Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Endocr Relat Cancer ; 23(5): 457-68, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26935838

RESUMO

The bioactive lipid sphingosine-1-phosphate (S1P) is a potent inducer of ML-1 thyroid cancer cell migration and invasion. It evokes migration and invasion by activating S1P receptor 1 and 3 (S1P1,3) and downstream signaling intermediates as well as through cross-communication with vascular endothelial growth factor receptor 2 (VEGFR2). However, very little is known about the role of S1P receptors in thyroid cancer. Furthermore, the currently used treatments for thyroid cancer have proven to be rather unsuccessful. Thus, due to the insufficiency of the available treatments for thyroid cancer, novel and targeted therapies are needed. The S1P receptor functional antagonist FTY720, an immunosuppressive drug currently used for treatment of multiple sclerosis, has shown promising effects as an inhibitor of cancer cell proliferation and invasion. In this study, we investigated the effect of FTY720 on invasion and proliferation of several thyroid cancer cell lines. We present evidence that FTY720 attenuated basal as well as S1P-evoked invasion of these cell lines. Furthermore, FTY720 potently downregulated S1P1, protein kinase Cα(PKCα), PKCßI, and VEGFR2. It also attenuated S1P-evoked phosphorylation of ERK1/2. Our results also showed that FTY720 attenuated S1P-induced MMP2 intracellular expression, S1P-induced secretion of MMP2 and MMP9, and decreased basal MMP2 and MMP9 activity. Moreover, in FTY720-treated cells, proliferation was attenuated, p21 and p27 were upregulated, and the cells were arrested in the G1 phase of the cell cycle. FTY720 attenuated cancer cell proliferation in the chick embryo chorioallantoic membrane assay. Thus, we suggest that FTY720 could be beneficial in the treatment of thyroid cancer.


Assuntos
Antineoplásicos/farmacologia , Cloridrato de Fingolimode/farmacologia , Neoplasias da Glândula Tireoide/patologia , Animais , Linhagem Celular , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Embrião de Galinha , Membrana Corioalantoide/efeitos dos fármacos , Membrana Corioalantoide/patologia , Humanos , Imunossupressores/farmacologia , Lisofosfolipídeos/metabolismo , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Neoplasias da Glândula Tireoide/metabolismo
2.
J Biol Chem ; 290(26): 16116-31, 2015 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-25971967

RESUMO

The identity of calcium channels in the thyroid is unclear. In human follicular thyroid ML-1 cancer cells, sphingolipid sphingosine 1-phosphate (S1P), through S1P receptors 1 and 3 (S1P1/S1P3), and VEGF receptor 2 (VEGFR2) stimulates migration. We show that human thyroid cells express several forms of transient receptor potential canonical (TRPC) channels, including TRPC1. In TRPC1 knockdown (TRPC1-KD) ML-1 cells, the basal and S1P-evoked invasion and migration was attenuated. Furthermore, the expression of S1P3 and VEGFR2 was significantly down-regulated. Transfecting wild-type ML-1 cells with a nonconducting TRPC1 mutant decreased S1P3 and VEGFR2 expression. In TRPC1-KD cells, receptor-operated calcium entry was decreased. To investigate whether the decreased receptor expression was due to attenuated calcium entry, cells were incubated with the calcium chelator BAPTA-AM (1,2-bis(o-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid). In these cells, and in cells where calmodulin and calmodulin-dependent kinase were blocked pharmacologically, S1P3 and VEGFR2 expression was decreased. In TRPC1-KD cells, both hypoxia-inducible factor 1α expression and the secretion and activity of MMP2 and MMP9 were attenuated, and proliferation was decreased in TRPC1-KD cells. This was due to a prolonged G1 phase of the cell cycle, a significant increase in the expression of the cyclin-dependent kinase inhibitors p21 and p27, and a decrease in the expression of cyclin D2, cyclin D3, and CDK6. Transfecting TRPC1 to TRPC1-KD cells rescued receptor expression, migration, and proliferation. Thus, the expression of S1P3 and VEGFR2 is mediated by a calcium-dependent mechanism. TRPC1 has a crucial role in this process. This regulation is important for the invasion, migration, and proliferation of thyroid cancer cells.


Assuntos
Proliferação de Células , Receptores de Lisoesfingolipídeo/genética , Receptores de Fatores de Crescimento do Endotélio Vascular/genética , Canais de Cátion TRPC/metabolismo , Neoplasias da Glândula Tireoide/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Movimento Celular , Ciclina D2/genética , Ciclina D2/metabolismo , Quinases Ciclina-Dependentes/genética , Quinases Ciclina-Dependentes/metabolismo , Humanos , Receptores de Lisoesfingolipídeo/metabolismo , Receptores de Fatores de Crescimento do Endotélio Vascular/metabolismo , Esfingolipídeos/metabolismo , Canais de Cátion TRPC/genética , Neoplasias da Glândula Tireoide/genética , Neoplasias da Glândula Tireoide/fisiopatologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética
3.
Mol Cell Endocrinol ; 404: 113-22, 2015 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-25643979

RESUMO

The bioactive lipid sphingosine-1-phosphate (S1P) has emerged as a potent inducer of cancer cell migration and invasion. Previously, we have shown that S1P induces invasion of ML-1 follicular thyroid cancer cells via S1P receptors 1 and 3 (S1P1,3). Matrix metalloproteinases (MMPs) are zinc-dependent proteolytic enzymes used by cells for degradation of the extracellular matrix during invasion and migration. In the present study, we examined the role of MMP2 and MMP9 for S1P-induced invasion of ML-1 cells, and found that S1P regulates the secretion and activity of MMP2 and MMP9 via S1P1,3. Both pharmacological inhibitors and siRNA knockdown of MMP2 and MMP9 could attenuate S1P-induced invasion. Additionally, we show that calpains and Rac1 mediate S1P-induced secretion of MMP2 and MMP9. In conclusion, MMP2 and MMP9 participate in S1P-evoked follicular ML-1 thyroid cancer cell invasion.


Assuntos
Adenocarcinoma Folicular/patologia , Lisofosfolipídeos/farmacologia , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Esfingosina/análogos & derivados , Neoplasias da Glândula Tireoide/patologia , Adenocarcinoma Folicular/enzimologia , Calpaína/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Humanos , Esfingosina/farmacologia , Neoplasias da Glândula Tireoide/enzimologia , Proteínas rac1 de Ligação ao GTP/metabolismo
4.
PLoS One ; 8(6): e66189, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23824493

RESUMO

Sphingosine-1-phosphate (S1P) is a bioactive lipid, which regulates several cancer-related processes including migration and angiogenesis. We have previously shown S1P to induce migration of follicular ML-1 thyroid cancer cells. Hypoxia-induced factor-1 (HIF-1) is an oxygen-sensitive transcription factor, which adapts cells to hypoxic conditions through increased survival, motility and angiogenesis. Due to these properties and its increased expression in response to intratumoral hypoxia, HIF-1 is considered a significant regulator of tumor biology. We found S1P to increase expression of the regulatory HIF-1α subunit in normoxic ML-1 cells. S1P also increased HIF-1 activity and expression of HIF-1 target genes. Importantly, inhibition or knockdown of HIF-1α attenuated the S1P-induced migration of ML-1 cells. S1P-induced HIF-1α expression was mediated by S1P receptor 3 (S1P3), Gi proteins and their downstream effectors MEK, PI3K, mTOR and PKCßI. Half-life measurements with cycloheximide indicated that S1P treatment stabilized the HIF-1α protein. On the other hand, S1P activated translational regulators eIF-4E and p70S6K, which are known to control HIF-1α synthesis. In conclusion, we have identified S1P as a non-hypoxic regulator of HIF-1 activity in thyroid cancer cells, studied the signaling involved in S1P-induced HIF-1α expression and shown S1P-induced migration to be mediated by HIF-1.


Assuntos
Adenocarcinoma Folicular/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Lisofosfolipídeos/metabolismo , Esfingosina/análogos & derivados , Adenocarcinoma Folicular/patologia , Linhagem Celular Tumoral , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/biossíntese , MAP Quinase Quinase Quinases/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteína Quinase C beta/metabolismo , Transdução de Sinais , Esfingosina/metabolismo , Serina-Treonina Quinases TOR/metabolismo
5.
Endocrinology ; 151(7): 2994-3005, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20501673

RESUMO

Sphingosine 1-phosphate (S1P) and vascular endothelial growth factor receptor 2 (VEGFR-2) signaling have been shown to integrate in many biological processes. The follicular thyroid carcinoma cell line ML-1 expresses VEGFR-2 and secretes substantial amounts of both vascular endothelial growth factor (VEGF)-A and VEGF-C. ML-1 cells also express S1P-receptors (S1P(1-3,5)). S1P is able to phosphorylate VEGFR-2, and inhibiting VEGFR-2 attenuates S1P-induced migration and down-regulates S1P(1) expression in ML-1 cells. In the present study, we focused on the interactions between S1P(1) and VEGFR-2. We show that S1P receptors form complexes with VEGFR-2 and that the S1P(1)/VEGFR-2 complex associates with protein kinase C (PKC)-alpha and ERK1/2. Furthermore, the complex evokes bidirectional signaling since the S1P-induced ERK1/2 phosphorylation is sensitive to VEGFR-2 kinase inhibition and VEGF-A-induced ERK1/2 phosphorylation is sensitive to pertussis toxin treatment as well as S1P(1) small interfering RNA (siRNA) treatment. Both S1P- and VEGF-A-induced haptotaxis is sensitive to pertussis toxin treatment and S1P(1) siRNA treatment. Phosphorylation of ERK1/2 evoked by both VEGF-A and the S1P(1) agonist SEW-2871 is inhibited by PKC-alpha and PKC-betaI siRNA. We hypothesize that VEGFR-2 forms a signaling complex with S1P(1), evoking bidirectional signaling regulating both ERK1/2 phosphorylation and haptotaxis of ML-1 cells.


Assuntos
Movimento Celular/efeitos dos fármacos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Proteína Quinase C-alfa/metabolismo , Receptores de Lisoesfingolipídeo/metabolismo , Neoplasias da Glândula Tireoide/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Western Blotting , Linhagem Celular Tumoral , Movimento Celular/genética , Humanos , Imuno-Histoquímica , Imunoprecipitação , Lisofosfolipídeos/farmacologia , Fosforilação/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/genética , Receptores de Lisoesfingolipídeo/agonistas , Receptores de Lisoesfingolipídeo/antagonistas & inibidores , Receptores de Lisoesfingolipídeo/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Esfingosina/análogos & derivados , Esfingosina/farmacologia , Neoplasias da Glândula Tireoide/genética , Fator A de Crescimento do Endotélio Vascular/farmacologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/agonistas , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...