Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
J Bioenerg Biomembr ; 55(3): 169-177, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37219648

RESUMEN

The tumor microenvironment (TME) consists of various cell types, including fibroblasts. The TME plays a central role in the promotion of tumor progression. In the present study, we investigated whether lysophosphatidic acid (LPA) receptor-mediated signaling regulates cellular functions by the TME of pancreatic cancer PANC-1 cells. To obtain fibroblast 3T3 cell supernatants, 3T3 cells were cultured in 5% charcoal stripped FCS-DMEM for 48 h. LPAR2 and LPAR3 expression levels were elevated in PANC-1 cells cultured in 3T3 cell supernatants. While PANC-1 cell motility was decreased by 3T3 cell supernatants, the cell survival to cisplatin (CDDP) of PANC-1 cells was markedly enhanced. Moreover, the cell survival to CDDP of PANC-1 cells cultured in 3T3 cell supernatants was increased by GRI-977,143 (LPA2 agonist) and (2 S)-OMPT (LPA3 agonist). Since hypoxia is caused by the restriction of adequate vascular networks to deliver oxygen into solid tumors, PANC-1 cells were cultured in 3T3 cell supernatants at 1% O2 conditions. The cell survival to CDDP of PANC-1 cells cultured in 3T3 cell supernatants at 1% O2 was significantly elevated, correlating with LPAR2 and LPAR3 expressions. These results suggest that LPA signaling via LPA2 and LPA3 is involved in the promotion of malignant properties by the TME in PANC-1 cells.


Asunto(s)
Neoplasias Pancreáticas , Receptores del Ácido Lisofosfatídico , Ratones , Animales , Humanos , Receptores del Ácido Lisofosfatídico/metabolismo , Lisofosfolípidos/metabolismo , Lisofosfolípidos/farmacología , Cisplatino/farmacología , Fibroblastos/metabolismo , Fibroblastos/patología , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Movimiento Celular , Hipoxia/metabolismo , Microambiente Tumoral , Neoplasias Pancreáticas
2.
J Bioenerg Biomembr ; 54(2): 109-117, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35260987

RESUMEN

Lysophosphatidic acid (LPA) signaling via LPA receptors (LPA1 to LPA6) exhibits a variety of malignant properties in cancer cells. Intracellular ATP depletion leads to the development of necrosis and apoptosis. The present study aimed to evaluate the effects of LPA receptor-mediated signaling on the regulation of cancer cell functions associated with ATP reduction. Long-term ethidium bromide (EtBr) treated (MG63-EtBr) cells were established from osteosarcoma MG-63 cells. The intracellular ATP levels of MG63-EtBr cells were significantly lower than that of MG-63 cells. LPAR2, LPAR3, LPAR4 and LPAR6 gene expressions were elevated in MG63-EtBr cells. The cell motile and invasive activities of MG63-EtBr cells were markedly higher than those of MG-63 cells. The cell motile activity of MG-63 cells was increased by LPA4 and LPA6 knockdowns. In cell survival assay, cells were treated with cisplatin (CDDP) every 24 h for 3 days. The cell survival to CDDP of MG63-EtBr cells was lower than that of MG-63 cells. LPA2 knockdown decreased the cell survival to CDDP of MG-63 cells. The cell survival to CDDP of MG-63 cells was inhibited by (2 S)-OMPT (LPA3 agonist). Moreover, the cell survival to CDDP of MG-63 cells was enhanced by LPA4 and LPA6 knockdowns. These results indicate that LPA signaling via LPA receptors is involved in the regulation of cellular functions associated with ATP reduction in MG-63 cells treated with EtBr.


Asunto(s)
Neoplasias Óseas , Osteosarcoma , Adenosina Trifosfato/farmacología , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/genética , Movimiento Celular , Etidio/farmacología , Regulación Neoplásica de la Expresión Génica , Humanos , Lisofosfolípidos/metabolismo , Osteosarcoma/tratamiento farmacológico , Osteosarcoma/genética , Receptores del Ácido Lisofosfatídico/genética , Receptores del Ácido Lisofosfatídico/metabolismo
3.
Anal Biochem ; 630: 114322, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34343482

RESUMEN

Autotaxin (ATX) plays an important role in (patho-)physiological lysophosphatidic acid (LPA) signaling. Here we describe the establishment of novel cell-based ATX assay formats. ATX-mediated LPA generation is detected by using a stable LPA receptor reporter cell line. In a first assay variant, ATX-mediated LPA generation is started in the absence of cells and the reaction mix is transferred to the reporter cells after stopping the reaction (two-tube assay). In a second assay variant, ATX is added to the reporter cells expressing the known autotaxin binding partners integrin ß1, integrin ß3 and the LPA receptor 1. LPA generation is started in the presence of cells and is detected in real-time (one-tube assay). Structurally diverse ATX inhibitors with different binding modes were characterized in both cell-based assay variants and were also tested in the well-established biochemical choline release assay. ATX inhibitors displayed similar potencies, regardless if the assay was performed in the absence or presence of cells, and comparable results were obtained in all three assay formats. In summary, our novel cell-based ATX assay formats are well-suited for sensitive detection of enzyme activity as well as for the characterization of ATX inhibitors in the presence and absence of cells.


Asunto(s)
Hidrolasas Diéster Fosfóricas/análisis , Células Cultivadas , Humanos , Lisofosfolípidos/química , Lisofosfolípidos/metabolismo , Modelos Moleculares , Hidrolasas Diéster Fosfóricas/metabolismo
4.
Int J Mol Sci ; 22(17)2021 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-34502491

RESUMEN

Besides serving as a structural membrane component and intermediate of the glycerolipid metabolism, lysophosphatidic acid (LPA) has a prominent role as a signaling molecule through its binding to LPA receptors at the cell surface. Extracellular LPA is primarily produced from lysophosphatidylcholine (LPC) through the activity of secreted lysophospholipase D, autotaxin (ATX). The degradation of extracellular LPA to monoacylglycerol is mediated by lipid phosphate phosphatases (LPPs) at the cell membrane. This review summarizes and interprets current literature on the role of the ATX-LPA-LPP3 axis in the regulation of energy homeostasis, insulin function, and adiposity at baseline and under conditions of obesity. We also discuss how the ATX-LPA-LPP3 axis influences obesity-related metabolic complications, including insulin resistance, fatty liver disease, and cardiomyopathy.


Asunto(s)
Metabolismo Energético , Lisofosfolípidos/metabolismo , Enfermedades Metabólicas/metabolismo , Fosfatidato Fosfatasa/metabolismo , Hidrolasas Diéster Fosfóricas/metabolismo , Transducción de Señal , Animales , Humanos
5.
Biochem Biophys Res Commun ; 503(4): 2698-2703, 2018 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-30093116

RESUMEN

Lysophosphatidic acid (LPA) receptors (LPA1 to LPA6) regulate a variety of malignant properties in cancer cells. In the present study, we investigated the roles of LPA receptors in the promotion of cellular functions during tumor progression in fibrosarcoma cells. To obtain long-term anticancer drug treated cells, human fibrosarcoma HT1080 cells were treated with methotrexate (MTX) and cisplatin (CDDP) for 6 months. LPAR2 and LPAR5 expressions were significantly higher in MTX-treated (HT-MTX) cells than in HT1080 cells. The cell motile and invasive activities of HT-MTX cells were significantly elevated compared with HT1080 cells. Although LPAR5 expression was increased in MTX and CDDP treated (HT-M-C) cells, no change of LPAR2 expression was observed. The cell motile and invasive activities of HT-M-C cells were lower than those of HT1080 cells. Moreover, to evaluate whether LPA receptors promote cell invasive activity, highly invasion (HT1080-M6) cells were established from HT1080 cells. The cell invasive activity of HT1080-M6 cells was approximately 4.5 times higher than HT1080 cell invasion. LPAR2 expression was markedly elevated in HT1080-M6 cells compared with HT1080 cells. The high cell invasion activity of HT1080-M6 cells was significantly suppressed by an antagonist of LPA2, H2L5186303. These results suggest that LPA2 acts as a key regulator of malignant properties in HT1080 cells.


Asunto(s)
Fibroblastos/metabolismo , Regulación Neoplásica de la Expresión Génica , Receptores del Ácido Lisofosfatídico/genética , Antineoplásicos/farmacología , Derivados del Benceno/farmacología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Cisplatino/farmacología , Resistencia a Antineoplásicos/genética , Fibroblastos/efectos de los fármacos , Fibroblastos/patología , Humanos , Metotrexato/farmacología , Receptores del Ácido Lisofosfatídico/antagonistas & inhibidores , Receptores del Ácido Lisofosfatídico/metabolismo , Transducción de Señal
6.
J Recept Signal Transduct Res ; 38(4): 367-371, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-30396320

RESUMEN

Lysophosphatidic acid (LPA) is a simple biological lipid and mediates several biological functions with LPA receptors (LPA1 to LPA6). In the present study, to assess whether LPA receptors promote cell-invasive activity of pancreatic cancer cells, highly invasion PANC-R9 cells were established from PANC-1 cells, using Matrigel-coated Cell Culture Insert. The cell-invasive activity of PANC-R9 cells was shown to be approximately 15 times higher than that of PANC-1 cells. LPAR1 expression level was markedly elevated in PANC-R9 cells in comparison with PANC-1 cells, while LPAR3 expression level was reduced. The cell-invasive activity of PANC-R9 cells was enhanced by LPA, but LPA had no impact on PANC-1 cell invasion. Before initiation of the cell invasion assay, PANC-R9 cells were pretreated with dioctanoylglycerol pyrophosphate (DGPP), an antagonist of LPA1/LPA3. The invasive activity of PANC-R9 cells was markedly suppressed by DGPP. Autotaxin (ATX) is a key enzyme that catalyzes the conversion of lysophosphatidylcholine (LPC) to LPA. ATX expression level was elevated in PANC-R9 cells compared with PANC-1 cells. In the presence of LPC, the cell motile activity of PANC-R9 cells was markedly stimulated. In contrast, LPC did not affect the cell motile activity of PANC-1 cells. PANC-R9 cell motility was inhibited by an ATX inhibitor, PF-8380. These results suggest that LPA signaling via LPA1 is a potent molecular target for the regulation of tumor progression in PANC-1 cells.


Asunto(s)
Lisofosfatidilcolinas/metabolismo , Neoplasias Pancreáticas/tratamiento farmacológico , Hidrolasas Diéster Fosfóricas/genética , Receptores del Ácido Lisofosfatídico/genética , Benzoxazoles/farmacología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica , Humanos , Lisofosfatidilcolinas/genética , Invasividad Neoplásica/genética , Neoplasias Pancreáticas/genética , Ácidos Fosfatidicos/metabolismo , Piperazinas/farmacología , Receptores del Ácido Lisofosfatídico/metabolismo , Transducción de Señal/efectos de los fármacos
7.
J Recept Signal Transduct Res ; 38(1): 71-75, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29369010

RESUMEN

Lysophosphatidic acid (LPA) is a simple physiological lipid and exhibits a variety of cellular responses via the activation of G protein-coupled transmembrane LPA receptors (LPA receptor-1 (LPA1) to LPA6). The aim of our study was to investigate effects of LPA receptors on soft agar colony formation in colon cancer cells treated with anticancer drugs. DLD1 cells were treated with fluorouracil (5-FU) or cisplatin (CDDP) for at least six months (DLD-5FU and DLD-CDDP cells, respectively). LPAR1 gene expression was markedly elevated in DLD-5FU cells. In contrast, DLD-CDDP cells showed the high expression of LPAR6 gene. In colony formation assay, DLD-5FU cells formed markedly large-sized colonies, while no colony formation was observed in DLD1 and DLD-CDDP cells. The large-sized colonies formed in DLD-5FU cells were suppressed by LPA1 knockdown. In contrast, LPA6 knockdown increased the size of colonies. In addition, DLD-5FU cells were further treated with CDDP for three months (DLD-C-F cells). DLD-CDDP cells were also treated with 5-FU (DLD-F-C cells). DLD-C-F cells formed large-sized colonies, but not DLD-F-C cells, correlating with LPAR1 and LPAR6 gene expression levels. These results suggest that LPA1 and LPA6 may regulate the colony formation activity in DLD1 cells treated with anticancer drugs.


Asunto(s)
Neoplasias del Colon/tratamiento farmacológico , Receptores del Ácido Lisofosfatídico/genética , Células Madre/efectos de los fármacos , Antineoplásicos/administración & dosificación , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Cisplatino/administración & dosificación , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Fluorouracilo/administración & dosificación , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Lisofosfolípidos/genética , Lisofosfolípidos/metabolismo
8.
Exp Cell Res ; 352(1): 139-145, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28189636

RESUMEN

Lysophosphatidic acid (LPA) signaling via G protein-coupled LPA receptors exhibits a variety of biological effects, such as cell proliferation, motility and differentiation. The aim of this study was to evaluate the roles of LPA1 and LPA3 in cellular functions during tumor progression in pancreatic cancer cells. LPA1 and LPA3 knockdown cells were generated from PANC-1 cells. The cell motile and invasive activities of PANC-1 cells were inhibited by LPA1 and LPA3 knockdown. In gelatin zymography, LPA1 and LPA3 knockdown cells indicated the low activation of matrix metalloproteinase-2 (MMP-2) in the presence of LPA. Next, to assess whether LPA1 and LPA3 regulate cellular functions induced by anticancer drug, PANC-1 cells were treated with cisplatin (CDDP) for approximately 6 months. The cell motile and invasive activities of long-term CDDP treated cells were markedly higher than those of PANC-1 cells, correlating with the expression levels of LPAR1 and LPAR3 genes. In soft agar assay, the long-term CDDP treated cells formed markedly large sized colonies. In addition, the cell motile and invasive activities enhanced by CDDP were significantly suppressed by LPA1 and LPA3 knockdown as well as colony formation. These results suggest that LPA signaling via LPA1 and LPA3 play an important role in the regulation of cellular functions during tumor progression in PANC-1 cells.


Asunto(s)
Lisofosfolípidos/farmacología , Neoplasias Pancreáticas/patología , Receptores del Ácido Lisofosfatídico/metabolismo , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Western Blotting , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Cisplatino/farmacología , Progresión de la Enfermedad , Quimioterapia Combinada , Humanos , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 2 de la Matriz/metabolismo , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/metabolismo , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores del Ácido Lisofosfatídico/antagonistas & inhibidores , Receptores del Ácido Lisofosfatídico/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas
9.
J Cell Sci ; 128(21): 3871-7, 2015 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-26345369

RESUMEN

Lysophosphatidic acid (LPA) is emerging as an angiogenic factor, because knockdown of the enzyme that produces it (autotaxin, also known as ENPP2) and its receptors cause severe developmental vascular defects in both mice and fish. In addition, overexpression of autotaxin in mice causes similar vascular defects, indicating that the extracellular amount of LPA must be tightly regulated. Here, we focused on an LPA-degrading enzyme, lipid phosphate phosphatase 3 (LPP3, also known as PPAP2B), and showed that LPP3 was localized in specific cell-cell contact sites of endothelial cells and suppresses LPA signalling through the LPA6 receptor (also known as LPAR6). In HEK293 cells, overexpression of LPP3 dramatically suppressed activation of LPA6. In human umbilical vein endothelial cells (HUVECs), LPA induced actin stress fibre formation through LPA6, which was substantially upregulated by LPP3 knockdown. LPP3 was localized to cell-cell contact sites and was missing in non-contact sites to which LPA-induced actin stress fibre formation mediated by LPA6 was restricted. Interestingly, the expression of LPP3 in HUVECs was dramatically increased after forskolin treatment in a process involving Notch signalling. These results indicate that LPP3 regulates and localizes LPA signalling in endothelial cells, thereby stabilizing vessels through Notch signalling for proper vasculature.


Asunto(s)
Lisofosfolípidos/metabolismo , Fosfatidato Fosfatasa/metabolismo , Animales , Línea Celular , Colforsina/farmacología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones , Ratones Noqueados , Fosfatidato Fosfatasa/genética , Receptores Notch/genética , Receptores Notch/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
10.
Biochem Biophys Res Commun ; 483(1): 652-657, 2017 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-27993681

RESUMEN

Lysophosphatidic acid (LPA) is an extracellular biological lipid and interacts with six subtypes of G protein-coupled LPA receptors (LPA1 to LPA6). LPA receptors exhibit a variety of cellular functions, depending on types of cancer cells. In this study, to assess the roles of LPA4 and LPA6 in cell growth and motile activities of colon cancer cells, LPA4 and LPA6 knockdown cells were established from DLD1 and HCT116 cells. LPA treatment increased the cell growth activities of LPA4 and LPA6 knockdown cells, compared with control cells. The cell motile activities of LPA4 and LPA6 knockdown cells were significantly higher than those of control cells. To evaluate the effects of LPA4 and LPA6 on cell motile activity induced by anticancer drug, long-term fluorouracil (5-FU) treated (DLD-5FU) cells were generated. The expression levels of LPAR1, LPAR4 and LPAR6 genes were significantly increased in DLD-5FU cells. DLD-5FU cells showed the high cell motile activity, compared with DLD1 cells. The increased cell motile activity was markedly stimulated by LPA4 and LPA6 knockdown. In contrast, the cell motile activity enhanced by 5-FU treatment was suppressed by LPA1 knockdown. These results suggest that LPA signaling via LPA4 and LPA6 negatively regulates the cell motile activities of DLD1 and HCT116 cells as well as long-term 5-FU treated cells.


Asunto(s)
Neoplasias del Colon/patología , Lisofosfolípidos/metabolismo , Receptores del Ácido Lisofosfatídico/metabolismo , Receptores Purinérgicos/metabolismo , Antimetabolitos Antineoplásicos/farmacología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Neoplasias del Colon/metabolismo , Fluorouracilo/farmacología , Células HCT116 , Humanos , Lisofosfolípidos/farmacología , Receptores Purinérgicos P2 , Transducción de Señal
11.
Biochim Biophys Acta ; 1851(2): 172-83, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25463482

RESUMEN

Erythrocytes and megakaryocytes (MK) are derived from a common progenitor that undergoes lineage specification. Lysophosphatidic acid (LPA), a lipid growth factor was previously shown to be a regulator for erythropoietic process through activating LPA receptor 3 (LPA3). However, whether LPA affects megakaryopoiesis remains unclear. In this study, we used K562 leukemia cell line as a model to investigate the roles of LPA in MK differentiation. We demonstrated that K562 cells express both LPA2 and LPA3, and the expression levels of LPA2 are higher than LPA3. Treatment with phorbol 12-myristate 13-acetate, a commonly used inducer of megakaryopoiesis, reciprocally regulates the expressions of LPA2 and LPA3. By pharmacological blockers and knockdown experiments, we showed that activation of LPA2 suppresses whereas, LPA3 promotes megakaryocytic differentiation in K562. The LPA2-mediated inhibition is dependent on ß-catenin translocation, whereas reactive oxygen species (ROS) generation is a downstream signal for activation of LPA3. Furthermore, the hematopoietic transcriptional factors GATA-1 and FLI-1, appear to be involved in these regulatory mechanisms. Taken together, our results suggested that LPA2 and LPA3 may function as a molecular switch and play opposing roles during megakaryopoiesis of K562 cells.


Asunto(s)
Leucemia Eritroblástica Aguda/metabolismo , Megacariocitos/metabolismo , Receptores del Ácido Lisofosfatídico/metabolismo , Trombopoyesis , Factor de Transcripción GATA1/metabolismo , Humanos , Integrina beta3/metabolismo , Células K562 , Leucemia Eritroblástica Aguda/genética , Megacariocitos/efectos de los fármacos , Proteínas de Microfilamentos/metabolismo , Interferencia de ARN , Especies Reactivas de Oxígeno/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores del Ácido Lisofosfatídico/antagonistas & inhibidores , Receptores del Ácido Lisofosfatídico/genética , Transducción de Señal , Acetato de Tetradecanoilforbol/farmacología , Trombopoyesis/efectos de los fármacos , Factores de Tiempo , Transactivadores , Transfección , beta Catenina/metabolismo
12.
Mol Carcinog ; 55(11): 1573-1583, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26377854

RESUMEN

Lysophosphatidic acid (LPA) signaling via LPA receptors (LPA1 to LPA6 ) mediates a variety of cellular functions, including cell motility. In the present study, we investigated the effects of LPA receptors on cell motile activity during multi-stage hepatocarcinogenesis in rat liver epithelial WB-F344 cells treated with chemical liver carcinogens. Cells were treated with a initiator (N-nitrosodiethylamine (DEN)) and three promoters (phenobarbital (PB), okadaic acid (OA) and clofibrate) every 24 h for 2 days. Cell motile activity was elevated by DEN, correlating with Lpar3 expression. PB, OA, and clofibrate elevated Lpar1 expression and inhibited cell motile activity. To evaluate the effects of long-term treatment on cell motility, cells were treated with DEN and/or PB for at least 6 months. Lpar3 expression and cell motile activity were significantly elevated by the long-term DEN treatment with or without further PB treatment. In contrast, long-term PB treatment with or without further DEN elevated Lpar1 expression and inhibited cell motility. When the synthesis of extracellular LPA was blocked by a potent ATX inhibitor S32826 before cell motility assay, the cell motility induced by DEN and PB was markedly suppressed. These results suggest that activation of the different LPA receptors may regulate the biological functions of cells treated with chemical carcinogens. © 2015 Wiley Periodicals, Inc.


Asunto(s)
Carcinógenos/farmacología , Dietilnitrosamina/efectos adversos , Células Epiteliales/efectos de los fármacos , Neoplasias Hepáticas Experimentales/metabolismo , Receptores del Ácido Lisofosfatídico/metabolismo , Animales , Línea Celular , Movimiento Celular/efectos de los fármacos , Clofibrato/efectos adversos , Clofibrato/farmacología , Células Epiteliales/citología , Regulación de la Expresión Génica/efectos de los fármacos , Neoplasias Hepáticas Experimentales/inducido químicamente , Ácido Ocadaico/efectos adversos , Ácido Ocadaico/farmacología , Fenobarbital/efectos adversos , Fenobarbital/farmacología , Ratas , Ratas Endogámicas F344
13.
J Pathol ; 235(3): 456-65, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25294670

RESUMEN

Undifferentiated nasopharyngeal carcinoma (NPC) is a highly metastatic disease that is consistently associated with Epstein-Barr virus (EBV) infection. In this study, we have investigated the contribution of lysophosphatidic acid (LPA) signalling to the pathogenesis of NPC. Here we demonstrate two distinct functional roles for LPA in NPC. First, we show that LPA enhances the migration of NPC cells and second, that it can inhibit the activity of EBV-specific cytotoxic T cells. Focusing on the first of these phenotypes, we show that one of the LPA receptors, LPA receptor 5 (LPAR5), is down-regulated in primary NPC tissues and that this down-regulation promotes the LPA-induced migration of NPC cell lines. Furthermore, we found that EBV infection or ectopic expression of the EBV-encoded LMP2A was sufficient to down-regulate LPAR5 in NPC cell lines. Our data point to a central role for EBV in mediating the oncogenic effects of LPA in NPC and identify LPA signalling as a potential therapeutic target in this disease.


Asunto(s)
Regulación hacia Abajo/fisiología , Infecciones por Virus de Epstein-Barr/fisiopatología , Regulación Neoplásica de la Expresión Génica/fisiología , Lisofosfolípidos/fisiología , Neoplasias Nasofaríngeas/fisiopatología , Receptores del Ácido Lisofosfatídico/fisiología , Transducción de Señal/fisiología , Adenocarcinoma/patología , Adenocarcinoma/fisiopatología , Carcinoma , Línea Celular Tumoral , Movimiento Celular/fisiología , Herpesvirus Humano 4/fisiología , Humanos , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/patología , Hidrolasas Diéster Fosfóricas/fisiología , Receptores del Ácido Lisofosfatídico/genética , Linfocitos T Citotóxicos/patología , Proteínas de la Matriz Viral/fisiología
14.
Korean J Physiol Pharmacol ; 20(6): 629-639, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27847440

RESUMEN

The present study was designed to investigate the characteristics of gintonin, one of components isolated from Korean Ginseng on secretion of catecholamines (CA) from the isolated perfused model of rat adrenal gland and to clarify its mechanism of action. Gintonin (1 to 30 µg/ml), perfused into an adrenal vein, markedly increased the CA secretion from the perfused rat adrenal medulla in a dose-dependent fashion. The gintonin-evoked CA secretion was greatly inhibited in the presence of chlorisondamine (1 µM, an autonomic ganglionic bloker), pirenzepine (2 µM, a muscarinic M1 receptor antagonist), Ki14625 (10 µM, an LPA1/3 receptor antagonist), amiloride (1 mM, an inhibitor of Na+/Ca2+ exchanger), a nicardipine (1 µM, a voltage-dependent Ca2+ channel blocker), TMB-8 (1 µM, an intracellular Ca2+ antagonist), and perfusion of Ca2+-free Krebs solution with 5mM EGTA (a Ca2+chelater), while was not affected by sodium nitroprusside (100 µM, a nitrosovasodialtor). Interestingly, LPA (0.3~3 µM, an LPA receptor agonist) also dose-dependently enhanced the CA secretion from the adrenal medulla, but this facilitatory effect of LPA was greatly inhibited in the presence of Ki 14625 (10 µM). Moreover, acetylcholine (AC)-evoked CA secretion was greatly potentiated during the perfusion of gintonin (3 µg/ml). Taken together, these results demonstrate the first evidence that gintonin increases the CA secretion from the perfused rat adrenal medulla in a dose-dependent fashion. This facilitatory effect of gintonin seems to be associated with activation of LPA- and cholinergic-receptors, which are relevant to the cytoplasmic Ca2+ increase by stimulation of the Ca2+ influx as well as by the inhibition of Ca2+ uptake into the cytoplasmic Ca2+ stores, without the increased nitric oxide (NO). Based on these results, it is thought that gintonin, one of ginseng components, can elevate the CA secretion from adrenal medulla by regulating the Ca2+ mobilization for exocytosis, suggesting facilitation of cardiovascular system. Also, these findings show that gintonin might be at least one of ginseng-induced hypertensive components.

15.
Acta Crystallogr D Biol Crystallogr ; 71(Pt 5): 1039-50, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25945569

RESUMEN

Lysophosphatidic acid (LPA) is a phospholipid growth factor with myriad effects on biological systems. LPA is usually present bound to animal plasma proteins such as albumin or gelsolin. When LPA complexes with plasma proteins, it binds to its cognate receptors with higher affinity than when it is free. Recently, gintonin from ginseng was found to bind to LPA and to activate mammalian LPA receptors. Gintonin contains two components: ginseng major latex-like protein 151 (GLP) and ginseng ribonuclease-like storage protein. Here, the crystal structure of GLP is reported, which belongs to the plant Bet v 1 superfamily, and a model is proposed for how GLP binds LPA. Amino-acid residues of GLP recognizing LPA were identified using site-directed mutagenesis and isothermal titration calorimetry. The resulting GLP mutants were used to study the activation of LPA receptor-dependent signalling pathways. In contrast to wild-type GLP, the H147A mutant did not bind LPA, elicit intracellular Ca(2+) transients in neuronal cells or activate Ca(2+)-dependent Cl(-) channels in Xenopus oocytes. Based on these results, a mechanism by which GLP recognizes LPA and its requirement to activate G protein-coupled LPA receptors to elicit diverse biological responses were proposed.


Asunto(s)
Embrión de Mamíferos/metabolismo , Hipocampo/metabolismo , Lisofosfolípidos/metabolismo , Oocitos/metabolismo , Proteínas de Plantas/química , Proteínas de Plantas/metabolismo , Secuencia de Aminoácidos , Animales , Calcio/metabolismo , Células Cultivadas , Electrofisiología , Embrión de Mamíferos/citología , Femenino , Hipocampo/citología , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Mutación/genética , Oocitos/citología , Proteínas de Plantas/genética , Conformación Proteica , Homología de Secuencia de Aminoácido , Transducción de Señal , Xenopus laevis/crecimiento & desarrollo , Xenopus laevis/metabolismo
16.
Biochem Biophys Res Commun ; 461(1): 59-64, 2015 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-25849892

RESUMEN

Lysophosphatidic acid (LPA) is an extracellular biological lipid which interacts with G protein-coupled LPA receptors (LPA1 to LPA6). LPA signaling via LPA receptors mediates several cellular responses. In the present study, to assess the roles of LPA4, LPA5 and LPA6 in cellular functions of pancreatic cancer cells, we generated LPA receptor knockdown cells from PANC-1 cells (PANC-sh4, PANC-sh5 and PANC-sh6 cells, respectively). In cell motility assay, PANC-sh4 and PANC-sh5 cells enhanced the cell motile activities, compared with control cells. In contrast, the cell motile activity of PANC-sh6 cells was suppressed. The invasive activities of PANC-sh4 and PANC-sh5 cells were markedly stimulated, while PANC-sh6 cells showed the low invasive activity. In colony assay, PANC-sh4 and PANC-sh5 cells formed the large sized colonies, but not PANC-sh6 cells. When endothelial cells were incubated with supernatants from PANC-sh4 and PANC-sh5 cells, the cell motility and tube formation of endothelial cells were significantly induced, but not PANC-sh6 cells. These results suggest that the diverse roles of LPA4, LPA5 and LPA6 are involved in the activation of tumor progression in pancreatic cancer cells.


Asunto(s)
Lisofosfolípidos/metabolismo , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Receptores del Ácido Lisofosfatídico/metabolismo , Receptores Purinérgicos P2/metabolismo , Línea Celular Tumoral , Movimiento Celular , Humanos , Invasividad Neoplásica , Receptores Purinérgicos
17.
Mol Cell Biochem ; 408(1-2): 147-54, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26116018

RESUMEN

Lysophosphatidic acid (LPA) signaling via LPA receptors provides a variety of cellular functions, including angiogenesis. In this study, to assess an involvement of LPA receptors in cell motile activities of endothelial cells during chemotherapy, F-2 cells were treated with cisplatin (CDDP) and doxorubicin (DOX) at a concentration of 0.01 µM every 24 h for at least 1 month. The treatment of CDDP and DOX inhibited the expression levels of the LPA receptor-1 (Lpar1), Lpar2, and Lpar3 genes in F-2 cells. The cell motile activities of CDDP and DOX treated cells were relatively lower than those of untreated cells. Next, we investigated whether cancer cells could stimulate the cell motile activities of F-2 cells treated with CDDP and DOX. For cell motility assay, CDDP- and DOX-treated cells were co-cultured with pancreatic cancer PANC-1 cells. The cell motile activities of CDDP- and DOX-treated cells were significantly enhanced by the existence of PANC-1 cells, correlating with the LPA receptor expressions. In addition, the elevated cell motile activities were suppressed by the pretreatment of an autotaxin inhibitor S32826. These results suggest that LPA signaling via LPA receptors may regulate the cell motile activities of F-2 cells treated with anticancer drugs.


Asunto(s)
Antineoplásicos/farmacología , Cisplatino/farmacología , Doxorrubicina/farmacología , Células Endoteliales/efectos de los fármacos , Lisofosfolípidos/metabolismo , Receptores del Ácido Lisofosfatídico/genética , Línea Celular , Movimiento Celular/efectos de los fármacos , Técnicas de Cocultivo , Células Endoteliales/citología , Células Endoteliales/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Páncreas/citología , Páncreas/patología
18.
Growth Factors ; 32(3-4): 117-22, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24798396

RESUMEN

Lysophosphatidic acid (LPA) is a bioactive lipid that interacts with G protein-coupled LPA receptors (LPA receptor-1 (LPA1) to LPA6). Here, we investigated the effects of LPA signaling via LPA5 on cellular functions of sarcoma cells by generating Lpar5 overexpressing and Lpar5 knockdown cells from rat osteosarcoma and malignant fibrous histiocytoma cells, respectively. The cell motility activity of Lpar5 overexpressing cells was significantly lower, while Lpar5 knockdown cells showed high cell motility, compared with respective controls. Gelatin zymography showed that LPA5 suppressed the activation of matrix metalloproteinase-2. LPA5 also inhibited the cell motility activity of endothelial cells, correlating with the expression levels of vascular endothelial growth factor genes. These results suggest that LPA signaling via LPA5 negatively regulates the cellular functions of rat sarcoma cells.


Asunto(s)
Receptores del Ácido Lisofosfatídico/metabolismo , Sarcoma/metabolismo , Animales , Células COS , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Chlorocebus aethiops , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Células Endoteliales/fisiología , Lisofosfolípidos/farmacología , Metaloproteinasa 2 de la Matriz/metabolismo , Ratas , Receptores del Ácido Lisofosfatídico/genética , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
19.
Biochem Biophys Res Commun ; 446(2): 585-9, 2014 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-24632199

RESUMEN

Lysophosphatidic acid (LPA) signaling via G protein-coupled LPA receptors (LPA1-LPA6) mediates a variety of biological functions, including cell migration. Recently, we have reported that LPA1 inhibited the cell motile activities of mouse fibroblast 3T3 cells. In the present study, to evaluate a role of LPA5 in cellular responses, Lpar5 knockdown (3T3-L5) cells were generated from 3T3 cells. In cell proliferation assays, LPA markedly stimulated the cell proliferation activities of 3T3-L5 cells, compared with control cells. In cell motility assays with Cell Culture Inserts, the cell motile activities of 3T3-L5 cells were significantly higher than those of control cells. The activity levels of matrix metalloproteinases (MMPs) were measured by gelatin zymography. 3T3-L5 cells stimulated the activation of Mmp-2, correlating with the expression levels of Mmp-2 gene. Moreover, to assess the co-effects of LPA1 and LPA5 on cell motile activities, Lpar5 knockdown (3T3a1-L5) cells were also established from Lpar1 over-expressing (3T3a1) cells. 3T3a1-L5 cells increased the cell motile activities of 3T3a1 cells, while the cell motile activities of 3T3a1 cells were significantly lower than those of control cells. These results suggest that LPA5 may act as a negative regulator of cellular responses in mouse fibroblast 3T3 cells, similar to the case for LPA1.


Asunto(s)
Células 3T3/metabolismo , Movimiento Celular/fisiología , Metaloproteinasa 9 de la Matriz/metabolismo , Receptores del Ácido Lisofosfatídico/metabolismo , Transducción de Señal/fisiología , Animales , Regulación hacia Abajo/fisiología , Activación Enzimática , Ratones
20.
J Recept Signal Transduct Res ; 34(3): 201-4, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24460192

RESUMEN

Lysophosphatidic acid (LPA) signaling via G protein-coupled transmembrane LPA receptors (LPA1 to LPA6) mediates a variety of cellular functions, including cell proliferation, migration, morphogenesis, and differentiation. Recently, we demonstrated that the different induction of LPA receptors by estrogens regulates cell motile activity of rat liver epithelial WB-F344 cells. In the present study, to assess whether endocrine disruptors (EDs) are involved in cellular functions through LPA signaling, we measured cell motile activity and LPA receptor expressions in WB-F344 cells treated with bisphenol A (BPA) and 4-nonylphenol (4-NP). Using quantitative real time RT-PCR analysis, the Lpar1 expression was elevated in BPA-treated cells, whereas the Lpar3 expression was decreased. In contrast, 4-NP increased the Lpar3 expression, but not the Lpar1 and Lpar2. For cell motility assay with a Cell Culture Insert, cell motile activity of BPA-treated cells was significantly lower than that of untreated cells. In contrast, 4-NP markedly enhanced cell motile activity. The effects of BPA and 4-NP on cell motility were inhibited by the Lpar1 or Lpar3 knockdown. These results suggest that BPA and 4-NP may regulate cell motile activity through the different induction of LPA receptors in WB-F344 cells.


Asunto(s)
Compuestos de Bencidrilo/administración & dosificación , Disruptores Endocrinos/administración & dosificación , Células Epiteliales/fisiología , Hígado/fisiología , Lisofosfolípidos/metabolismo , Fenoles/administración & dosificación , Receptores del Ácido Lisofosfatídico/metabolismo , Animales , Línea Celular , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Proliferación Celular/efectos de los fármacos , Proliferación Celular/fisiología , Relación Dosis-Respuesta a Droga , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Hígado/citología , Hígado/efectos de los fármacos , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA