Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Biomed Pharmacother ; 137: 111331, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33578235

RESUMO

SCOPE: To investigate the effect of Qingjie Fuzheng Granule (QFG) on lymphangiogenesis and lymphatic metastasis in colorectal cancer. METHODS: The effects of QFG on the expression and secretion of vascular endothelial growth factor-C (VEGF-C) in HCT-116 cells were investigated both in vitro and in vivo. HCT-116 cells were treated with different concentrations (0.2, 0.5, and 1.0 mg/mL) of QFG. The VEGF-C expression level was determined using RT-qPCR and western blotting, and the VEGF-C concentration in supernatant was measured by ELISA. Tumor xenograft models of HCT-116 cells were generated using BALB/c nude mice, and the mice were randomly divided into a control group (gavaged with normal saline) and QFG group (gavaged with 2 g/kg QFG). The effect of QFG on tumor growth was evaluated by comparing the volume and weight of tumors between two groups. Immunohistochemistry (IHC) and RT-qPCR were performed to detect the expression levels of VEGF-C, vascular endothelial growth factor receptor 3 (VEGFR-3), and LYVE-1 (lymphatic vessel endothelial hyaluronan receptor 1). ELISA was performed to measure the concentration of serum VEGF-C. TMT proteomics technology and Reactome pathway analysis were used to explore the mechanism of QFG inhibiting lymphangiogenesis in tumor. The VEGF-C (5 ng/mL)-stimulated human lymphatic endothelial cell (HLEC) model was conducted to evaluate the effect of QFG on lymphangiogenesis in vitro. The model cells were treated with different concentrations (0.2, 0.5, and 1.0 mg/mL) of QFG. Cell viability was then determined using an MTT assay. The cell migration, invasion, and tube-formation ability were analyzed using transwell migration, matrigel invasion and tube formation assays, respectively. The underlying mechanism was uncovered, the levels of VEGFR-3, matrix metalloproteinase 2 (MMP-2), matrix metalloproteinase 9 (MMP-9), p-PI3K/PI3K, p-AKT/AKT and p-mTOR/ mTOR were detected using western blotting. RESULTS: QFG significantly reduced VEGF-C expression and secretion in HCT-116 cells. QFG evidently suppressed in vivo tumor growth and the expression of VEGF-C, VEGFR-3, and LYVE-1. The serum VEGF-C level was also reduced by QFG. Moreover, TMT proteomics technology and Reactome pathway analysis identified 95 differentially expressed protein and multiple enriched pathway about matrix metalloproteinase and extracellular matrix, which is direct associate with lymphangiogenesis. In vitro experiment, QFG inhibited the viability, migration, invasion and tube formation of HLECs. Additionally, QFG reduced the VEGFR-3, MMP-2, MMP-9 expression levels, and the p-PI3K/PI3K, p-AKT/AKT, p-mTOR/ mTOR ratios. CONCLUSION: QFG can exert its effect on both tumor cells and HLECs, exhibiting ani- lymphangiogenesis in colorectal cancer via the VEGF-C/VEGFR-3 dependent PI3K/AKT pathway pathway.


Assuntos
Inibidores da Angiogênese/farmacologia , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/patologia , Linfangiogênese/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Animais , Linhagem Celular , Células Endoteliais/efeitos dos fármacos , Humanos , Inibidores de Metaloproteinases de Matriz/farmacologia , Proteínas de Membrana Transportadoras/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Proteína Oncogênica v-akt/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/efeitos dos fármacos , Fator C de Crescimento do Endotélio Vascular/efeitos dos fármacos , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/efeitos dos fármacos
2.
Pain Pract ; 15(6): 530-7, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24807396

RESUMO

BACKGROUND: Vascular endothelial growth factor-C (VEGF-C), tumor necrosis factor-α (TNF-α), and interleukin-1ß(IL-1ß) have been shown to be associated with the recurrence and metastasis of breast cancer after surgery. This study tested the hypothesis that patients undergoing surgery for breast cancer, who received postoperative analgesia with flurbiprofen axetil combined with small doses of fentanyl (FA), exhibited reduced levels of VEGF-C, TNF-α, and IL-1ß compared with those patients receiving fentanyl alone (F). METHOD: Forty-women with primary breast cancer undergoing a modified radical mastectomy were randomized to receive postoperative analgesia with flurbiprofen axetil combined with fentanyl or fentanyl alone. Venous blood was sampled before anesthesia, at the end of surgery, and at 48 hours after surgery, and the serum was analyzed. The primary endpoint was changes in the VEGF-C concentrations in serum. RESULTS: Group FA patients reported similar analgesic effects as group F patients at 2, 24, and 48 hours. At 48 hours, mean postoperative concentrations of VEGF-C in group F patients were higher than in group FA patients, 730.9 versus. 354.1 pg/mL (P = 0.003), respectively. The mean postoperative concentrations of TNF-α in group F patients were also higher compared with group FA patients 27.1 vs. 15.8 pg/mL (P = 0.005). Finally, the mean postoperative concentrations of IL-1ß in group F were also significantly higher than in group FA 497.5 vs. 197.7 pg/mL (P = 0.001). CONCLUSION: In patients undergoing a mastectomy, postoperative analgesia with flurbiprofen axetil, combined with fentanyl, were associated with decreases in serum concentrations of VEGF-C, TNF-α, and IL-1ß compared with patients receiving doses of only fentanyl.


Assuntos
Analgésicos/administração & dosagem , Fentanila/administração & dosagem , Flurbiprofeno/análogos & derivados , Interleucina-1beta/sangue , Fator de Necrose Tumoral alfa/sangue , Fator C de Crescimento do Endotélio Vascular/sangue , Adulto , Idoso , Neoplasias da Mama/sangue , Neoplasias da Mama/cirurgia , Quimioterapia Combinada/métodos , Feminino , Flurbiprofeno/administração & dosagem , Humanos , Interleucina-1beta/efeitos dos fármacos , Pessoa de Meia-Idade , Dor Pós-Operatória/prevenção & controle , Fator de Necrose Tumoral alfa/efeitos dos fármacos , Fator C de Crescimento do Endotélio Vascular/efeitos dos fármacos
3.
Braz. j. med. biol. res ; 47(12): 1021-1028, 12/2014. tab, graf
Artigo em Inglês | LILACS | ID: lil-727663

RESUMO

DNA hypomethylation may activate oncogene transcription, thus promoting carcinogenesis and tumor development. S-adenosylmethionine (SAM) is a methyl donor in numerous methylation reactions and acts as an inhibitor of intracellular demethylase activity, which results in hypermethylation of DNA. The main objectives of this study were to determine whether DNA hypomethylation correlated with vascular endothelial growth factor-C (VEGF-C) expression, and the effect of SAM on VEGF-C methylation and gastric cancer growth inhibition. VEGF-C expression was assayed by Western blotting and RT-qPCR in gastric cancer cells, and by immunohistochemistry in tumor xenografts. VEGF-C methylation was assayed by bisulfite DNA sequencing. The effect of SAM on cell apoptosis was assayed by flow cytometry analyses and its effect on cancer growth was assessed in nude mice. The VEGF-C promoters of MGC-803, BGC-823, and SGC-7901 gastric cancer cells, which normally express VEGF-C, were nearly unmethylated. After SAM treatment, the VEGF-C promoters in these cells were highly methylated and VEGF-C expression was downregulated. SAM also significantly inhibited tumor growth in vitro and in vivo. DNA methylation regulates expression of VEGF-C. SAM can effectively induce VEGF-C methylation, reduce the expression of VEGF-C, and inhibit tumor growth. SAM has potential as a drug therapy to silence oncogenes and block the progression of gastric cancer.


Assuntos
Animais , Humanos , Masculino , Antineoplásicos/farmacologia , Metilação de DNA/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , S-Adenosilmetionina/farmacologia , Neoplasias Gástricas/tratamento farmacológico , Fator C de Crescimento do Endotélio Vascular/metabolismo , Apoptose/efeitos dos fármacos , Western Blotting , Linhagem Celular Tumoral , Carcinogênese/efeitos dos fármacos , Metilação de DNA/genética , Citometria de Fluxo , Regulação Neoplásica da Expressão Gênica/fisiologia , Xenoenxertos/efeitos dos fármacos , Imuno-Histoquímica , Camundongos Nus , Oncogenes/efeitos dos fármacos , Regiões Promotoras Genéticas/efeitos dos fármacos , Reação em Cadeia da Polimerase em Tempo Real , RNA Mensageiro/análise , Neoplasias Gástricas/metabolismo , Fator C de Crescimento do Endotélio Vascular/efeitos dos fármacos , Fator C de Crescimento do Endotélio Vascular/genética
4.
Braz J Med Biol Res ; 47(12): 1021-8, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25387667

RESUMO

DNA hypomethylation may activate oncogene transcription, thus promoting carcinogenesis and tumor development. S-adenosylmethionine (SAM) is a methyl donor in numerous methylation reactions and acts as an inhibitor of intracellular demethylase activity, which results in hypermethylation of DNA. The main objectives of this study were to determine whether DNA hypomethylation correlated with vascular endothelial growth factor-C (VEGF-C) expression, and the effect of SAM on VEGF-C methylation and gastric cancer growth inhibition. VEGF-C expression was assayed by Western blotting and RT-qPCR in gastric cancer cells, and by immunohistochemistry in tumor xenografts. VEGF-C methylation was assayed by bisulfite DNA sequencing. The effect of SAM on cell apoptosis was assayed by flow cytometry analyses and its effect on cancer growth was assessed in nude mice. The VEGF-C promoters of MGC-803, BGC-823, and SGC-7901 gastric cancer cells, which normally express VEGF-C, were nearly unmethylated. After SAM treatment, the VEGF-C promoters in these cells were highly methylated and VEGF-C expression was downregulated. SAM also significantly inhibited tumor growth in vitro and in vivo. DNA methylation regulates expression of VEGF-C. SAM can effectively induce VEGF-C methylation, reduce the expression of VEGF-C, and inhibit tumor growth. SAM has potential as a drug therapy to silence oncogenes and block the progression of gastric cancer.


Assuntos
Antineoplásicos/farmacologia , Metilação de DNA/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , S-Adenosilmetionina/farmacologia , Neoplasias Gástricas/tratamento farmacológico , Fator C de Crescimento do Endotélio Vascular/metabolismo , Animais , Apoptose/efeitos dos fármacos , Western Blotting , Carcinogênese/efeitos dos fármacos , Linhagem Celular Tumoral , Metilação de DNA/genética , Citometria de Fluxo , Regulação Neoplásica da Expressão Gênica/fisiologia , Xenoenxertos/efeitos dos fármacos , Humanos , Imuno-Histoquímica , Masculino , Camundongos Nus , Oncogenes/efeitos dos fármacos , Regiões Promotoras Genéticas/efeitos dos fármacos , RNA Mensageiro/análise , Reação em Cadeia da Polimerase em Tempo Real , Neoplasias Gástricas/metabolismo , Fator C de Crescimento do Endotélio Vascular/efeitos dos fármacos , Fator C de Crescimento do Endotélio Vascular/genética
5.
Int J Clin Exp Pathol ; 6(4): 586-97, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23573305

RESUMO

BACKGROUND: Vascular endothelial growth factor (VEGF)-A and VEGF-C are two important molecules involving in tumor development and metastasis via angiogenesis and lymphangiogenesis. However, the combined effect of VEGF-A and VEGF-C on the growth of gastric cancer (GC) is not clear. METHODS: The correlations of VEGF-A and VEGF-C expressions with clinicopathologic parameters and prognosis were evaluated in patients with GC. Furthermore, lentivirus-mediated RNA interfering (RNAi) targeting VEGF-A and/or VEGF-C was employed to silence their expressions in SGC7901 GC cell line. Cell proliferation and apoptosis were measured in vitro. Suppressive effect lentivirus-mediated VEGF-A and/or VEGF-C silencing on GC growth was evaluated in GC bearing mice. RESULTS: The patients with high expression of both VEGF-A and VEGF-C (A+C+) had larger tumor size, higher peritumoral lymphatic vessel density(P-LVD), microvessel density(MVD), lymphatic vessel invasion (LVI), lymph node(LN) metastasis, and worse prognosis than those with low expression of both VEGF-A and VEGF-C (P<0.05). Lentivirus-mediated RNAi significantly reduced the mRNA and protein expression of VEGF-A and VEGF-C in the SGC7901 cells. The Lenti-miRNA-VEGF-A+VEGF-C significantly inhibited the cell proliferation and tumor growth, compared with Lenti-miRNA-VEGF-A or Lenti-miRNA-VEGF-C (P<0.05). In addition, Lenti-miRNA- VEGF-A+VEGF-C markedly lowered the tumor size in vivo in comparison with Lenti-miRNA-VEGF-A or Lenti-miRNA-VEGF-C (P<0.05). CONCLUSION: Expressions of both VEGF-A and VEGF-C predict worse prognosis of GC patients. Combined silencing of VEGF-A and VEGF-C markedly suppresses cancer growth than silencing of VEGF-A or VEGF-C. Thus, to inhibit the expressions of VEGF-A and VEGF-C may become a novel strategy for the treatment of GC.


Assuntos
Inativação Gênica , RNA Interferente Pequeno/farmacologia , Neoplasias Gástricas/diagnóstico , Neoplasias Gástricas/metabolismo , Fator A de Crescimento do Endotélio Vascular/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fator C de Crescimento do Endotélio Vascular/efeitos dos fármacos , Fator C de Crescimento do Endotélio Vascular/metabolismo , Adenocarcinoma/diagnóstico , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Apoptose/efeitos dos fármacos , Biomarcadores Tumorais/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Progressão da Doença , Feminino , Seguimentos , Humanos , Técnicas In Vitro , Lentivirus/genética , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Pessoa de Meia-Idade , Metástase Neoplásica/tratamento farmacológico , Neovascularização Patológica/tratamento farmacológico , Prognóstico , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/uso terapêutico , Neoplasias Gástricas/patologia , Fator A de Crescimento do Endotélio Vascular/genética , Fator C de Crescimento do Endotélio Vascular/genética , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Am Surg ; 76(6): 614-7, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20583517

RESUMO

Flap necrosis is one of the major complications of reconstructive surgery and sildenafil citrate has been shown to decrease flap necrosis in preclinical animal models. However, the mechanisms underlying sildenafil's therapeutic efficacy are not known. As with other phosphodiesterase 5 selective inhibitors, sildenafil causes vasodilation and enhanced blood flow. In addition, sildenafil can also alter gene expression. This study is designed to test the hypothesis that increased expression of angiogenic growth factors may be responsible for therapeutic efficacy of sildenafil. A modified McFarlane flap measuring 3 x 10 cm was created on the dorsal skin of male Sprague-Dawley rats. For growth factor expression experiment, rats were administered either vehicle or sildenafil 10 mg/Kg intraperitoneal (IP). Ribonucleic acid (RNA) extracted from skin flap was analyzed to assess the messenger ribonucleic acid (mRNA) levels of different angiogenic growth factors. For skin flap viability experiment, fibrin film impregnated with vehicle, fibroblast growth factor (FGF) (5.0 microg) or vascular endothelial growth factor (VEGF) (2.0 microg) was applied to the wound. The skin flap was then returned to its native position and stapled in place. Total affected area (area of necrosis and blood flow stasis) of each rat on postoperative day 14 was analyzed with orthogonal polarization spectral imaging. Daily systemic treatment with sildenafil significantly (P < 0.05) increased the expression of FGF1 and FGF Receptor 3 on postoperative day 3 by 5.08- and 4.78-fold, respectively. In addition, sildenafil increased the expression of VEGF-A, VEGF-B, and VEGF-C by 2.66-, 2.02-, and 2.00-fold, respectively. Subcutaneous treatment with FGF but not VEGF-A tended to decrease total affected area in rats. These data demonstrate that sildenafil altered the expression of FGF and VEGF. Altered expression of growth factors may be, at least partly, responsible for the beneficial effects of sildenafil citrate on skin viability.


Assuntos
Inibidores de Fosfodiesterase/farmacologia , Piperazinas/farmacologia , Sulfonas/farmacologia , Retalhos Cirúrgicos/irrigação sanguínea , Fator A de Crescimento do Endotélio Vascular/efeitos dos fármacos , Animais , Fator 1 de Crescimento de Fibroblastos/metabolismo , Masculino , Neovascularização Fisiológica/efeitos dos fármacos , Análise de Sequência com Séries de Oligonucleotídeos , Purinas/farmacologia , Ratos , Ratos Sprague-Dawley , Citrato de Sildenafila , Retalhos Cirúrgicos/fisiologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fator B de Crescimento do Endotélio Vascular/efeitos dos fármacos , Fator B de Crescimento do Endotélio Vascular/metabolismo , Fator C de Crescimento do Endotélio Vascular/efeitos dos fármacos , Fator C de Crescimento do Endotélio Vascular/metabolismo
7.
Endothelium ; 15(5-6): 265-75, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19065318

RESUMO

The role of estrogen replacement therapy in postmenopausal women remains controversial. The authors hypothesized that contradictory results with estrogen therapy may be explained by estrogen's potent proangiogenic property, which could be protective in women without atherosclerotic disease but in the presence of chronic inflammation, could lead to destabilization of atherosclerotic plaques. The authors thus examined the interaction between 17beta-estradiol (E2) and the inflammatory cytokine tumor necrosis factor alpha (TNFalpha) in an early stage of angiogenesis. Human umbilical endothelial cells were grown to confluence. Migration was assessed with a wound assay and proliferation was assessed with 5-bromo-2'-deoxyuridine (BrDU). Cells were treated with medium alone, TNFalpha at 0.3, 1, or 20 ng/ml, E2 at 20 nM, or the combination of E2 and TNFalpha. The authors used real-time polymerase chain reaction (PCR) to measure changes in expression of the angiogenesis genes angiopoeitin-2 (Ang-2), vacular endothelial growth factor (VEGF)-A and -C, and interleukin (IL)-8. A large dose of TNFalpha (20 ng/ml) inhibited healing at 24 to 48 h and the addition of E2 preserved some healing. E2 by itself doubled migration, with only a minimal effect on proliferation. A low dose of TNFalpha (0.3 ng/ml) had no effect on migration, 1.0 ng/ml moderately increased it, but the addition of E2 to both doses of TNFalpha increased migration. There was no change in migration when cells were pretreated with E2 and given TNFalpha after wounding, whereas pretreatment with TNFalpha followed by E2 significantly increased wound healing. The nitric oxide synthase (NOS) inhibitor N-nitro-l-arginine-methyl ester (l-NAME) completely blocked the E2 effect on migration. TNFalpha (0.3 and 1.0 ng/ml) increased expression of VEGF-C (2.8 +/- 0.1- and 2.5 +/- 0.2-fold, respectively) and IL-8 (32.8 +/- 1.2- and 42.7 +/- 3.6-fold, respectively) mRNA, but E2 had no significant effect on these molecules. E2 increases the angiogenic activity of TNFalpha. This could potentially worsen the stability of complex atherosclerotic plaques and increase cardiovascular events.


Assuntos
Movimento Celular/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Estrogênios/farmacologia , Neovascularização Fisiológica/efeitos dos fármacos , Fator de Necrose Tumoral alfa/farmacologia , Angiopoietina-2/metabolismo , Aterosclerose/induzido quimicamente , Aterosclerose/metabolismo , Aterosclerose/fisiopatologia , Movimento Celular/fisiologia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Relação Dose-Resposta a Droga , Interações Medicamentosas/fisiologia , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Estradiol/metabolismo , Estradiol/farmacologia , Terapia de Reposição de Estrogênios/efeitos adversos , Estrogênios/metabolismo , Feminino , Humanos , Interleucina-8/efeitos dos fármacos , Interleucina-8/genética , Interleucina-8/metabolismo , Neovascularização Fisiológica/fisiologia , Óxido Nítrico Sintase Tipo III/antagonistas & inibidores , Óxido Nítrico Sintase Tipo III/metabolismo , RNA Mensageiro/efeitos dos fármacos , RNA Mensageiro/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Fator C de Crescimento do Endotélio Vascular/efeitos dos fármacos , Fator C de Crescimento do Endotélio Vascular/genética , Fator C de Crescimento do Endotélio Vascular/metabolismo , Cicatrização/efeitos dos fármacos , Cicatrização/fisiologia
8.
Pharmacology ; 82(2): 148-55, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18667841

RESUMO

We have previously reported that dihydroartemisinin is found to have a potent ability in influencing lymphatic endothelial cell migration and tube formation. In this study, we investigated the effect of artemisinin on tumor growth, lymphangiogenesis, metastasis and survival in mouse Lewis lung carcinoma (LLC) models. We found that orally administered artemisinin inhibited lymph node and lung metastasis and prolonged survival without retarding tumor growth. Consistent with the decrease in lymph node metastasis, tumor lymphangiogenesis and expression of vascular endothelial growth factor C (VEGF-C) was significantly decreased in artemisinin-treated mice, as compared to control mice. Furthermore, IL-1beta-induced p38 mitogen-activated protein kinase (MAPK) activation and upregulation of VEGF-C mRNA and protein in LLC cells was also suppressed by artemisinin or by the p38 MAPK inhibitor SB-203580, suggesting that p38 MAPK could serve as a mediator of proinflammatory cytokine-induced VEGF-C expression. These data indicate that artemisinin may be useful for the prevention of lymph node metastasis by downregulating VEGF-C and reducing tumor lymphangiogenesis.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Artemisininas/farmacologia , Carcinoma Pulmonar de Lewis/tratamento farmacológico , Fator C de Crescimento do Endotélio Vascular/efeitos dos fármacos , Administração Oral , Animais , Carcinoma Pulmonar de Lewis/patologia , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Neoplasias Pulmonares/prevenção & controle , Neoplasias Pulmonares/secundário , Linfangiogênese/efeitos dos fármacos , Metástase Linfática/prevenção & controle , Camundongos , Camundongos Endogâmicos C57BL , Metástase Neoplásica/prevenção & controle , Taxa de Sobrevida , Fator C de Crescimento do Endotélio Vascular/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
9.
Kidney Int ; 71(8): 771-7, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17299523

RESUMO

Lymphatic complications are common side effects of mammalian target of rapamycin (mTOR) inhibitor-based immunosuppression in kidney transplantation. Therefore, we investigated whether the mTOR inhibitor rapamycin, besides its known antihemangiogenic effect, also impedes regenerative lymphangiogenesis. In a murine skin flap model, rapamycin impaired recovery of lymphatic flow across surgical incisions resulting in prolonged wound edema in these animals. Importantly, the antilymphangiogenic effect of rapamycin was not related to a general inhibition of wound healing as demonstrated an in vivo Matrigeltrade mark lymphangiogenesis assay and a model of lymphangioma. Rapamycin concentrations as low as 1 ng/ml potently inhibited vascular endothelial growth factor (VEGF)-C driven proliferation and migration, respectively, of isolated human lymphatic endothelial cells (LECs) in vitro. Mechanistically, mTOR inhibition impairs downstream signaling of VEGF-A as well as VEGF-C via mTOR to the p70S6 kinase in LECs. In conclusion, we provide extensive experimental evidence for an antilymphangiogenic activity of mTOR inhibition suggesting that the early use of mTOR inhibitor following tissue injury should be avoided. Conversely, the antilymphangiogenic properties of rapamycin and its derivates may provide therapeutic value for the prevention and treatment of malignancies, respectively.


Assuntos
Células Endoteliais/efeitos dos fármacos , Endotélio Linfático/efeitos dos fármacos , Imunossupressores/farmacologia , Linfangiogênese/efeitos dos fármacos , Proteínas Quinases/efeitos dos fármacos , Sirolimo/farmacologia , Animais , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Colágeno , Combinação de Medicamentos , Imunossupressores/uso terapêutico , Laminina , Linfangioma/tratamento farmacológico , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias Peritoneais/tratamento farmacológico , Fosforilação/efeitos dos fármacos , Proteoglicanas , Proteínas Quinases S6 Ribossômicas 70-kDa/efeitos dos fármacos , Sirolimo/uso terapêutico , Serina-Treonina Quinases TOR , Fator A de Crescimento do Endotélio Vascular/efeitos dos fármacos , Fator C de Crescimento do Endotélio Vascular/efeitos dos fármacos
11.
Am J Respir Crit Care Med ; 174(3): 326-30, 2006 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-16690974

RESUMO

RATIONALE: In mice, vascular endothelial growth factor-C (VEGF-C) plays an important role in development of the lymphatic system and in pathogenesis of pulmonary inflammation. Its role in development of the lymphatic system in human lung and in lung injury in newborns remains unclear. OBJECTIVES: We studied the role of VEGF-C in developing human lung, and in acute and chronic lung injury in preterm infants. METHODS: Included in the immunohistochemistry study were 10 fetuses, 15 control neonates without primary lung disease, 15 preterm infants with respiratory distress syndrome, and 8 infants with bronchopulmonary dysplasia. Tracheal aspirate fluid samples of intubated very-low-birth-weight infants during Postnatal Weeks 1-5 were analyzed with ELISA. RESULTS: Bronchiolar staining for VEGF-C was observed in all 48 samples. Alveolar epithelial staining was seen in most fetuses (8/10). In addition, staining was observed in alveolar macrophages in bronchopulmonary dysplasia (4/8), and late respiratory distress syndrome (2/7). VEGF receptor-3 (VEGFR-3) staining was observed in lymphatic endothelium adjacent to vascular endothelium. VEGF-C was expressed consistently in tracheal aspirate fluid, being highest during the first 2 postnatal days. Antenatal administration of glucocorticoids was associated with higher VEGF-C in tracheal aspirate fluid. CONCLUSIONS: The pattern of pulmonary VEGF-C and VEGFR-3 protein expression and consistent VEGF-C protein appearance in tracheal aspirate fluid in human preterm infants indicate a role for VEGF-C in the physiologic development of the lymphatic system of the lung.


Assuntos
Feto/metabolismo , Recém-Nascido de muito Baixo Peso/metabolismo , Pulmão/metabolismo , Traqueia/metabolismo , Fator C de Crescimento do Endotélio Vascular/metabolismo , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Brônquios/patologia , Líquido da Lavagem Broncoalveolar , Feminino , Desenvolvimento Fetal , Idade Gestacional , Humanos , Imuno-Histoquímica , Recém-Nascido , Recém-Nascido Prematuro/metabolismo , Pulmão/efeitos dos fármacos , Pulmão/embriologia , Pulmão/crescimento & desenvolvimento , Masculino , Análise de Regressão , Síndrome do Desconforto Respiratório do Recém-Nascido/etiologia , Fator C de Crescimento do Endotélio Vascular/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...