Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Environ Sci Technol ; 2024 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-38344765

RESUMO

Volatile sulfur compounds, such as dimethyl sulfide (DMS), carbonyl sulfide (OCS), and carbon disulfide (CS2), have significant implications for both atmospheric chemistry and climate change. Despite the crucial role of oceans in regulating their atmospheric budgets, our comprehension of their cycles in seawater remains insufficient. To address this gap, a field investigation was conducted in the western North Pacific to clarify the sources, sinks, and biogeochemical controls of these gases in two different marine environments, including relatively eutrophic Kuroshio-Oyashio extension (KOE) and oligotrophic North Pacific subtropical gyre. Our findings revealed higher concentrations of these gases in both seawater and the atmosphere in the KOE compared to the subtropical gyre. In the KOE, nutrient-rich upwelling stimulated rapid DMS biological production, while reduced seawater temperatures hindered the removal of OCS and CS2, leading to their accumulation. Furthermore, we have quantitatively evaluated the relative contribution of each pathway to the source and sink of DMS, OCS, and CS2 within the mixed layer and identified vertical exchange as a potential sink in most cases, transporting substantial amounts of these gases from the mixed layer to deeper waters. This research advances our understanding of sulfur gas source-sink dynamics in seawater, contributing to the assessment of their marine emissions and atmospheric budgets.

2.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 30(3): 844-850, 2022 Jun.
Artigo em Zh | MEDLINE | ID: mdl-35680815

RESUMO

OBJECTIVE: To provide a research basis for a safe and effective cell therapy for ß-thalassemia through optimization of HS4 region of the third generation lentiviral vector for stable expression of ß-globin. METHODS: The human ß-globin HS4 region in the third generation lentiviral expression vector was optimized to construct the lenti-HBB, and the transcription and translation of ß-globin gene were analyzed by RT-PCR and Western blot after the transduction of lenti-HBB in MEL cell line. Furthermore, the erythroid differentiation of CD34+ cells which were transduced lentiviral virus carrying human ß-globin from normal human umbilical cord blood cells and peripheral blood cells of patients with ß-thalassemia major were confirmed by colony formation assay, cell smear assay and flow cytometry. The safety and effectiveness of the optimized lenti-HBB were verified by NSG mouse in vivo test. RESULTS: The human ß-globin was expressed stably in the MEL cells, and CD34+ cells from health umbilical cord blood as well as PBMC from patient with ß-thalassemia major transduced with lenti-HBB could be differentiated to mature red blood cells. The ß-globin expression and differentiation in CD34+ cells were demonstrated successfully in the NSG mouse for about 35 months after post-transplant. CONCLUSION: Stable ß-globin expression through the optimization of HS4 from CD34+ in the third generation lentiviral vector is safe and effective for patients with severe ß-thalassemia and other ß-globin abnormal diseases.


Assuntos
Globinas beta , Talassemia beta , Animais , Terapia Genética , Vetores Genéticos , Humanos , Lentivirus/genética , Leucócitos Mononucleares , Camundongos , Globinas beta/genética , Talassemia beta/genética , Talassemia beta/terapia
3.
Mol Ther ; 18(2): 413-20, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19773745

RESUMO

To enhance the strength of activation afforded by tumor antigen-specific receptors, we investigated the effect of adding combined CD28 and 4-1BB costimulatory signaling domains to a chimeric antigen receptor (CAR) specific for prostate-specific membrane antigen (PSMA). Having transferred receptors encompassing the CD28, 4-1BB, and/or CD3zeta cytoplasmic domains in primary human CD8(+) T cells, we find that the P28BBz receptor, which includes all three signaling domains, is superior to receptors that only include one or two of these domains in promoting cytokine release, in vivo T-cell survival and tumor elimination following intravenous T-cell administration to tumor-bearing severe combined immunodeficient (SCID)/beige mice. Upon in vitro exposure to PSMA, the P28BBZ receptor-induced the strongest PI(3)Kinase/Akt activation and Bcl-X(L) expression, and the least apoptosis in transduced peripheral blood CD8(+) T cells. These findings further support the concept of integrating optimized costimulatory properties into recombinant antigen receptors to augment the survival and function of genetically targeted T cells within the tumor microenvironment.


Assuntos
Antígenos CD28/metabolismo , Linfócitos T CD8-Positivos/imunologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/metabolismo , Proteína bcl-X/metabolismo , Animais , Antígenos de Superfície/imunologia , Antígenos CD28/genética , Células Cultivadas , Citometria de Fluxo , Glutamato Carboxipeptidase II/imunologia , Humanos , Immunoblotting , Camundongos , Camundongos SCID , Células NIH 3T3 , Neoplasias/genética , Neoplasias/terapia , Fosfatidilinositol 3-Quinases/genética , Proteínas Proto-Oncogênicas c-akt/genética , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/genética , Proteína bcl-X/genética
4.
Chin Med J (Engl) ; 132(12): 1435-1440, 2019 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-31205101

RESUMO

BACKGROUND: Previous studies have shown that endogenous T cells play an important role in the prolonged survival time of high-grade glioma (HGG) patients. Our objectives were to investigate the features of T-cell receptor (TCR) repertoires in HGG patients and to elucidate any potential therapeutic value. METHODS: During November 2011 and December 2018, tumor tissues and blood samples of 35 patients with HGG who underwent surgery at Beijing Tiantan Hospital or Beijing Shijitan Hospital were selected after surgery. After isolating DNA from samples, multiple rounds of PCR were performed to establish a DNA immune repertoire (IR). Then, the sequences and frequencies of the complementarity-determining 3 (CDR3) region in TCR beta chain (TRB) were identified by high-throughput sequencing and IR analysis. A survival follow-up was conducted monthly thereafter until December 2018. Finally, the t test and Mann-Whitney test were used to compare statistical differences between two sets of data. RESULTS: The Shannon diversity index (SHDI) of TRB sequences of HGG patients was significantly lower than that of healthy individuals (7.34 vs. 8.45, P = 0.001). The SHDI of TRB sequences of glioblastoma (GBM) patients with more than 16 months survival time was much higher than that of GBM patients with shorter survival times in both tumor tissues (3.48 ±â€Š0.31 vs. 6.21 ±â€Š0.33, t = -5.49, P = 0.002) and blood cells (6.02 ±â€Š0.66 vs. 7.44 ±â€Š0.32, t = -2.20, P = 0.036). In addition, patients achieved a distinctly higher proportion compared to that of healthy individuals in the proportion of TRBV9 and TRBV5 functional regions (9.83% vs. 6.83%, P = 0.001). Surgical tissue from patients who survived more than 16 months yielded a much higher proportion of TRBV4 and TRBV9 regions (7.14% vs. 3.28%, t = 3.18, P = 0.019). In surgical tissues from two GBM patients who survived for longer than 46 months, we found a potentially therapeutic TCR sequence. CONCLUSIONS: HGG patients have less species diversity of TCR repertoires compared with that of healthy individuals. TRBV9 regions in TCRs may be protective factors for long-term survival of GBM patients.


Assuntos
Glioma/genética , Glioma/terapia , Receptores de Antígenos de Linfócitos T/química , Receptores de Antígenos de Linfócitos T/genética , Adulto , Idoso , Feminino , Glioma/metabolismo , Glioma/mortalidade , Humanos , Imunoterapia , Masculino , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase , Fatores de Tempo , Adulto Jovem
5.
Cancer Res ; 65(19): 9080-8, 2005 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-16204083

RESUMO

The genetic transfer of antigen receptors is a powerful approach to rapidly generate tumor-specific T lymphocytes. Unlike the physiologic T-cell receptor, chimeric antigen receptors (CARs) encompass immunoglobulin variable regions or receptor ligands as their antigen recognition moiety, thus permitting T cells to recognize tumor antigens in the absence of human leukocyte antigen expression. CARs encompassing the CD3zeta chain as their activating domain induce T-cell proliferation in vitro, but limited survival. The requirements for genetically targeted T cells to function in vivo are less well understood. We have, therefore, established animal models to assess the therapeutic efficacy of human peripheral blood T lymphocytes targeted to prostate-specific membrane antigen (PSMA), an antigen expressed in prostate cancer cells and the neovasculature of various solid tumors. In vivo specificity and antitumor activity were assessed in mice bearing established prostate adenocarcinomas, using serum prostate-secreted antigen, magnetic resonance, computed tomography, and bioluminescence imaging to investigate the response to therapy. In three tumor models, orthotopic, s.c., and pulmonary, we show that PSMA-targeted T cells effectively eliminate prostate cancer. Tumor eradication was directly proportional to the in vivo effector-to-tumor cell ratio. Serial imaging further reveals that the T cells must survive for at least 1 week to induce durable remissions. The eradication of xenogeneic tumors in a murine environment shows that the adoptively transferred T cells do not absolutely require in vivo costimulation to function. These results thus provide a strong rationale for undertaking phase I clinical studies to assess PSMA-targeted T cells in patients with metastatic prostate cancer.


Assuntos
Imunoterapia Adotiva/métodos , Neoplasias da Próstata/imunologia , Neoplasias da Próstata/terapia , Linfócitos T/imunologia , Animais , Antígenos de Superfície/genética , Antígenos de Superfície/imunologia , Linhagem Celular Tumoral , Epitopos de Linfócito T/genética , Epitopos de Linfócito T/imunologia , Glutamato Carboxipeptidase II/genética , Glutamato Carboxipeptidase II/imunologia , Humanos , Memória Imunológica/imunologia , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/secundário , Neoplasias Pulmonares/terapia , Ativação Linfocitária , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/imunologia , Camundongos , Camundongos SCID , Células NIH 3T3 , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/imunologia , Transdução Genética
6.
Drug Des Devel Ther ; 11: 1905-1915, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28721010

RESUMO

Leptomeningeal metastasis (LM) of high-grade glioma is a highly lethal disease requiring new effective therapeutic measures. For both de novo or relapsed glioma with LM, intrathecal cytarabine chemotherapy is not frequently used for first-line and relapse protocols. We encountered a clinical case demonstrating effective application of cytarabine in high-grade glioma with LM, prompting us to explore the effects of cytarabine on malignant glioma and molecular mechanisms of such effects through in vivo and in vitro experiments. The U87 cell line was selected to represent human glioma for studies. Cell viability was measured by MTT assay, plate colony formation assay, and trypan-blue dye exclusion test. Apoptosis was assessed by flow cytometry. Protein expression levels were detected by Western blot assay and immunohistochemistry. mRNA expression was examined by quantitative real-time reverse transcription polymerase chain reaction. Cytarabine inhibited tumor growth during the in vivo experiment. The present study confirmed that cytarabine inhibits proliferation and promotes apoptosis of U87 cells, and molecular analysis of this effect showed that cytarabine significantly reduces expression of phosphatidylinositol 3-kinase/serine/threonine kinase also known as the protein kinase B/mechanistic target of rapamycin (PI3K/Akt/mTOR) pathway, Ki-67, BCL2, and 4-1BB, and upregulates Bax and cleaved caspase-3. Our findings indicated that intrathecal administration of cytarabine manifests potential in prophylaxis and treatment of malignant glioma with LM. Effective medications for high-grade glioma with LM should contain cytarabine.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Neoplasias Encefálicas/tratamento farmacológico , Citarabina/farmacologia , Glioma/tratamento farmacológico , Glioma/secundário , Neoplasias Meníngeas/tratamento farmacológico , Neoplasias Meníngeas/secundário , Proteína Oncogênica v-akt/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/efeitos dos fármacos , Serina-Treonina Quinases TOR/efeitos dos fármacos , Adolescente , Animais , Apoptose/efeitos dos fármacos , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Intervalo Livre de Doença , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glioma/patologia , Humanos , Masculino , Neoplasias Meníngeas/patologia , Camundongos , Camundongos SCID , Transdução de Sinais , Ensaio Tumoral de Célula-Tronco , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Biochem Pharmacol ; 71(6): 761-71, 2006 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-16426580

RESUMO

ERCC1 is a critical gene within the nucleotide excision repair pathway. Overexpression of ERCC1 through promoter-mediating transcriptional regulation is associated with repair of cisplatin-induced DNA damage and clinical resistance to platinum-chemotherapy. Several transcriptional repressors and activators within the 5'-flanking region of the ERCC1 gene may be involved in the up-regulation of this gene. Minimal sequence within the promoter region required for ERCC1 transcription was analyzed by CAT assay and demonstrated that the region of -220 to -110 is essential to constitutive expression of ERCC1 gene in ovarian cancer cell line A2780/CP70. A more forward upstream region seems to be responsible for cisplatin-induced expression. Study of the functional cis-element in this region by electrophoretic mobility shift assay indicates that a MZF1-like site as well as an AP1-like site responded in a time-dependent manner to cisplatin stimulation with altered binding activities. EMSA with MZF1 ZN1-4 consensus oligonucleotides suggests that the MZF1 N-terminal domain of zinc finger cluster may bind to the MZF1-like site of the ERCC1 promoter region. MZF1 mRNA in A2780/CP70 cells decreased upon cisplatin exposure as analyzed by quantitative PCR, suggesting that MZF1 may mediate cisplatin-invoked gene expression in these cells. Overexpression of MZF1 repressed the ERCC1 promoter activity as determined in co-transfection assay, suggesting that MZF1 might be a repressor of ERCC1 transcription upon cisplatin exposure. In summary, our studies revealed a core promoter region and adjacent drug-responsible region within the ERCC1 promoter. The drug-responsible region contains cis-elements of activator, AP1 and repressor, MZF1. In response to cisplatin treatment, decreased MZF1 and increased AP1 binding activities appear to be the leading mechanism of up-regulation of ERCC1 expression. Our findings imply potential therapeutic strategies to antagonize drug resistant mechanisms in treatment of human ovarian cancer.


Assuntos
Adenocarcinoma/genética , Reparo do DNA , Proteínas de Ligação a DNA/genética , Endonucleases/genética , Neoplasias Ovarianas/genética , Fatores de Transcrição/genética , Dedos de Zinco , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Antineoplásicos/farmacologia , Sequência de Bases , Linhagem Celular Tumoral , Cisplatino/farmacologia , Cisplatino/uso terapêutico , Proteínas de Ligação a DNA/metabolismo , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Endonucleases/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Fatores de Transcrição Kruppel-Like , Dados de Sequência Molecular , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Regiões Promotoras Genéticas/efeitos dos fármacos , RNA Mensageiro/análise , RNA Neoplásico/análise , Fator de Transcrição AP-1/metabolismo , Fatores de Transcrição/metabolismo , Regulação para Cima
8.
Cancer Res ; 62(20): 5689-97, 2002 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-12384526

RESUMO

The p70 S6 kinase (p70(S6k)) is a Ser/Thr kinase that plays an important role in cell growth, transformation, and the transition of the cell cycle in mammalian cells. Because UV radiation has been reported to induce activation of p70(S6k), which is believed to play some role in the carcinogenic effects of sun exposure, the present study investigated the signaling pathways involved in this activation induced by UV radiation in mouse epidermal JB6 Cl41 cells. Exposure of cells to UV radiation led to marked increases in p70(S6k) activity and phosphorylation at Thr(389) and Thr(421)/Ser(424). UV radiation also generated reactive oxygen species as measured by electron spin resonance and by H(2)O(2) and O( minus sign, dot below )(2) fluorescence staining assays in JB6 Cl 41 cells. The scavenging of UV-generated H(2)O(2) by N-acety-L-cyteine (a general antioxidant) or catalase (a specific H(2)O(2) scavenger) inhibited p70(S6k) phosphorylation at Thr(389) and Thr(421)/Ser(424), whereas pretreatment of cells with sodium formate (an.OH radical scavenger) or superoxide dismutase (an O( minus sign, dot below )(2) radical scavenger) did not show any inhibitory effects. Importantly, UV-induced increases in p70(S6k) phosphorylation at Thr(389) and Thr(421)/Ser(424) were dramatically inhibited by pretreatment of cells with rapamycin, LY294002, or PD98059, whereas overexpression of dominant-negative mutants of PKClambda/iota and Akt1 did not inhibit p70(S6k) phosphorylation at Thr(389) and Thr(421)/Ser(424). These results demonstrated that H(2)O(2), phosphatidylinositol 3-kinase, and mammalian target of rapamycin were important players for UV-induced p70(S6k) phosphorylation at Thr(389) and Thr(421)/Ser(424), whereas Akt and atypical protein kinase C were not involved in this activation. The role of H(2)O(2) in p70(S6k) phosphorylation at Thr(389) and Thr(421)/Ser(424) was further supported by the findings that treatment of cells with H(2)O(2) also caused p70(S6k) phosphorylation at Thr(389) and Thr(421)/Ser(424).


Assuntos
Peróxido de Hidrogênio/metabolismo , Proteína Quinase C/metabolismo , Proteínas Quinases/metabolismo , Proteínas Serina-Treonina Quinases , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Animais , Células Cultivadas , Espectroscopia de Ressonância de Spin Eletrônica , Indução Enzimática/efeitos da radiação , Camundongos , Fosforilação/efeitos da radiação , Proteínas Proto-Oncogênicas c-akt , Espécies Reativas de Oxigênio/metabolismo , Proteínas Quinases S6 Ribossômicas 70-kDa/biossíntese , Serina/metabolismo , Transdução de Sinais/efeitos da radiação , Pele/citologia , Pele/metabolismo , Pele/efeitos da radiação , Serina-Treonina Quinases TOR , Treonina/metabolismo , Raios Ultravioleta
9.
Nat Med ; 15(7): 808-13, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19525962

RESUMO

Self-renewing cell populations such as hematopoietic stem cells and memory B and T lymphocytes might be regulated by shared signaling pathways. The Wnt-beta-catenin pathway is an evolutionarily conserved pathway that promotes hematopoietic stem cell self-renewal and multipotency by limiting stem cell proliferation and differentiation, but its role in the generation and maintenance of memory T cells is unknown. We found that induction of Wnt-beta-catenin signaling by inhibitors of glycogen sythase kinase-3beta or the Wnt protein family member Wnt3a arrested CD8(+) T cell development into effector cells. By blocking T cell differentiation, Wnt signaling promoted the generation of CD44(low)CD62L(high)Sca-1(high)CD122(high)Bcl-2(high) self-renewing multipotent CD8(+) memory stem cells with proliferative and antitumor capacities exceeding those of central and effector memory T cell subsets. These findings reveal a key role for Wnt signaling in the maintenance of 'stemness' in mature memory CD8(+) T cells and have major implications for the design of new vaccination strategies and adoptive immunotherapies.


Assuntos
Linfócitos T CD8-Positivos/fisiologia , Células-Tronco Hematopoéticas/fisiologia , Memória Imunológica , Transdução de Sinais/fisiologia , Proteínas Wnt/fisiologia , Animais , Linfócitos T CD8-Positivos/citologia , Diferenciação Celular , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Glicogênio Sintase Quinase 3 beta , Fator 1-alfa Nuclear de Hepatócito , Receptores de Hialuronatos/análise , Selectina L/análise , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Fator 1 de Transcrição de Linfócitos T/fisiologia , beta Catenina/fisiologia
10.
Biochem Biophys Res Commun ; 358(1): 92-8, 2007 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-17470361

RESUMO

Cisplatin (CDDP) and its analogues are widely used for the treatment of a variety of human solid tumors. However, the molecular mechanism of its action remains to be understood. Vascular endothelial growth factor (VEGF) is a potent inducer of angiogenesis and is upregulated in many human cancers. In this study we demonstrated that CDDP-inhibited VEGF expression in human ovarian cancer cells. We found that CDDP inhibited the VEGF reporter activity in a dose-dependent manner, indicating that CDDP-inhibited transcriptional activation of VEGF. We also found that: (1) luciferase activity mediated by the VEGF reporter containing a mutation of the HIF-1 binding site was much lower than that of the reporter containing a wild-type HIF-1 binding site in ovarian cancer cells, thus confirming that HIF-1 is a major transcriptional regulator of VEGF expression; and that (2) CDDP greatly inhibited VEGF reporter activity containing the wild-type but not the mutant HIF-1 binding site. This result indicates that CDDP-inhibited VEGF transcriptional activation specifically by decreasing HIF-1 activity. Co-transfection of a dominant negative construct of HIF-1 inhibited VEGF reporter activity in ovarian cancer cells. CDDP-inhibited VEGF transcriptional activation specifically through the expression of HIF-1alpha, but not HIF-1beta. We demonstrated that VEGF receptor KDR was expressed in ovarian cancer cells, and that CDDP-inhibited VEGF expression was linked with cellular apoptosis, which was rescued by VEGF treatment. These results suggest a novel mechanism of CDDP's anti-tumor activity in ovarian cancer cells via HIF-1 expression and VEGF transcriptional activation.


Assuntos
Antineoplásicos/farmacologia , Apoptose , Cisplatino/farmacologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Translocador Nuclear Receptor Aril Hidrocarboneto/metabolismo , Linhagem Celular Tumoral , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Luciferases/genética , Luciferases/metabolismo , Mutação , Neoplasias Ovarianas , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética
11.
Mol Cell Biochem ; 255(1-2): 19-23, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14971642

RESUMO

Arsenite is ubiquitous in the environment, particularly in the form of contaminated water. Although this metal is a known human carcinogen, its exact mechanism of action remains unclear. P70S6K1 phosphorylates the ribosomal 40S protein leading to increased protein translation, and is an important regulator of cell growth and proliferation. Hypoxia inducible factor-1 (HIF-1) is a basic helix-loop-helix transcription factor composed of two subunits, HIF-1alpha and HIF-1beta. HIF-1 activates the transcription of a number of genes that mediate angiogenesis and tumor formation. In this study we demonstrated that arsenite treatment increased levels of p70S6K1 phosphorylation and p70S6K1 activity in a PI3K and mTOR sensitive manner. We have also shown that arsenite specifically induces HIF-1alpha, but not HIF-1beta, protein levels in prostate cancer cells in a mTOR-dependent manner.


Assuntos
Arsenitos/farmacologia , Proteínas de Ligação a DNA , Receptores de Hidrocarboneto Arílico/biossíntese , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Transdução de Sinais/efeitos dos fármacos , Teratogênicos/farmacologia , Fatores de Transcrição/biossíntese , Translocador Nuclear Receptor Aril Hidrocarboneto , Linhagem Celular Tumoral , Ativação Enzimática/efeitos dos fármacos , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia , Masculino , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação/efeitos dos fármacos , Neoplasias da Próstata/metabolismo , Proteínas Quinases/metabolismo , Sirolimo/farmacologia , Serina-Treonina Quinases TOR
12.
Biochem Biophys Res Commun ; 324(2): 471-80, 2004 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-15474452

RESUMO

Ovarian cancer has the highest mortality rate of any gynecological disease affecting women in Western countries. VEGF is a crucial inducer of angiogenesis both in vivo and in vitro. VEGF is commonly upregulated in ovarian cancer and is regulated by HIF-1. SU5416 is known to inhibit various stages of tumor growth. In this study, we show that SU5416 inhibited VEGF mRNA expression in ovarian cancer cells in a dose-dependent manner. SU5416 inhibited VEGF expression at the transcriptional level through the HIF-1 DNA binding site. HIF-1 is composed of HIF-1alpha and HIF-1beta subunits. SU5416 specifically decreased HIF-1alpha, but not HIF-1beta protein levels. To understand the signaling pathways regulating SU5416-inhibited VEGF and HIF-1alpha expression, we found that SU5416 inhibited PI3K activity. AKT is a downstream target of PI3K. We found that SU5416 also inhibited AKT and p70S6K1 activation and activity in a dose-dependent manner. These results demonstrate that SU5416 inhibited VEGF and HIF-1alpha expression through the inhibition of PI3K/AKT/p70S6K1 pathway in ovarian cancer cells. These results indicate that SU5416 may be an effective agent for ovarian cancer treatment through the inhibition of VEGF and HIF-1 expression, and the activation of PI3K/AKT/p70S6K1 signaling pathway.


Assuntos
Indóis/farmacologia , Neovascularização Patológica , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Pirróis/farmacologia , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Fatores de Transcrição/antagonistas & inibidores , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Sítios de Ligação , Northern Blotting , Linhagem Celular Tumoral , Proliferação de Células , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Feminino , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia , Immunoblotting , Luciferases/metabolismo , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/enzimologia , Neoplasias Ovarianas/metabolismo , Fosforilação , Plasmídeos/metabolismo , Ligação Proteica , Proteínas Proto-Oncogênicas c-akt , RNA/metabolismo , RNA Mensageiro/metabolismo , Transdução de Sinais , Sais de Tetrazólio/farmacologia , Tiazóis/farmacologia , Fatores de Tempo , Ativação Transcricional , Transfecção , Regulação para Cima , Fator A de Crescimento do Endotélio Vascular/metabolismo
13.
Am J Physiol Cell Physiol ; 287(2): C281-91, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15028555

RESUMO

Ovarian cancer is one of the most common cancers among women. Recent studies demonstrated that the gene encoding the p110alpha catalytic subunit of phosphatidylinositol 3-kinase (PI3K) is frequently amplified in ovarian cancer cells. PI3K is involved in multiple cellular functions, including proliferation, differentiation, antiapoptosis, tumorigenesis, and angiogenesis. In this study, we demonstrate that the inhibition of PI3K activity by LY-294002 inhibited ovarian cancer cell proliferation and induced G(1) cell cycle arrest. This effect was accompanied by the decreased expression of G(1)-associated proteins, including cyclin D1, cyclin-dependent kinase (CDK) 4, CDC25A, and retinoblastoma phosphorylation at Ser(780), Ser(795), and Ser(807/811). Expression of CDK6 and beta-actin was not affected by LY-294002. Expression of the cyclin kinase inhibitor p16(INK4a) was induced by the PI3K inhibitor, whereas steady-state levels of p21(CIP1/WAF1) were decreased in the same experiment. The inhibition of PI3K activity also inhibited the phosphorylation of AKT and p70S6K1, but not extracellular regulated kinase 1/2. The G(1) cell cycle arrest induced by LY-294002 was restored by the expression of active forms of AKT and p70S6K1 in the cells. Our study shows that PI3K transmits a mitogenic signal through AKT and mammalian target of rapamycin (mTOR) to p70S6K1. The mTOR inhibitor rapamycin had similar inhibitory effects on G(1) cell cycle progression and on the expression of cyclin D1, CDK4, CDC25A, and retinoblastoma phosphorylation. These results indicate that PI3K mediates G(1) progression and cyclin expression through activation of an AKT/mTOR/p70S6K1 signaling pathway in the ovarian cancer cells.


Assuntos
Fase G1/fisiologia , Neoplasias Ovarianas , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Quinases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Transdução de Sinais/fisiologia , Antibióticos Antineoplásicos/farmacologia , Proteínas Sanguíneas/farmacologia , Divisão Celular/fisiologia , Cromonas/farmacologia , Ciclina D1/metabolismo , Quinase 4 Dependente de Ciclina , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Quinases Ciclina-Dependentes/metabolismo , Inibidores Enzimáticos/farmacologia , Feminino , Fase G1/efeitos dos fármacos , Humanos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Morfolinas/farmacologia , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação , Inibidores de Proteínas Quinases , Proteínas Proto-Oncogênicas c-akt , Proteína do Retinoblastoma/metabolismo , Proteínas Quinases S6 Ribossômicas 70-kDa/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Sirolimo/farmacologia , Serina-Treonina Quinases TOR , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA