγ-Ser273 and the metabolic stability in mouse liver microsome assay. To date, there are no published reports on FXR/PPARγ dual partial agonists with biological profiles similar to 18. Thus, the analog would be a feasible candidate as an unprecedented approach to NAFLD associated with type 2 diabetes mellitus.


Asunto(s)
Diabetes Mellitus Tipo 2 , Enfermedad del Hígado Graso no Alcohólico , Ratones , Animales , PPAR gamma/agonistas , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Factores de Transcripción , Hipoglucemiantes/farmacología
3.
Elife ; 122023 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-36779701

RESUMEN

In the developing central nervous system, oligodendrocyte precursor cells (OPCs) differentiate into oligodendrocytes, which form myelin around axons. Oligodendrocytes and myelin are essential for the function of the central nervous system, as evidenced by the severe neurological symptoms that arise in demyelinating diseases such as multiple sclerosis and leukodystrophy. Although many cell-intrinsic mechanisms that regulate oligodendrocyte development and myelination have been reported, it remains unclear whether interactions among oligodendrocyte-lineage cells (OPCs and oligodendrocytes) affect oligodendrocyte development and myelination. Here, we show that blocking vesicle-associated membrane protein (VAMP) 1/2/3-dependent exocytosis from oligodendrocyte-lineage cells impairs oligodendrocyte development, myelination, and motor behavior in mice. Adding oligodendrocyte-lineage cell-secreted molecules to secretion-deficient OPC cultures partially restores the morphological maturation of oligodendrocytes. Moreover, we identified L-type prostaglandin D synthase as an oligodendrocyte-lineage cell-secreted protein that promotes oligodendrocyte development and myelination in vivo. These findings reveal a novel autocrine/paracrine loop model for the regulation of oligodendrocyte and myelin development.


Asunto(s)
Vaina de Mielina , Oligodendroglía , Animales , Ratones , Oligodendroglía/metabolismo , Vaina de Mielina/metabolismo , Neurogénesis/fisiología , Exocitosis , Diferenciación Celular/fisiología
4.
Biol Pharm Bull ; 45(8): 985-991, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35908908

RESUMEN

Prostaglandins (PGs) are lipid-derived autacoids that are synthesized from arachidonic acid by the action of cyclooxygenases and PG terminal synthases. PGs consist of PGD2, PGE2, PGF2α, prostacyclin (PGI2), and thromboxane A2, which act through G protein-coupled receptors. PGs sustain homeostatic functions and exert a variety of pathophysiological roles to regulate the development of various diseases such as obesity and dyslipidemia. Adipocytes (fat cells) have the unique capacity to accumulate large amounts of lipids as energy source in lipid droplets. Adipogenesis is the process of differentiation from preadipocytes to mature adipocytes, which is regulated by various adipogenic transcription factors. Obesity is defined as an abnormal increase in adipose tissue mass and is considered to be a risk factor for the development of lifestyle-related diseases including cardiovascular disease, hyperlipidemia, and type 2 diabetes mellitus. This review summarizes insights into the roles of PGD2, PGF2α, and their synthases in the regulation of adipogenesis and obesity.


Asunto(s)
Adipogénesis , Diabetes Mellitus Tipo 2 , Humanos , Obesidad , Prostaglandina D2 , Prostaglandinas , Prostaglandinas F
5.
Biochim Biophys Acta Mol Cell Res ; 1869(3): 119203, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34968576

RESUMEN

Leukotriene (LT) C4 synthase (LTC4S) catalyzes the conversion from LTA4 to LTC4, which is a proinflammatory lipid mediator in asthma and other inflammatory diseases. LTC4 is metabolized to LTD4 and LTE4, all of which are known as cysteinyl (Cys) LTs and exert physiological functions through CysLT receptors. LTC4S is expressed in adipocytes. However, the function of CysLTs and the regulatory mechanism in adipocytes remain unclear. In this study, we investigated the expression of LTC4S and production of CysLTs in murine adipocyte 3T3-L1 cells and their underlying regulatory mechanisms. Expression of LTC4S and production of LTC4 and CysLTs increased during adipogenesis, whereas siRNA-mediated suppression of LTC4S expression repressed adipogenesis by reducing adipogenic gene expression. The CysLT1 receptor, one of the two LTC4 receptors, was expressed in adipocytes. LTC4 and LTD4 increased the intracellular triglyceride levels and adipogenic gene expression, and their enhancement was suppressed by co-treatment with pranlukast, a CysLT1 receptor antagonist. Moreover, the expression profiles of LTC4S gene/protein during adipogenesis resembled those of peroxisome proliferator-activated receptor (PPAR) γ. LTC4S expression was further upregulated by treatment with troglitazone, a PPARγ agonist. Promoter-luciferase and chromatin immunoprecipitation assays showed that PPARγ directly bound to the PPAR response element of the LTC4S gene promoter in adipocytes. These results indicate that the LTC4S gene expression was enhanced by PPARγ, and LTC4 and LTD4 activated adipogenesis through CysLT1 receptors in 3T3-L1 cells. Thus, LTC4S and CysLT1 receptors are novel potential targets for the treatment of obesity.


Asunto(s)
Adipocitos/citología , Adipogénesis , Glutatión Transferasa/genética , Leucotrieno C4/farmacología , Leucotrieno D4/farmacología , PPAR gamma/metabolismo , Receptores de Leucotrienos/metabolismo , Células 3T3-L1 , Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Animales , Regulación de la Expresión Génica , Glutatión Transferasa/metabolismo , Ratones , PPAR gamma/genética , Regiones Promotoras Genéticas , Receptores de Leucotrienos/genética
6.
Int J Mol Sci ; 22(23)2021 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-34884648

RESUMEN

Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are severe respiratory disorders that are caused by aspiration, sepsis, trauma, and pneumonia. A clinical feature of ALI/ARDS is the acute onset of severe hypoxemia, and the mortality rate, which is estimated at 38-50%, remains high. Although prostaglandins (PGs) are detected in the bronchoalveolar lavage fluid of patients with ALI/ARDS, the role of PGF2α in ALI remains unclear. We aimed to clarify the role of PGF2α/PGF2α receptor (FP) signaling in acid-induced ALI using an FP receptor antagonist, AL8810. Intratracheal injection of hydrochloric acid (HCl) increased neutrophil migration into the lungs, leading to respiratory dysfunction. Pre-administration of AL8810 further increased these features. Moreover, pre-treatment with AL8810 enhanced the HCl-induced expression of pro-inflammatory cytokines and neutrophil migratory factors in the lungs. Administration of HCl decreased the gene expression of lung surfactant proteins, which was further reduced by co-administration of AL8810. Administration of AL8810 also increased lung edema and reduced mRNA expression of epithelial sodium channel in the lungs, indicating that AL8810 reduced fluid clearance. Furthermore, AL8810 also increased lipopolysaccharide-induced expression of adhesion molecules such as intracellular adhesion molecule-1 and E-selectin in human umbilical vein endothelial cells. These results indicate that inhibition of FP receptors by AL8810 exacerbated HCl-induced ALI.


Asunto(s)
Lesión Pulmonar Aguda/metabolismo , Pulmón/efectos de los fármacos , Neumonía/metabolismo , Receptores de Prostaglandina/antagonistas & inhibidores , Síndrome de Dificultad Respiratoria/metabolismo , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/patología , Animales , Modelos Animales de Enfermedad , Femenino , Ácido Clorhídrico/toxicidad , Pulmón/metabolismo , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Neumonía/inducido químicamente , Neumonía/inmunología , Neumonía/patología , Prostaglandinas F/metabolismo , Síndrome de Dificultad Respiratoria/inducido químicamente , Síndrome de Dificultad Respiratoria/patología
7.
Eur J Pharm Biopharm ; 167: 116-126, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34363979

RESUMEN

This study aimed to design dry powder inhaler formulations using a hydrophilic polymeric polysaccharide, phytoglycogen (PyG), as a multi-functional additive that increases the phagocytic activity of macrophage-like cells and enhances pulmonary delivery of drugs. The safety and usefulness of PyG were determined using in vitro cell-based studies. Dry powder inhaler formulations of an antitubercular drug, rifampicin, were fabricated by spray drying with PyG. The cytotoxicity, effects on phagocytosis, particle size, and morphology were evaluated. The aerosolization properties of the powder formulations were evaluated using an Andersen cascade impactor (ACI). Scanning electron microscope images of the particles on each ACI stage were captured to observe the deposition behavior. PyG showed no toxicity in A549, Calu-3, or RAW264.7 cell lines. At concentrations of 0.5 and 1 g/L, PyG facilitated the cellular uptake of latex beads and the expression of pro-inflammatory cytokine genes in RAW264.7 cells. Formulations with outstanding inhalation potential were produced. The fine particle fraction (aerodynamic size 2-7 µm) of the porous particle batch reached nearly 60%, whereas in the formulation containing wrinkled carrier particles, the extra-fine particle fraction (aerodynamic particle size < 2 µm) was 25.0% ± 1.7%. The deposition of porous and wrinkled particles on individual ACI stages was distinct. The inclusion of PyG dramatically improved the inhalation performance of porous and wrinkled powder formulations. These easily inhaled immunostimulatory carrier particles may advance the state of research by enhancing the therapeutic effect and alveolar delivery of antitubercular drugs.


Asunto(s)
Antituberculosos/administración & dosificación , Sistemas de Liberación de Medicamentos , Glucógeno/química , Rifampin/administración & dosificación , Células A549 , Administración por Inhalación , Aerosoles , Animales , Antituberculosos/química , Antituberculosos/toxicidad , Línea Celular Tumoral , Química Farmacéutica/métodos , Inhaladores de Polvo Seco , Excipientes/química , Humanos , Ratones , Tamaño de la Partícula , Porosidad , Células RAW 264.7 , Rifampin/química , Rifampin/toxicidad , Distribución Tisular
8.
Chem Biol Interact ; 346: 109595, 2021 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-34302803

RESUMEN

Glycyrrhizic acid (GA), a major constituent of the root of licorice (Glycyrrhiza glabra), and has various biological activities, including anti-obesity property. However, the molecular mechanism of anti-adipogenic effect of GA is still unclear. In this study, we investigated the anti-adipogenic effects of GA in mouse adipocytic 3T3-L1 cells and elucidated its underlying molecular mechanism. GA decreased the intracellular triglyceride level. The expression levels of the adipogenic and lipogenic genes were lowered by treatment with GA in a concertation-dependent manner. In contrast, GA did not affect the lipolytic gene expression and the released glycerol level. GA suppressed the early stage of adipogenesis when it was added for 0-3 h after initiation of adipogenesis. Moreover, GA reduced the mRNA levels of CCAAT/enhancer binding protein (C/EBP) ß and C/EBPδ, both of which activate the early stage of adipogenesis. Furthermore, GA decreased phosphorylation of extracellular signal-regulated kinase [ERK: p44/42 mitogen-activated protein kinase (MAPK)] in the early stage of adipogenesis. In addition, a MAPK kinase (MEK) inhibitor, PD98059 reduced the C/EBPß and C/EBPδ gene expression. These results indicate that GA suppressed the early stage of adipogenesis through repressing the MEK/ERK-mediated C/EBPß and C/EBPδ expression in 3T3-L1 cells. Thus, GA has an anti-adipogenic ability and a possible agent for treatment of obesity.


Asunto(s)
Adipogénesis/efectos de los fármacos , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Proteína delta de Unión al Potenciador CCAAT/metabolismo , Diferenciación Celular/efectos de los fármacos , Ácido Glicirrínico/farmacología , Células 3T3-L1 , Animales , Proteína beta Potenciadora de Unión a CCAAT/genética , Proteína delta de Unión al Potenciador CCAAT/genética , Supervivencia Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Flavonoides/farmacología , Transportador de Glucosa de Tipo 4/metabolismo , Lipólisis/efectos de los fármacos , Ratones , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , PPAR gamma/metabolismo , Fosforilación/efectos de los fármacos , Triglicéridos/metabolismo
9.
Neurotoxicology ; 85: 186-200, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34077701

RESUMEN

Formononetin is an isoflavone, found in herbs like Trifolium pratense, which executes a variety of physiological activities including anti-neurodegenerative effect. However, the molecular mechanism of formononetin-mediated neuroprotection remains unclear. In this study, we investigated the protective effect of formononetin on hydrogen peroxide (H2O2)-induced death of human neuroblastoma SH-SY5Y cells and its underlying molecular mechanism. Formononetin suppressed H2O2-induced cytotoxicity. H2O2-induced increase in the intracellular reactive oxygen species (ROS) levels was decreased by formononetin, together with the enhanced expression of the antioxidant genes. H2O2-induced elevation of the Bax/Bcl-2 ratio and cleaved caspase-3 and caspase-7 levels were lowered by formononetin treatment. Moreover, formononetin repressed H2O2-induced phosphorylation of mitogen-activated protein kinases (MAPKs). Nuclear factor erythroid 2-related factor 2 (Nrf2) siRNA decreased antioxidant gene expression and elevated the H2O2-induced ROS level in the formononetin-treated cells. Furthermore, the phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) signaling is involved in the activation of the nuclear translocation of Nrf2. These results indicate that the neuroprotective effect of formononetin against H2O2-induced cell death is due to a decrease in the ROS level with the enhanced expression of the antioxidant genes through activation of the PI3K/Akt-Nrf2 signaling. In addition, formononetin suppressed apoptosis through inhibition of phosphorylation of MAPKs in SH-SY5Y cells. Thus, formononetin is a potential therapeutic agent for the treatment of neurodegenerative diseases.


Asunto(s)
Muerte Celular/efectos de los fármacos , Peróxido de Hidrógeno/toxicidad , Isoflavonas/farmacología , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Neuronas/efectos de los fármacos , Especies Reactivas de Oxígeno/antagonistas & inhibidores , Antioxidantes/metabolismo , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Muerte Celular/fisiología , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Expresión Génica , Humanos , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Factor 2 Relacionado con NF-E2/biosíntesis , Factor 2 Relacionado con NF-E2/genética , Neuronas/metabolismo , Fosfatidilinositol 3-Quinasas/biosíntesis , Fosfatidilinositol 3-Quinasas/genética , Fitoestrógenos/farmacología , Proteínas Proto-Oncogénicas c-akt/biosíntesis , Proteínas Proto-Oncogénicas c-akt/genética , Especies Reactivas de Oxígeno/metabolismo
10.
Bioorg Med Chem Lett ; 41: 128026, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33839252

RESUMEN

Farnesoid X receptor (FXR) controls gene-expression relevant to various diseases including nonalcoholic steatohepatitis and has become a drug target to regulate metabolic aberrations. However, some side effects of FXR agonists reported in clinical development such as an increase in blood cholesterol levels incentivize the development of partial agonists to minimize side effects. In this study, to identify a new partial agonist, we analyzed the computational structure-activity relationship (SAR) of FXR agonists previously developed in our laboratories using molecular dynamics simulations. SAR analysis showed that fluctuations in the H8 helix, by ligand binding, of the ligand-binding domain (LBD) of FXR may influence agonistic activity. Based on this observation, 6 was newly designed as a partial agonist and synthesized. As a result of biological evaluations, 6 showed weak agonistic activity (40.0% relative agonistic activity to the full-agonist GW4064) and a potent EC50 value (55.5 nM). The successful identification of the new potent partial agonist 6 suggested that helix fluctuation in the LBD induced by ligands could be one way to develop partial agonists.


Asunto(s)
Ácido Quenodesoxicólico/farmacología , Diseño de Fármacos , Simulación de Dinámica Molecular , Receptores Citoplasmáticos y Nucleares/agonistas , Sitios de Unión/efectos de los fármacos , Ácido Quenodesoxicólico/química , Relación Dosis-Respuesta a Droga , Humanos , Ligandos , Estructura Molecular , Receptores Citoplasmáticos y Nucleares/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Relación Estructura-Actividad
11.
ACS Med Chem Lett ; 12(3): 420-425, 2021 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-33738070

RESUMEN

We describe the discovery of analog 15 (FLG249), which is an orally active and nonsteroidal farnesoid X receptor (FXR) antagonist in mice with unique profiles, such as a propensity for ileum distribution and the significant control in the expression level of three FXR target genes in mouse ileum. Key design features incorporated in 15 were the introduction of metabolically stable groups in potent and metabolically labile antagonist 9. Our pursuit ultimately identified FXR antagonist 15, which has enabled its assessment in a drug discovery program.

12.
FEBS J ; 288(14): 4249-4266, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33452755

RESUMEN

Viral infection is a significant burden to health care worldwide. Statins, 3-hydroxy-3-methyl-glutaryl-coenzyme A reductase inhibitors, are widely used as cholesterol-lowering drugs. Recently, long-term statin therapy was shown to reduce the antiviral immune response; however, the underlying molecular mechanisms are unclear. Here, we found that simvastatin decreased polyinosinic-polycytidylic acid [poly(I:C)]-induced expression of antiviral interferon (IFN)-ß and IFN-stimulated genes (ISGs) in the bronchoalveolar lavage fluid (BALF) and lungs of mice with high-fat diet-induced hyperlipidemia. Macrophages were the dominant cell type in the BALF of poly(I:C)-treated mice. We examined the effects of simvastatin in primary lung macrophages and found that simvastatin suppressed poly(I:C)-induced expression of IFN-ß and ISGs. We examined the molecular mechanisms of statin-mediated inhibition of antiviral gene expression using murine macrophage-like cell line, J774.1/JA-4. Simvastatin and pitavastatin decreased poly(I:C)-induced expression of IFN-ß and ISGs. Moreover, they repressed poly(I:C)-induced phosphorylation of IFN regulatory factor (IRF) 3 and signal transducers and activators of transcription (STAT) 1, which is involved in Janus kinase (JAK)/STAT signaling. Mevalonate and geranylgeranyl pyrophosphate (GGPP), but not cholesterol, counteracted the negative effect of statins on IFN-ß and ISG expression and phosphorylation of IRF3 and STAT1. The geranylgeranyltransferase inhibitor suppressed poly(I:C)-induced expression of IFN-ß and ISGs and phosphorylation of IRF3 and STAT1. These results suggest that statins suppressed the expression of IFN-ß and ISGs in poly(I:C)-treated hyperlipidemic mice and murine macrophages and that these effects occurred through the inhibition of IRF3 and JAK/STAT signaling in macrophages. Furthermore, GGPP recovered the statin-suppressed IRF3 and JAK/STAT signaling in poly(I:C)-treated macrophages.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Hiperlipidemias/tratamiento farmacológico , Factores Reguladores del Interferón/metabolismo , Interferón beta/metabolismo , Macrófagos/efectos de los fármacos , Poli I-C/toxicidad , Animales , Hiperlipidemias/inducido químicamente , Hiperlipidemias/metabolismo , Hiperlipidemias/patología , Inductores de Interferón/toxicidad , Factor 3 Regulador del Interferón/antagonistas & inhibidores , Factor 3 Regulador del Interferón/genética , Factor 3 Regulador del Interferón/metabolismo , Factores Reguladores del Interferón/genética , Interferón beta/genética , Quinasas Janus/antagonistas & inhibidores , Quinasas Janus/genética , Quinasas Janus/metabolismo , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/patología , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Factor de Transcripción STAT3/antagonistas & inhibidores , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo
13.
Toxicology ; 450: 152686, 2021 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-33486071

RESUMEN

6-Hydroxydopamine (6-OHDA) is a neurotoxin that destroy dopaminergic neurons and widely used to establish animal models of Parkinson's disease. Prostaglandins (PGs) are involved in various cellular processes, including the damage and repair of neuronal cells. However, the function of PGF2α in neuronal cells remains unclear. In this study, we investigated the effects of PGF2α against 6-OHDA-mediated toxicity in human neuroblastoma SH-SY5Y cells and elucidated its underlying molecular mechanism. When the cells were treated with 6-OHDA (50 µM) for 6 h, the expression levels of PGF2α synthetic enzymes; cyclooxygenase-2 and aldo-keto reductase 1C3 as PGF2α synthase were enhanced in an incubation-time-dependent manner. In addition, the production of PGF2α was increased in 6-OHDA-treated cells. Fluprostenol, a PGF2α receptor (FP) agonist (500 nM), suppressed 6-OHDA-induced cell death by decreasing the production of reactive oxygen species (ROS) and increasing the expression of the anti-oxidant genes. These fluprostenol-mediated effects were inhibited by co-treatment with AL8810, an FP receptor antagonist (1 µM) or transfection with FP siRNA (20 nM). Moreover, 6-OHDA-induced phosphorylation of extracellular signal-regulated kinase (ERK), a member of the mitogen-activated protein kinase family, was inhibited by co-incubation with AL8810. Furthermore, fluprostenol itself enhanced ERK phosphorylation and further elevated the 6-OHDA-induced phosphorylation of ERK. In addition, 6-OHDA induced nuclear translocation of nuclear factor erythroid 2-related factor 2 (Nrf2), activating anti-oxidant gene expression, was repressed by co-culturing with AL8810. These results indicate that PGF2α suppressed 6-OHDA-induced neuronal cell death by enhancing anti-oxidant gene expression via the FP receptor-ERK-Nrf2 signaling. Thus, FP receptor is a potential target for inhibition of ROS-mediated neuronal cell death.


Asunto(s)
Dinoprost/biosíntesis , Sistema de Señalización de MAP Quinasas/fisiología , Factor 2 Relacionado con NF-E2/metabolismo , Neuroprotección/fisiología , Oxidopamina/toxicidad , Receptores de Prostaglandina/metabolismo , Adrenérgicos/toxicidad , Línea Celular Tumoral , Dinoprost/agonistas , Relación Dosis-Respuesta a Droga , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Neuroprotección/efectos de los fármacos , Prostaglandinas F Sintéticas/farmacología
14.
FASEB J ; 34(11): 15197-15207, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32985737

RESUMEN

Although it is known that prostaglandin (PG) F2α level is elevated in the plasma of patients with sepsis, the roles of PGF2α is still unknown. We aimed to clarify the roles of PGF2α in the regulation of lipopolysaccharide (LPS)-induced systemic inflammation. At 24 hours after LPS administration, neutrophil infiltration in peritoneal cavity, the mRNA expression of pro-inflammatory cytokines such as tumor necrosis factor-α, interleukin (IL)-1ß, IL-6, and macrophage inflammatory protein-2, and tissue damages in lung, liver, and kidney were all increased. Inhibition of FP receptors significantly decreased LPS-induced neutrophil infiltration and lowered the mRNA expression of the pro-inflammatory cytokines. At 6 hour after LPS administration, the level of anti-inflammatory cytokine, IL-10 in peritoneal lavage fluid was higher than that in naïve mice. Inhibition of FP receptors in these mice increased IL-10 level further. Stimulation of isolated peritoneal neutrophils by LPS increased the gene expression of IL-10, which was further increased by AL8810 treatment. Administration of an anti-IL-10 antibody antagonized the AL8810-decreased mRNA expression of pro-inflammatory cytokines and tissue damages. These results indicate that inhibition of FP receptors by AL8810 attenuated LPS-induced systemic inflammation in mice via enhanced IL-10 production.


Asunto(s)
Antiinflamatorios/farmacología , Citocinas/metabolismo , Dinoprost/análogos & derivados , Inflamación/prevención & control , Lipopolisacáridos/toxicidad , Neutrófilos/efectos de los fármacos , Receptores de Prostaglandina/antagonistas & inhibidores , Animales , Dinoprost/farmacología , Inflamación/inducido químicamente , Inflamación/metabolismo , Inflamación/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Neutrófilos/metabolismo , Neutrófilos/patología
15.
Bioorg Med Chem ; 28(14): 115512, 2020 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-32616182

RESUMEN

As a cellular bile acid sensor, farnesoid X receptor (FXR) participates in regulation of bile acid, lipid and glucose homeostasis, and liver protection. With respect to the bone metabolism, FXR positively regulates bone metabolism through both bone formation and resorption of the bone remodeling pathways. Some of FXR agonists possessing isoxazole moiety are undergoing clinical trials for the treatment of non-alcoholic steatohepatitis. To date, therefore, the activation of FXR leads to considerable interest in FXR as potential therapeutic targets. We have identified a series of nonsteroidal FXR agonists bearing N1-methyl benzimidazole and isoxazole moieties that are bridged with aromatic derivatives. They showed affinity to FXR, but also weak affinity toward the vitamin D receptor (VDR) that involves regulation of calcium and phosphate homeostasis and is activated by bile acids. The deployment of FXR agonists without activity against VDR as off-target is therefore crucial in the development of FXR ligands. Our efforts focusing on increasing the agonist properties towards FXR led to the discovery of 19, which activates FXR at and below nanomolar levels (EC50 = 26.5 ± 10.5 nM TR-FRET and 0.8 ± 0.2 nM luciferase, respectively) and functions as a FXR agonist: the affinity toward FXR over eight nuclear receptors, including VDR [IC50 (VDR) / EC50 (FXR) > 5000] and TGR5, effects FXR target genes, and activates bone morphogenetic protein-2-induced differentiation of mouse bone marrow-derived mesenchymal stem cell-like ST2 cells into osteoblast.


Asunto(s)
Bencimidazoles/farmacología , Receptores de Calcitriol/antagonistas & inhibidores , Receptores Citoplasmáticos y Nucleares/agonistas , Bencimidazoles/síntesis química , Bencimidazoles/química , Relación Dosis-Respuesta a Droga , Humanos , Estructura Molecular , Receptores de Calcitriol/metabolismo , Relación Estructura-Actividad
16.
Biochem Biophys Res Commun ; 527(1): 49-55, 2020 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-32446390

RESUMEN

Farnesoid X receptor (FXR), a bile acid receptor, is known to be involved in the promotion of adipogenesis. However, the regulation mechanism of FXR-promoted adipogenesis remains unclear. In this study, we investigated the regulation mechanism of FXR-mediated activation of adipogenesis in murine adipocyte 3T3-L1 cells. Chenodeoxycholic acid (CDCA), a potent FXR agonist, enhanced the accumulation of intracellular triglycerides and the expression of the adipogenic and lipogenic genes, while guggulsterone, an FXR antagonist, suppressed CDCA-activated adipogenesis. Moreover, troglitazone, a peroxisome proliferator-activated receptor (PPAR) γ agonist, elevated the expression of the FXR gene during adipogenesis, similar to that of the PPARγ gene. Chromatin immunoprecipitation assay demonstrated that PPARγ bound to the PPAR-responsive element of the FXR gene in a PPARγ agonist-dependent manner. Furthermore, FXR activation induced the expression of the stearoyl-CoA desaturase (SCD) gene in adipocytes. The FXR-response element (FXRE) was found in the SCD gene promoter, and FXR bound to the FXRE of the SCD gene promoter, and its binding efficiency was enhanced by CDCA in adipocytes. These results indicate that FXR assisted lipogenesis through the enhanced expression of SCD in a PPARγ-dependent manner in adipocytes, making this study the first to identify the role of FXR in the promotion of lipogenesis by PPARγ activation.


Asunto(s)
Adipocitos/metabolismo , Adipogénesis , Lipogénesis , PPAR gamma/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Células 3T3-L1 , Adipocitos/citología , Animales , Ratones , Estearoil-CoA Desaturasa/genética
17.
Artículo en Inglés | MEDLINE | ID: mdl-32036038

RESUMEN

Macrophages are the effector immune cells with plasticity to differentiate as M1 (classically activated) and M2 (alternatively activated) phenotypes. Prostaglandins (PGs) have various important roles and are involved in the regulation of macrophage activation. However, the role of PGF2α in macrophage activation remains unclear. We investigated the role of PGF2α receptor (FP)-mediated signaling in the M1 macrophage polarization using murine macrophage RAW264.7 cells. Stimulation with lipopolysaccharide (LPS) + interferon (IFN)-γ increased the mRNA expression of the M1 macrophage markers such as inducible nitric oxide synthase, tumor necrosis factor-α, and CD11c. Pre-treatment with AL8810, an FP receptor antagonist, further enhanced the expression of these genes. In contrast, treatment with fluprostenol, an FP receptor agonist, decreased the LPS + IFN-γ-induced expression of M1 markers. LPS-induced M1 macrophage polarization was dependent on the activation of NF-κB p65. Treatment with IκB kinase ß inhibitor reduced AL8810-induced mRNA expression of the M1 markers. Stimulation with LPS + IFN-γ increased the expression of IL-10. Pre-treatment with AL8810 lowered LPS + IFN-γ-induced IL-10 expression, and further enhanced LPS + IFN-γ-stimulated nuclear translocation of NF-κB p65. In contrast, co-treatment with IL-10 reversed AL8810-induced nuclear translocation of NF-κB p65. These results indicate that the FP receptor signaling was involved in the control of M1 polarization of macrophages via IL-10-regulated nuclear translocation of NF-κB p65.


Asunto(s)
Interleucina-10/metabolismo , Activación de Macrófagos/inmunología , Macrófagos/inmunología , Receptores de Prostaglandina/metabolismo , Factor de Transcripción ReIA/metabolismo , Animales , Núcleo Celular/metabolismo , Dinoprost/análogos & derivados , Dinoprost/farmacología , Quinasa I-kappa B/antagonistas & inhibidores , Quinasa I-kappa B/metabolismo , Interferón gamma/inmunología , Lipopolisacáridos/inmunología , Activación de Macrófagos/efectos de los fármacos , Macrófagos/citología , Macrófagos/metabolismo , Ratones , Prostaglandinas F Sintéticas/farmacología , Células RAW 264.7 , Receptores de Prostaglandina/agonistas , Receptores de Prostaglandina/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Tiofenos/farmacología
18.
J Biochem ; 167(6): 623-631, 2020 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-31960927

RESUMEN

Macrophages, which develop by changing their functions according to various environmental conditions and stimuli, defend against the pathogens and play roles in homoeostasis and disease states. Bicarbonate (HCO3-) is important in the maintenance of intracellular and extracellular pH in the body. However, the effects of bicarbonate on macrophage function have not been examined. In this study, we investigated the effects of bicarbonate on macrophage activation in lipopolysaccharide (LPS) and interferon (IFN)-γ (LPS + IFN-γ)-stimulated murine macrophage-like RAW264.7 cells. The expression of the interleukin (IL)-6, inducible nitric oxide (NO) synthase and cyclooxygenase-2 genes was enhanced by sodium bicarbonate (NaHCO3) in a concentration-dependent manner in LPS + IFN-γ-stimulated RAW264.7 cells. The production of IL-6, NO2- and prostaglandin E2 was also increased by treatment with NaHCO3 in these cells. Moreover, NaHCO3-mediated elevation of inflammatory gene expression was abrogated by solute carrier (SLC) transporter inhibitors. Furthermore, its NaHCO3-mediated activation was negated by a JAK inhibitor , tofacitinib. NaHCO3-enhanced phosphorylation of STAT1, and its enhancement was abrogated by pre-treating with SLC transporter inhibitors in LPS + IFN-γ-stimulated RAW264.7 cells. In addition, similar results were obtained in murine bone marrow-derived macrophages. These results indicate that bicarbonate enhanced the inflammatory response through the JAK/STAT signalling in LPS + IFN-γ-stimulated macrophages.


Asunto(s)
Interferón gamma/farmacología , Quinasas Janus/metabolismo , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Factor de Transcripción STAT1/metabolismo , Transducción de Señal/efectos de los fármacos , Bicarbonato de Sodio/farmacología , Animales , Ciclooxigenasa 2/genética , Expresión Génica/efectos de los fármacos , Inflamación/inducido químicamente , Inflamación/genética , Interleucina-6/genética , Inhibidores de las Cinasas Janus/farmacología , Quinasas Janus/antagonistas & inhibidores , Activación de Macrófagos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Óxido Nítrico/genética , Piperidinas/farmacología , Pirimidinas/farmacología , Células RAW 264.7 , Proteínas Recombinantes/farmacología
19.
Molecules ; 24(22)2019 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-31744088

RESUMEN

The modulators of farnesoid X receptor (FXR), a bile acid receptor, regulate various biological processes including bile acid metabolism, and are associated with the control of fatty liver and osteoporosis. Thus, the control of FXR activity and development of FXR modulators are critical not only for research, but also for clinical application. In this study, we synthesized novel FXR agonists 1-4 possessing isoxazole and N-substituted benzimidazole moieties, and compared their effects on osteoblast differentiation with the known FXR agonists, chenodeoxycholic acid and a synthetic compound, GW4064. Two (3 and 4) of the four novel FXR agonists 1-4 showed high specificities for FXR. Computer-assisted modeling suggested that the binding of the FXR agonist 3 with ligand binding domain of FXR was similar to GW4064. FXR was expressed in mouse bone marrow-derived mesenchymal stem cell (MSC)-like ST2 cells (ST-2 MSCs). The FXR agonists activated the BMP-2-induced differentiation of ST-2 MSCs into osteoblasts and enhanced the expression of RUNX2. Moreover, the potency of the FXR agonist 3 was comparable to GW4064 in promoting osteoblast differentiation of ST-2 MSCs. These results indicate that FXR activation enhanced the BMP-2-induced differentiation of MSCs into osteoblasts through activating RUNX2 expression. FXR could be a potential therapeutic target for the treatment of bone diseases such as osteoporosis.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Isoxazoles/síntesis química , Isoxazoles/farmacología , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de los fármacos , Osteoblastos/citología , Receptores Citoplasmáticos y Nucleares/agonistas , Animales , Supervivencia Celular/efectos de los fármacos , Técnicas de Química Sintética , Fluoroinmunoensayo , Genes Reporteros , Humanos , Isoxazoles/química , Ratones , Modelos Moleculares , Receptores Citoplasmáticos y Nucleares/química , Relación Estructura-Actividad
20.
J Agric Food Chem ; 67(36): 10107-10115, 2019 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-31434473

RESUMEN

We examined the antiobesity effect of a limonoid 7-deacetoxy-7-oxogedunin, named CG-1, purified from the seeds of Carapa guianensis, Meliaceae, known as andiroba in high-fat-diet (HFD)-fed mice. C57BL/6 mice were fed a low-fat diet or an HFD and orally administered CG-1 (20 mg/kg) for 7 weeks. CG-1 lowered the body weight gain and improved the serum triglyceride level and insulin sensitivity in HFD-fed mice. The expression level of the adipogenesis-related genes was lowered by CG-1 in the visceral white adipose tissue (vWAT). The mRNA expression level of the macrophage-related genes decreased in vWAT following the administration of CG-1 to HFD-fed mice. It is noteworthy that CG-1 activated the brown adipose tissue (BAT) with enhanced expression of uncoupling protein 1 and increased the rectal temperature in HFD-fed mice. These results indicate that the limonoid CG-1 decreased body weight gain and ameliorated hypertriglyceridemia and insulin resistance with the activation of BAT in HFD-fed mice.


Asunto(s)
Tejido Adiposo Pardo/efectos de los fármacos , Fármacos Antiobesidad/administración & dosificación , Resistencia a la Insulina , Limoninas/administración & dosificación , Meliaceae/química , Obesidad/tratamiento farmacológico , Extractos Vegetales/administración & dosificación , Tejido Adiposo Pardo/metabolismo , Animales , Dieta Alta en Grasa/efectos adversos , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/genética , Obesidad/metabolismo , Obesidad/fisiopatología , Semillas/química , Triglicéridos/sangre , Proteína Desacopladora 1/genética , Proteína Desacopladora 1/metabolismo , Aumento de Peso/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA