Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Neurochem ; 156(4): 499-512, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32438456

RESUMO

Glioblastomas (GBMs) are highly aggressive primary brain tumors characterized by cellular heterogeneity, insensitivity to chemotherapy and poor patient survival. Lysophosphatidic acid (LPA) is a lysophospholipid that acts as a bioactive signaling molecule and plays important roles in diverse biological events during development and disease, including several cancer types. Microglial cells, the resident macrophages of the central nervous system, express high levels of Autotaxin (ATX,Enpp2), an enzyme that synthetizes LPA. Our study aimed to investigate the role of LPA on tumor growth and invasion in the context of microglia-GBM interaction. First, through bioinformatics studies, patient data analysis demonstrated that more aggressive GBM expressed higher levels of ENPP2, which was also associated with worse patient prognosis with proneural GBM. Using GBM-microglia co-culture system we then demonstrated that GBM secreted factors were able to increase LPA1 and ATX in microglia, which could be further enhanced by hypoxia. On the other hand, interaction with microglial cells also increased ATX expression in GBM. Furthermore, microglial-induced GBM proliferation and migration could be inhibited by pharmacological inhibition of LPA1 , suggesting that microglial-derived LPA could support tumor growth and invasion. Finally, increased LPA1 expression was observed in GBM comparing with other gliomas and could be also associated with worse patient survival. These results show for the first time a microglia-GBM interaction through the LPA pathway with relevant implications for tumor progression. A better understanding of this interaction can lead to the development of new therapeutic strategies setting LPA as a potential target for GBM treatment.


Assuntos
Neoplasias Encefálicas/metabolismo , Movimento Celular/fisiologia , Glioblastoma/metabolismo , Lisofosfolipídeos/metabolismo , Microglia/metabolismo , Receptores de Ácidos Lisofosfatídicos/biossíntese , Animais , Neoplasias Encefálicas/patologia , Proliferação de Células/fisiologia , Células Cultivadas , Feminino , Glioblastoma/patologia , Humanos , Masculino , Camundongos , Microglia/patologia
2.
Anticancer Res ; 40(2): 733-741, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-32014915

RESUMO

BACKGROUND/AIM: GPR87 is a member of the cell surface molecular G protein-coupled receptors (GPCR) family and suggested to contribute to the viability of human tumor cells. Its tumor-specific expression and cell surface location make it a potential molecule for targeted therapy. In the present study, we aimed to examine the effect of silencing GPR87 expression and explore the possibility of establishing gene therapy against GPR87-overexpressing lung cancer. MATERIALS AND METHODS: Twenty malignant cell lines were investigated and GPR87-overexpressing H358 and PC9 lung cancer cells were subjected to inhibiting experiments. A short hairpin siRNA targeting the GPR87 gene was transformed into an adenoviral vector (Ad-shGPR87). Real-time RT-PCR and western blot analyses were performed to evaluate gene and protein expression. Tumors derived from human H358 cells were subcutaneously implanted in nude mice for in vivo experiments. RESULTS AND CONCLUSION: About 50% (10/20) malignant cells showed GPR87-overexpression, especially for lung cancer cells (70%, 7/10). Ad-shGPR87 effectively down-regulated the GPR87 expression, and significantly inhibited the cell proliferation in GPR87-overexpressing H358 and PC9 cells. Treatment with Ad-shGPR87 exerted a significant antitumor effect against the GPR87-expressing H358 xenografts. In addition, the gene expression of H3.3, a recently proved activator for GPR87 transcription, was positively correlated with GPR87 gene expression. Furthermore, a significant decrease of KRAS and c-Myc expression was observed in both cell lines after Ad-shGPR87 infection. In conclusion, GPR87 may play a critical role in cancer cell proliferation, and indicate its potential as a novel target for lung cancer treatment.


Assuntos
Terapia Genética/métodos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/terapia , RNA Interferente Pequeno/administração & dosagem , Receptores de Ácidos Lisofosfatídicos/antagonistas & inibidores , Adenoviridae/genética , Animais , Linhagem Celular Tumoral , Proliferação de Células/genética , Expressão Gênica , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Nus , Proteínas Proto-Oncogênicas p21(ras)/genética , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , RNA Interferente Pequeno/genética , Receptores de Ácidos Lisofosfatídicos/biossíntese , Receptores de Ácidos Lisofosfatídicos/genética , Transdução de Sinais , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Am J Reprod Immunol ; 82(5): e13176, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31357233

RESUMO

PROBLEM: Recent advances in lipid research have revealed that impairments in lipid mediator signaling can be involved in the pathoetiology of a variety of diseases. We previously reported aberrant expression of autotaxin, a key enzyme for lysophosphatidic acid (LPA) production, in placentas from women with preeclampsia. The present study aimed to further explore the involvement of LPA signaling in the pathoetiology of preeclampsia. METHOD OF STUDY: Term placentas were obtained from deliveries after uncomplicated pregnancy (n = 18) and those complicated by preeclampsia (n = 24). First-trimester placental tissues were collected after elective terminations of pregnancy (n = 20). Placental expression of the six identified LPARs (LPAR1-6) was analyzed at protein and mRNA levels. RESULTS: In normal pregnancy, the mRNA expression levels of all LPARs except LPAR4 were significantly higher in term. Levels of mRNA encoding LPAR2-5 were significantly increased in preeclampsia placentas compared with those in the normal term placentas. Using Western immunoblotting, only LPAR3 was noted to be increased at the protein level in placentas from preeclamptic pregnancies. This was validated by immunohistochemistry. CONCLUSION: In summary, the placental expression of LPARs, particularly LPAR3, is enhanced in preeclampsia, suggesting that disturbances in placental LPA signaling may be involved in the pathogenesis of preeclampsia.


Assuntos
Regulação da Expressão Gênica , Placenta/metabolismo , Pré-Eclâmpsia/metabolismo , Receptores de Ácidos Lisofosfatídicos/biossíntese , Adulto , Feminino , Humanos , Placenta/patologia , Pré-Eclâmpsia/patologia , Gravidez
4.
Med Sci Monit ; 25: 4705-4715, 2019 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-31235682

RESUMO

BACKGROUND The aim of this study was to evaluate lysophosphatidic acid receptor-2 (LPA2) and Krüppel-like factor 5 (KLF5) protein expression in gastric adenocarcinoma and their correlation with patient clinicopathological characteristics and prognosis. MATERIAL AND METHODS Fifty-one gastric adenocarcinoma tissue samples, 21 gastric intraepithelial neoplasia (GIN) samples, and 13 normal gastric tissue samples were collected to test for LPA2 and KLF5 expression by tissue microarray and immunohistochemistry assay. LPA2 and KLF5 positive expression rate between gastric adenocarcinoma, GIN, and normal gastric tissue were compared. The relationship between LPA2 expression, KLF5 expression, and patients' clinicopathological characteristics and prognosis were evaluated. RESULTS The positive expression rate of LPA2 and KLF5 were statistical different in gastric adenocarcinoma, GIN, and normal gastric tissue (P<0.05). LPA2 positive expression was associated with tumor invasion depth, Lauren type, vascular invasion, local lymph node metastasis, and clinical stage (P<0.05). There was no correlation between LPA2 expression (hazard ratio [HR]=1.84, 95% confidence interval [CI]: 0.89-3.80, P>0.05), KLF5 expression (HR=1.13, 95% CI: 0.53-2.36, P>0.05), and gastric cancer patients' overall survival. CONCLUSIONS LPA2 and KLF5 protein expressions were differently expressed in gastric adenocarcinoma, GIN, and normal gastric tissue, and differences were correlated with patients' clinical characteristic. However, LPA2 and KLF5 expressions were not correlated with the patients' prognosis.


Assuntos
Fatores de Transcrição Kruppel-Like/metabolismo , Receptores de Ácidos Lisofosfatídicos/metabolismo , Neoplasias Gástricas/metabolismo , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Adulto , Idoso , Feminino , Humanos , Imuno-Histoquímica , Fatores de Transcrição Kruppel-Like/biossíntese , Fatores de Transcrição Kruppel-Like/genética , Masculino , Pessoa de Meia-Idade , Prognóstico , Modelos de Riscos Proporcionais , Receptores de Ácidos Lisofosfatídicos/biossíntese , Receptores de Ácidos Lisofosfatídicos/genética , Neoplasias Gástricas/patologia
5.
Toxicol Lett ; 288: 65-70, 2018 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-29454887

RESUMO

Estrogen is reported to be involved in mammary tumorigenesis. To unveil metabolic signatures for estrogen-induced mammary tumorigenesis, we carried out serum metabolomic analysis in an estrogen-induced mammary tumor model, female August Copenhagen-Irish/Segaloff (ACI/Seg) rats, using liquid chromatography-mass spectrometry. In contrast to the control group, all rats with an implanted 17ß-estradiol (E2) pellet developed mammary tumors during this experiment. E2 treatment significantly suppressed body weight gain. But no significant differences in food consumption were observed between the two groups, suggesting that metabolic alteration depended on E2 treatment. Serum metabolomic analysis detected 116 features that were statistically different (p < 0.01) between the groups. Quantitation analysis revealed that several phospholipids such as phosphatidylcholines and lysophosphatidylcholines (LPCs) were identified as significantly different metabolites. E2-treated rat serum stimulated the proliferation of human breast cancer MDA-MB-231 cells. In addition, the proliferation effect was diminished by pretreating cells with either autotaxin inhibitor or antagonist for lysophosphatidic acid receptor whose ligands are metabolites of LPCs via autotaxin-mediated hydrolysis. In summary, our results suggest that not only are phospholipids potential biomarkers for mammary tumors but importantly, LPCs themselves could be associated with E2-induced mammary tumorigenesis in female ACI/Seg rats.


Assuntos
Estradiol/toxicidade , Neoplasias Mamárias Experimentais/induzido quimicamente , Neoplasias Mamárias Experimentais/metabolismo , Metabolômica , Fosfolipídeos/sangue , Animais , Biomarcadores , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Implantes de Medicamento , Ingestão de Alimentos/efeitos dos fármacos , Estradiol/administração & dosagem , Feminino , Humanos , Camundongos , Diester Fosfórico Hidrolases/biossíntese , Diester Fosfórico Hidrolases/genética , Ratos Endogâmicos ACI , Receptores de Ácidos Lisofosfatídicos/biossíntese , Receptores de Ácidos Lisofosfatídicos/efeitos dos fármacos , Receptores de Ácidos Lisofosfatídicos/genética , Transdução de Sinais/efeitos dos fármacos , Aumento de Peso/efeitos dos fármacos
6.
Biomed Pharmacother ; 101: 49-57, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-29477472

RESUMO

AIMS: Lysophosphatidic acid is a lipid mediator that is supposed to be implicated in hepatic fibrosis. Silymarin and caffeine are natural compounds known for their anti-inflammatory and antioxidant effects. Our study aimed to explore the effect of silymarin, caffeine, and their combination on lysophosphatidic acid receptor 1 (LPAR1) pathway in thioacetamide (TAA)-induced hepatic fibrosis. MAIN METHODS: Hepatic fibrosis was induced in male Sprague-Dawley rats by intraperitoneal injection of 200 mg/kg of TAA twice a week for 8 weeks. Silymarin (50 mg/kg), caffeine (50 mg/kg), and their combination (50 mg/kg silymarin + 50 mg/kg caffeine) were orally given to rats every day for 8 weeks along with TAA injection. Liver functions were measured. Histopathological examination of liver tissues was performed using hematoxylin and eosin and Masson's trichrome staining. mRNA expressions of LPAR1, transforming growth factor beta 1 (TGF-ß1), connective tissue growth factor (CTGF), and alpha smooth muscle actin (α-SMA) were measured using RT-PCR. LPAR1 tissue expression was scored using immunohistochemistry. KEY FINDINGS: Silymarin, caffeine, and their combination significantly improved liver function. They caused significant decrease in fibrosis and necro-inflammatory scores. Combination of silymain and caffeine caused a significant decrease in the necro-inflammatory score than the single treatment with silymarin or caffeine. In addition, silymarin, caffeine, and their combination significantly decreased hepatic LPAR1, TGF-ß1, CTGF, and α-SMA gene expressions and LPAR1 tissue expression. SIGNIFICANCE: Silymarin, caffeine, and their combination protect against liver fibrosis through down-regulation of LPAR1, TGF-ß1, and CTGF.


Assuntos
Antioxidantes/administração & dosagem , Cafeína/administração & dosagem , Cirrose Hepática/tratamento farmacológico , Cirrose Hepática/metabolismo , Receptores de Ácidos Lisofosfatídicos/biossíntese , Silimarina/administração & dosagem , Animais , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/fisiologia , Quimioterapia Combinada , Expressão Gênica , Cirrose Hepática/induzido quimicamente , Masculino , Ratos , Ratos Sprague-Dawley , Receptores de Ácidos Lisofosfatídicos/antagonistas & inibidores , Receptores de Ácidos Lisofosfatídicos/genética , Tioacetamida/toxicidade
7.
Life Sci ; 197: 73-79, 2018 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-29412173

RESUMO

AIM: The intestinal mucus layer helps maintain intestinal homeostasis. In this study, we investigated the effects of lysophosphatidic acids (LPA) on differentiation of human colon carcinoma cell line, HT-29, to goblet cells with and without sodium butyrate, a known differentiation factor for intestinal cells. MAIN METHODS: Number and average size of cells with goblet-like morphology in five photographs per dish were measured for assessment of differentiation of HT-29 cells to goblet cells as well as their relative portion of surface of to whole surface area of the photograph. KEY FINDINGS: Our results revealed that 18:1 LPA enhanced butyrate-induced differentiation of HT-29 cells. Because increased mRNA expression of LPA5 and decreased mRNA expression of LPA6 were observed in HT-29 cells after treatment with butyrate, we explored the effects of alkyl LPA and 20:4 LPA, which show preferentially higher affinities to LPA5 and LPA6, respectively. As a result, the cell differentiation to goblet cell was increased by alkyl LPA but decreased by 20:4 LPA. Further, alkyl LPA and 18:1 LPA, but not 20:4 LPA, were found to reduce the numbers of cells surviving after incubation in a standard culture medium containing 10% fetal calf serum. SIGNIFICANCE: We suggest that the three LPAs positively and negatively affect the differentiation of HT-29 cells to goblet cells, which may be associated with their reduced survival through the activation of distinct LPA receptor(s).


Assuntos
Diferenciação Celular/efeitos dos fármacos , Colo/metabolismo , Células Caliciformes/metabolismo , Lisofosfolipídeos/farmacologia , Ácido Butírico/farmacologia , Colo/citologia , Regulação da Expressão Gênica/efeitos dos fármacos , Células Caliciformes/citologia , Humanos , Receptores de Ácidos Lisofosfatídicos/biossíntese
8.
Leuk Res ; 66: 39-48, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29407582

RESUMO

The NM23 gene is overexpressed in many hematological malignancies and its overexpression predicts poor treatment outcomes. NM23 overexpression is thought to suppress myeloid differentiation of leukemia cells, but the molecular mechanism is unknown. In breast cancer cells, the lysophosphatidic acid (LPA) receptor EDG2/lpa1 was downregulated by NM23-H1 overexpression, and this reciprocal expression pattern was associated with suppressed or induced cell motility/metastasis. Here, we examined the relationship between EDG2 and NM23 expression during myeloid differentiation of leukemia cells. NM23 expression decreased and EDG2 expression increased during all-trans retinoic acid (ATRA)-induced myeloid differentiation of HL-60, NB4, and THP-1 leukemia cells. Moreover, this inverse correlation was more evident when myeloid differentiation was enhanced by ellagic acid, an inhibitor of NM23 activity. In contrast, there was no inverse correlation between EDG2 and NM23 expression during erythroid differentiation of HEL and K562 cells. ATRA plus LPA enhanced the motility of leukemia cells as well as breast cancer cells in an EDG2-dependent manner. These results suggest a common molecular mechanism between myeloid differentiation of leukemia cells and migration of breast cancer cells depending on NM23 and EDG2 expression levels.


Assuntos
Diferenciação Celular , Regulação para Baixo , Regulação Leucêmica da Expressão Gênica , Leucemia/metabolismo , Células Mieloides/metabolismo , Nucleosídeo NM23 Difosfato Quinases/biossíntese , Proteínas de Neoplasias/biossíntese , Receptores de Ácidos Lisofosfatídicos/biossíntese , Regulação para Cima , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Feminino , Células HL-60 , Humanos , Células K562 , Leucemia/genética , Leucemia/patologia , Lisofosfolipídeos/farmacologia , Células MCF-7 , Células Mieloides/patologia , Nucleosídeo NM23 Difosfato Quinases/genética , Receptores de Ácidos Lisofosfatídicos/genética , Tretinoína/farmacologia , Células U937
9.
Mol Cell Biochem ; 431(1-2): 29-35, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28205098

RESUMO

Lysophosphatidic acid (LPA) is a simple biophysical lipid which interacts with at least six subtypes of G protein-coupled LPA receptors (LPA1-LPA6). In cancer cells, LPA signaling via LPA receptors is involved in the regulation of malignant properties, such as cell growth, motility, and invasion. The aim of this study was to assess whether LPA receptors regulate cellular functions of fibrosarcoma cells treated with anticancer drug. HT1080 cells were maintained by the stepwise treatment of cisplatin (CDDP) at a range of 0.01 to 1.0 µM for approximately 6 months. The cell motile and invasive activities of long-term CDDP-treated (HT-CDDP) cells were significantly stimulated by LPA treatment, while HT-CDDP cells in the static state showed the low cell motile and invasive activities in comparison with HT1080 cells. Since the expression level of LPAR2 gene was markedly elevated in HT-CDDP cells, LPA2 knockdown cells were generated from HT-CDDP cells. The cell motile and invasive activities of HT-CDDP cells were reduced by LPA2 knockdown. In colony assay, large-sized colonies formed by long-term CDDP treatment were suppressed by LPA2 knockdown. In addition, LPA2 knockdown cells reduced LPA production by autotaxin (ATX), correlating with ATX expression level. These results suggest that LPA signaling via LPA2 may play an important role in the regulation of cellular functions in HT1080 cells treated with CDDP.


Assuntos
Movimento Celular/efeitos dos fármacos , Cisplatino/farmacologia , Fibrossarcoma/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Proteínas de Neoplasias/biossíntese , Receptores de Ácidos Lisofosfatídicos/biossíntese , Movimento Celular/genética , Fibrossarcoma/genética , Fibrossarcoma/patologia , Regulação Neoplásica da Expressão Gênica/genética , Técnicas de Silenciamento de Genes , Humanos , Invasividade Neoplásica , Proteínas de Neoplasias/genética , Diester Fosfórico Hidrolases/biossíntese , Diester Fosfórico Hidrolases/genética , Receptores de Ácidos Lisofosfatídicos/genética
10.
Oncotarget ; 7(3): 2951-67, 2016 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-26701886

RESUMO

Hepatocellular carcinoma (HCC) is the most common primary cancer of the liver and is characterized by rapid tumor expansion and metastasis. Lysophosphatidic acid (LPA) signaling, via LPA receptors 1-6 (LPARs1-6), regulates diverse cell functions including motility, migration, and proliferation, yet the role of LPARs in hepatic tumor pathology is poorly understood. We sought to determine the expression and function of endothelial differentiation gene (EDG) LPARs (LPAR1-3) in human HCC and complimentary in vitro models. Human HCC were characterized by significantly elevated LPAR1/LPAR3 expression in the microenvironment between the tumor and non-tumor liver (NTL), a finding mirrored in human SKHep1 cells. Analysis of human tissue and human hepatic tumor cells in vitro revealed cells that express LPAR3 (HCC-NTL margin in vivo and SKHep1 in vitro) also express cancer stem cell markers in the absence of hepatocyte markers. Treatment of SKHep1 cells with exogenous LPA led to significantly increased cell motility but not proliferation. Using pharmacological agents and cells transfected to knock-down LPAR1 or LPAR3 demonstrated LPA-dependent cell migration occurs via an LPAR3-Gi-ERK-pathway independent of LPAR1. These data suggest cells that stain positive for both LPAR3 and cancer stem cell markers are distinct from the tumor mass per se, and may mediate tumor invasiveness/expansion via LPA-LPAR3 signaling.


Assuntos
Carcinoma Hepatocelular/patologia , Neoplasias Hepáticas/patologia , Lisofosfolipídeos/farmacologia , Células-Tronco Neoplásicas/patologia , Receptores de Ácidos Lisofosfatídicos/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Feminino , Células Hep G2 , Humanos , Lisofosfolipídeos/metabolismo , Masculino , Pessoa de Meia-Idade , Interferência de RNA , RNA Interferente Pequeno/genética , Receptores de Ácidos Lisofosfatídicos/biossíntese , Transdução de Sinais
11.
Int J Mol Sci ; 16(10): 24319-31, 2015 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-26473854

RESUMO

G protein-coupled receptor 87 (GPR87) is a newly deorphanized member of the cell surface molecule G protein-coupled receptor family. GPR signaling was shown to play a role in promotion of cell growth and survival, metastasis, and drug resistance. The overexpression of GPR87 has also been reported in many malignant tumors including bladder cancer. The aim of the present study is to examine the effect of silencing GPR87 expression with a replication-deficient recombinant adenoviral vector expressing short hairpin RNA targeting GPR87 (Ad-shGPR87) and to explore the underlying molecular mechanisms in bladder cancer cells. Six GPR87-expressing human bladder cancer cells, HT1197, HT1376, J82, RT112, TCCSUP and UMUC3, were used. Infection with Ad-shGPR87 effectively downregulated the GPR87 expression, and significantly reduced the percentage of viable cells in 4 of 6 cell lines as detected by an MTT assay. Significant inhibition on cell proliferation with Ad-shGPR87 was observed in the wild-type p53 bladder cancer cell lines (HT1197, RT112, TCCSUP and UMUC3), but not in the mutant p53 cells (HT1376 and J82). As represented by a wild-type p53 RT112 cell, Ad-shGPR87 infection significantly enhanced p53 and p21 expression and caused caspase-dependent apoptosis. Furthermore, the treatment with Ad-shGPR87 exerted a significant antitumor effect against the GPR87-expressing RT112 xenografts. GPR87 appeared to be a promising target for gene therapy, and Ad-shGPR87 had strong antitumor effects, specifically anti-proliferative and pro-apoptotic effects, against GPR87-expressing human bladder cancer cells.


Assuntos
Inibidor de Quinase Dependente de Ciclina p21/genética , Receptores de Ácidos Lisofosfatídicos/genética , Proteína Supressora de Tumor p53/genética , Neoplasias da Bexiga Urinária/patologia , Animais , Apoptose/genética , Linhagem Celular Tumoral , Proliferação de Células/genética , Sobrevivência Celular/genética , Inibidor de Quinase Dependente de Ciclina p21/biossíntese , Humanos , Camundongos , Camundongos SCID , Transplante de Neoplasias , Interferência de RNA , RNA Interferente Pequeno/genética , Receptores de Ácidos Lisofosfatídicos/biossíntese , Receptores de Ácidos Lisofosfatídicos/metabolismo , Transdução de Sinais/genética , Transplante Heterólogo , Proteína Supressora de Tumor p53/biossíntese , Neoplasias da Bexiga Urinária/genética
12.
J Urol ; 194(1): 238-44, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25524242

RESUMO

PURPOSE: LPA is one of several physiologically active lipid mediators that promote cell proliferation and invasion, and are present in serum, ascites and urine. LPA receptor is a G-protein coupled receptor that is considered a potential therapeutic target for some malignant cancers. We evaluated the expression of LPA receptors in bladder cancer and the effect of LPA in bladder cancer invasion. MATERIALS AND METHODS: Using real-time polymerase chain reaction and immunohistochemical staining we determined LPA receptor expression in bladder cancer specimens from patients with bladder cancer, including 12 with Ta or T1 and 15 with T2-T4 disease. ROCK expression, myosin light chain phosphorylation and Matrigel™ invasion assays were done and morphological observations were made to assess LPA effects in T24 cells, which were derived from bladder cancer. RESULTS: Notably LPA1 mRNA expression was significantly higher in muscle invasive bladder cancer specimens than in nonmuscle invasive specimens. Strong LPA1 expression was evident on cell membranes in muscle invasive specimens. T24 cell invasion was increased by LPA treatment and invasiveness was decreased by LPA1 siRNA or LPA1 inhibitor. LPA treatment increased ROCK1 expression and myosin light chain phosphorylation, and induced morphological changes, including lamellipodia formation and cell rounding. CONCLUSIONS: Results indicate that LPA signaling via LPA1 activation promoted bladder cancer invasion. LPA1 might be useful to detect bladder cancer with highly invasive potential and become a new therapeutic target for invasive bladder cancer treatment.


Assuntos
Receptores de Ácidos Lisofosfatídicos/biossíntese , Receptores de Ácidos Lisofosfatídicos/fisiologia , Neoplasias da Bexiga Urinária/metabolismo , Neoplasias da Bexiga Urinária/patologia , Idoso , Feminino , Humanos , Masculino , Invasividade Neoplásica , Células Tumorais Cultivadas , Neoplasias da Bexiga Urinária/cirurgia
13.
Mol Hum Reprod ; 20(10): 1016-25, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24994816

RESUMO

In the luteal phase, human endometrial stromal cells (HESCs) undergo proliferation, migration and differentiation during the decidualization process under the control of the ovarian steroids progesterone and estrogen. Proper decidualization of stromal cells is required for blastocyst implantation and the development of pregnancy. The proliferation, migration and differentiation of HESCs in decidualization do not require the presence of a blastocyst but are greatly accelerated during implantation. Lysophosphatidic acid (LPA) and sphingosine-1-phosphate (S1P) are potent bioactive lysophospholipids that have critical roles in various physiological and pathophysiological processes, including inflammation, angiogenesis and cancer. The expression of the enzymes involved in LPA and S1P turnover and their receptors in HESCs during decidualization has not been characterized yet. We found that the LPAR1 and LPAR6 and S1PR3 receptors are highly expressed in HESCs. LPAR1, autotaxin (ATX), an LPA producing enzyme and lipid phosphate phosphatase 3 were up-regulated during decidualization. Interestingly, the expression of all S1P receptor subtypes and LPA receptors (LPAR2-6) mRNA was down-regulated after decidualization. We found that SPHK1 is highly expressed in HESCs, and is up-regulated during decidualization. S1P phosphatase SGPP1 and S1P lyase SGPL1 are highly expressed in HESCs. SGPP1 mRNA expression was significantly up-regulated in decidualized HESCs. In conclusion, this study shows the first time that specific LPA and S1P receptors and their metabolizing enzymes are highly regulated in HESCs during decidualization. Furthermore, we suggest that LPAR1 receptor-mediated signaling in HESCs may be crucial in decidualization process. SPHK1 activity and high turnover of S1P and LPA might be essential for precise regulation of their signaling during decidualization of human endometrium and implantation.


Assuntos
Implantação do Embrião/fisiologia , Lisofosfolipídeos/metabolismo , Receptores de Ácidos Lisofosfatídicos/biossíntese , Receptores de Lisoesfingolipídeo/biossíntese , Esfingosina/análogos & derivados , Adulto , Diferenciação Celular , Movimento Celular , Proliferação de Células , Decídua/fisiologia , Endométrio/citologia , Estrogênios , Feminino , Humanos , Pessoa de Meia-Idade , Fosfatidato Fosfatase/biossíntese , Diester Fosfórico Hidrolases/biossíntese , Gravidez , Progesterona , RNA Mensageiro/biossíntese , Esfingosina/metabolismo , Receptores de Esfingosina-1-Fosfato , Células Estromais/citologia , Ativação Transcricional , Regulação para Cima
14.
BMC Cancer ; 14: 432, 2014 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-24928086

RESUMO

BACKGROUND: Oral squamous cell carcinoma is an aggressive neoplasm with serious morbidity and mortality, which typically spreads through local invasive growth. Lysophosphatidic acid (LPA) is involved in a number of biological processes, and may have a role in cancer cell migration and invasiveness. LPA is present in most tissues and can activate cells through six different LPA receptors (LPAR1-6). Although LPA is predominantly promigratory, some of the receptors may have antimigratory effects in certain cells. The signalling mechanisms of LPA are not fully understood, and in oral carcinoma cells the specific receptors and pathways involved in LPA-stimulated migration are unknown. METHODS: The oral carcinoma cell lines E10, SCC-9, and D2 were investigated. Cell migration was studied in a scratch wound assay, and invasion was demonstrated in organotypic three dimensional co-cultures. Protein and mRNA expression of LPA receptors was studied with Western blotting and qRT-PCR. Activation of signalling proteins was examined with Western blotting and isoelectric focusing, and signalling mechanisms were further explored using pharmacological agents and siRNA directed at specific receptors and pathways. RESULTS: LPA stimulated cell migration in the two oral carcinoma cell lines E10 and SCC-9, but was slightly inhibitory in D2. The receptor expression profile and the effects of specific pharmacological antagonist and agonists indicated that LPA-stimulated cell migration was mediated through LPAR3 in E10 and SCC-9. Furthermore, in both these cell lines, the stimulation by LPA was dependent on PKC activity. However, while LPA induced transactivation of EGFR and the stimulated migration was blocked by EGFR inhibitors in E10 cells, LPA did not induce EGFR transactivation in SCC-9 cells. In D2 cells, LPA induced EGFR transactivation, but this was associated with slowing of a very high inherent migration rate in these cells. CONCLUSION: The results demonstrate LPA-stimulated migration in oral carcinoma cells through LPAR3, mediated further by PKC, which acts either in concert with or independently of EGFR transactivation.


Assuntos
Carcinoma de Células Escamosas/genética , Receptores ErbB/genética , Neoplasias Bucais/genética , Receptores de Ácidos Lisofosfatídicos/biossíntese , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Receptores ErbB/biossíntese , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Lisofosfolipídeos/administração & dosagem , Neoplasias Bucais/patologia , Proteína Quinase C/genética , Receptores de Ácidos Lisofosfatídicos/genética , Transdução de Sinais/efeitos dos fármacos
15.
Mol Cell Biochem ; 393(1-2): 17-22, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24676544

RESUMO

LPA signaling via LPA receptors [LPA receptor-1 (LPA1)-LPA6] mediates the several cellular responses in cancer cells, including cell motility and invasion. In the present study, to investigate a role of LPA5 in the cell motile and invasive activities of sarcoma cells, LPAR5 knockdown (HOSL5 and HT1080L5) cells were generated from human osteosarcoma HOS and fibrosarcoma HT1080 cells, respectively. In cell motility assays with cell culture inserts, HOSL5 and HT1080L5 cells indicated the high cell motile activities, compared with control cells. The cell invasive activities of HOSL5 and HT1080L5 cells were significantly higher than those of control cells. Moreover, the activities of matrix metalloproteinase (MMP)-2 and MMP-9 were measured by gelatin zymography. MMP-2 was significantly activated in HOSL5 cells, but not MMP-9. The elevated activities of MMP-2 and MMP-9 were found in HT1080L5 cells, in comparison with control cells. These results suggest that LPA signaling via LPA5 negatively regulates the cell motile and invasive activities of human sarcoma cells.


Assuntos
Lisofosfolipídeos/metabolismo , Receptores de Ácidos Lisofosfatídicos/genética , Sarcoma/genética , Transdução de Sinais , Linhagem Celular Tumoral , Movimento Celular/genética , Humanos , Lisofosfolipídeos/genética , Metaloproteinase 2 da Matriz/biossíntese , Metaloproteinase 9 da Matriz/biossíntese , Invasividade Neoplásica/genética , Receptores de Ácidos Lisofosfatídicos/biossíntese , Sarcoma/patologia
16.
J Recept Signal Transduct Res ; 33(5): 286-90, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23909436

RESUMO

Angiogenesis stimulates the invasive and metastatic process of cancer cells. It is also known that activated fibroblasts promote cancer cell growth and enhance invasive and metastatic potential. Lysophosphatidic acid (LPA) is a biological mediator and interacts with G protein-coupled transmembrane LPA receptors (LPA1 to LPA6). In this study, to assess an involvement of LPA3 on angiogenesis and fibroblast activation, the Lpar3-expressing cells were generated from mouse lung cancer LL/2 cells, which unexpressed LPA3. The Lpar3-expressing cells were maintained in serum-free Dulbecco's modified Eagle's medium for 48 h, and cell motility assay was performed with a cell culture Insert. When endothelial F-2 cells and 3T3 fibroblasts were cultured with conditioned medium from the Lpar3-expressing cells, their cell motile activities were significantly lower than the Lpar3-unexpressing (control) cells. Expression levels of vascular endothelial growth factor (Vegf) and fibroblast growth factor (Fgf) genes in the Lpar3-expressing cells were measured by quantitative real time reverse transcription polymerase chain reaction analysis. The expressions of Vegf-A. Fgfa and Fgfb genes in the Lpar3-expressing cells were significantly lower than those in control cells, correlating with the effects on cell motile activities of F-2 and 3T3 cells. These results suggest that LPA signaling through LPA3 may inhibit angiogenesis and fibroblast activation in mouse lung cancer cells.


Assuntos
Neoplasias Pulmonares/metabolismo , Lisofosfolipídeos/metabolismo , Neoplasias Experimentais/genética , Neovascularização Patológica/metabolismo , Receptores de Ácidos Lisofosfatídicos/biossíntese , Células 3T3 , Animais , Movimento Celular/genética , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Fibroblastos/citologia , Fibroblastos/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Camundongos , Invasividade Neoplásica/genética , Metástase Neoplásica , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Receptores de Ácidos Lisofosfatídicos/genética , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/biossíntese , Fator A de Crescimento do Endotélio Vascular/genética
17.
PLoS One ; 8(2): e57742, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23451264

RESUMO

BACKGROUND: Lysophosphatidic acid (LPA) is a phospholipid growth factor involved in cell proliferation, differentiation, migration, inflammation, angiogenesis, wound healing, cancer invasion, and survival. This study was directed to evaluate the immunoexpression of LPA-1, cell proliferation, apoptosis, and angiogenesis markers in preneoplastic lesions induced with cadmium chloride in rat prostate. METHODS: The following parameters were calculated in ventral prostate of normal rats and rats that received Cd in drinking water during 24 months: percentages of cells immunoreactive to LPA-1 (LILPA1), PCNA (LIPCNA), MCM7 (LIMCM7), ubiquitin (LIUBI), apoptotic cells (LIAPO), and p53 (LIp53); volume fraction of Bcl-2 (VFBcl-2); and length of microvessels per unit of volume (LVMV/mm3). Data were analyzed using Student's t-test and Pearson correlation test. RESULTS: The LILPA1 in dysplastic lesions and normal epithelium of Cd-treated rats was significantly higher than those in the control group. Markers of proliferation were significantly increased in dysplastic lesions, whereas some apoptotic markers were significantly decreased. No significant differences between groups were found in VFBcl-2. Dysplastic lesions showed a significant increase of LIp53. The length of microvessels per unit of volume was elevated in dysplastic acini. Statistically significant correlations were found only between LILPA1 and LIUBI. CONCLUSIONS: Our results suggest that LPA-1 might be implicated in dysplastic lesions induced by cadmium chloride development. More studies are needed to confirm its potential contribution to the disease.


Assuntos
Lesões Pré-Cancerosas/metabolismo , Lesões Pré-Cancerosas/patologia , Próstata/metabolismo , Próstata/patologia , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Receptores de Ácidos Lisofosfatídicos/biossíntese , Animais , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Cloreto de Cádmio/toxicidade , Processos de Crescimento Celular/efeitos dos fármacos , Processos de Crescimento Celular/fisiologia , Masculino , Microvasos/metabolismo , Microvasos/patologia , Componente 7 do Complexo de Manutenção de Minicromossomo/genética , Componente 7 do Complexo de Manutenção de Minicromossomo/metabolismo , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Lesões Pré-Cancerosas/induzido quimicamente , Lesões Pré-Cancerosas/genética , Antígeno Nuclear de Célula em Proliferação/genética , Antígeno Nuclear de Célula em Proliferação/metabolismo , Próstata/irrigação sanguínea , Próstata/efeitos dos fármacos , Neoplasias da Próstata/irrigação sanguínea , Neoplasias da Próstata/induzido quimicamente , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de Ácidos Lisofosfatídicos/genética , Receptores de Ácidos Lisofosfatídicos/metabolismo , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Ubiquitina/genética , Ubiquitina/metabolismo
18.
Oncogene ; 32(26): 3198-206, 2013 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-22824789

RESUMO

The lysophosphatidic acid (LPA) receptor LPA1/Edg2 is the first identified LPA receptor. Although its wide tissue distribution and biological functions have been well studied, little is known about how LPA1 is transcriptionally regulated. In the current study, we showed that LPA1 is a physiological target of transforming growth factor beta (TGFß)-mediated repression. In both normal and neoplastic cells, TGFß inhibits LPA1 promoter activity, LPA1 mRNA expression and LPA1-dependent chemotaxis and tumor cell invasion. Knockdown of the TGFß intracellular effector Smad3 or Smad4 with lentivirally transduced short hairpin RNA relieved these inhibitory effects of TGFß. Interestingly, the LPA1 promoter contains two potential TGFß inhibitory elements (TIEs), each consisting of a Smad-binding site and an adjacent E2F4/5 element, structurally similar to the TIE found on the promoter of the well-defined TGFß target gene c-myc. Deletion and point mutation analyses indicate that the distal TIE located at 401 bp from the transcription initiation site, is required for TGFß repression of the LPA1 promoter. A DNA pull-down assay showed that the -401 TIE was capable of binding Samd3 and E2F4 in TGFß-treated cells. TGFß-induced binding of the Smad complex to the native -401 TIE sequence of the LPA1 gene promoter was further verified by chromatin immunoprecipitation assays. We therefore identified a novel role of TGFß in the control of LPA1 expression and LPA1-coupled biological functions, adding LPA1 to the list of TGFß-repressed target genes.


Assuntos
Receptores de Ácidos Lisofosfatídicos/metabolismo , Proteína Smad3/genética , Proteína Smad4/genética , Fator de Crescimento Transformador beta/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Regulação Neoplásica da Expressão Gênica , Humanos , Regiões Promotoras Genéticas , Interferência de RNA , RNA Mensageiro/biossíntese , RNA Interferente Pequeno , Receptores de Ácidos Lisofosfatídicos/biossíntese , Elementos de Resposta , Transdução de Sinais/genética , Proteína Smad3/metabolismo , Proteína Smad4/metabolismo , Transcrição Gênica
19.
Biochem Biophys Res Commun ; 428(1): 105-9, 2012 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-23058922

RESUMO

Lysophosphatidic acid (LPA) interacts with G protein-coupled transmembrane LPA receptors (LPA receptors; LPA(1)-LPA(6)). Recently, we demonstrated that each LPA receptor acts as a positive or negative regulator of cell migration ability. It is known that estrogens indicate a variety of biological functions, including cell motility. In the present study, to assess whether LPA signaling is involved in cell motile activity stimulated by estrogens, we measured cell motile activity and LPA receptor expressions of rat liver epithelial WB-F344 cells treated with 17ß-estradiol (E(2)), ethinyl estradiol (EE) and diethylstilbestrol (DES) at concentrations of 0.1 and 1.0 µM for 48 h. The cell motility of E(2) and EE treated cells was significantly higher than that of untreated cells. By contrast, DES markedly inhibited cell motile activity. Using quantitative real time RT-PCR analysis, Lpar1 and Lpar3 expressions in E(2) treated cells were significantly higher than those in untreated cells. In EE treated cells, Lpar3 expression was markedly elevated, whereas Lpar1 expression was decreased. On the other hand, Lpar1 expression was significantly increased in DES treated cells. Interestingly, the effects of E(2), EE and DES on cell motility were suppressed by Lpar1 or Lpar3 knockdown. These results suggest that the different induction of LPA receptors by estrogens may regulate cell motile activity of WB-F344 cells.


Assuntos
Movimento Celular/fisiologia , Estrogênios/fisiologia , Fígado/citologia , Receptores de Ácidos Lisofosfatídicos/biossíntese , Animais , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/fisiologia , Estrogênios/farmacologia , Ratos , Receptores de Ácidos Lisofosfatídicos/agonistas
20.
J Biol Chem ; 287(30): 24990-5000, 2012 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-22665482

RESUMO

One of the most common molecular changes in cancer is the increased endogenous lipid synthesis, mediated primarily by overexpression and/or hyperactivity of fatty acid synthase (FAS) and acetyl-CoA carboxylase (ACC). The changes in these key lipogenic enzymes are critical for the development and maintenance of the malignant phenotype. Previous efforts to control oncogenic lipogenesis have been focused on pharmacological inhibitors of FAS and ACC. Although they show anti-tumor effects in culture and in mouse models, these inhibitors are nonselective blockers of lipid synthesis in both normal and cancer cells. To target lipid anabolism in tumor cells specifically, it is important to identify the mechanism governing hyperactive lipogenesis in malignant cells. In this study, we demonstrate that lysophosphatidic acid (LPA), a growth factor-like mediator present at high levels in ascites of ovarian cancer patients, regulates the sterol regulatory element binding protein-FAS and AMP-activated protein kinase-ACC pathways in ovarian cancer cells but not in normal or immortalized ovarian epithelial cells. Activation of these lipogenic pathways is linked to increased de novo lipid synthesis. The pro-lipogenic action of LPA is mediated through LPA(2), an LPA receptor subtype overexpressed in ovarian cancer and other malignancies. Downstream of LPA(2), the G(12/13) and G(q) signaling cascades mediate LPA-dependent sterol regulatory element-binding protein activation and AMP-activated protein kinase inhibition, respectively. Moreover, inhibition of de novo lipid synthesis dramatically attenuated LPA-induced cell proliferation. These results demonstrate that LPA signaling is causally linked to the hyperactive lipogenesis in ovarian cancer cells, which can be exploited for development of new anti-cancer therapies.


Assuntos
Lipogênese/efeitos dos fármacos , Lisofosfolipídeos/farmacologia , Neoplasias Ovarianas/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Linhagem Celular Tumoral , Ácido Graxo Sintases/biossíntese , Feminino , Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Lisofosfolipídeos/metabolismo , Camundongos , Proteínas de Neoplasias/metabolismo , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/patologia , Receptores de Ácidos Lisofosfatídicos/biossíntese , Elementos Reguladores de Transcrição
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA