Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Anticancer Drugs ; 33(1): e752-e755, 2022 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-34387588

RESUMO

No targeted therapies are approved for non-small-cell lung cancer (NSCLC) with Kirsten rat sarcoma viral oncogene homolog (KRAS) mutation to date. Trametinib, a selective allosteric inhibitor of the MEK1/2, demonstrated debatable clinical activity in KRAS-mutant NSCLC. In this case, we present a recurrent advanced NSCLC with KRAS G12C mutation successfully treated with single-agent trametinib therapy. An 87-year-old man who underwent radiotherapy for the right lung adenocarcinoma was admitted to clinical oncology center for recurrent lesions in bilateral lungs. He was unwilling to perform second-line chemotherapy, but underwent molecular profiling and revealed the KRAS G12C mutation. The single-agent target therapy of trametinib showed clinical benefit without obvious toxicity. Furthermore, this report reviewed the previous date of the preclinical and clinical and summarized that KRAS G12C mutation may be more sensitive to the inhibition of mitogen-activated protein kinase kinase. This case advocates for routine screening of KRAS point mutations in the utility of precision medicine and suggests that treatment with trametinib in advanced NSCLC cases with KRAS G12C mutation is well tolerated and effective, especially for those very elderly or unsuitable for more aggressive chemotherapy.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Neoplasias Pulmonares/tratamento farmacológico , Inibidores de Proteínas Quinases/uso terapêutico , Proteínas Proto-Oncogênicas p21(ras)/genética , Piridonas/uso terapêutico , Pirimidinonas/uso terapêutico , Idoso de 80 Anos ou mais , Carcinoma Pulmonar de Células não Pequenas/genética , Humanos , Neoplasias Pulmonares/genética , Masculino , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores
2.
Breast ; 49: 225-232, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31911370

RESUMO

PURPOSE: The proliferation marker Ki67 has prognostic and predictive values in breast cancer, and the cutoff of the Ki67 label index (LI) is a key index for chemotherapy. However, poor interobserver consistency in Ki67 assessment has limited the clinical use of Ki67, especially in luminal cancers. Here, we reported a modified Ki67 assessment method, size-set semiautomatic counting (SSSAC) and investigated its interobserver reproducibility. METHODS: One hundred invasive breast cancer tissues were set immunostained for Ki67 in one laboratory, scanned as digital slides, and sent to 41 pathologists at the laboratories of 16 hospitals for Ki67 LI assessment using size-set semiautomatic counting (SSSAC), size-set visual assessment (SSVA) and size-set digital image analysis (SSDIA) with a specific image viewing software (Aperio Image Scope, Leica, Germany). The intraclass correlation coefficient (ICC) and Bland-Altman plot were used to evaluate interobserver reproducibility. The Wilcoxon signed-rank test was used to analyze the difference in the Ki67 values assessed by SSSAC and SSDIA. RESULTS: SSSAC demonstrated better interobserver reproducibility (ICC = 0.942) than SSVA (ICC = 0.802). The interobserver reproducibility was better in Ki67 homogeneously stained slides and centralized hot-spot slides than in scattered hot-spot slides. The Ki67 value assessed with SSSAC was obviously higher than that assessed with SSDIA (negative ranks (SSDIA < SSSAC): N = 80, sum of ranks = 4274.50; positive ranks (SSDIA > SSSAC): N = 17, sum of ranks = 478.50; Z = -6.837; P < 0.001). CONCLUSION: SSSAC shows satisfactory interobserver reproducibility in the Ki67 assessment of breast cancer and may be a candidate standard method for Ki67 LI assessment in breast cancer and other malignancies.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Carcinoma Ductal de Mama/metabolismo , Carcinoma Ductal de Mama/patologia , Interpretação de Imagem Assistida por Computador/métodos , Antígeno Ki-67/metabolismo , Neoplasias da Mama/diagnóstico , Carcinoma Ductal de Mama/diagnóstico , Feminino , Humanos , Imuno-Histoquímica , Variações Dependentes do Observador , Reprodutibilidade dos Testes
3.
J Pathol ; 247(2): 266-278, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30357833

RESUMO

Phosphatase and tensin homolog deleted on chromosome 10 (PTEN) deficiency in primary human glioblastoma (GBM) is associated with increased invasiveness and poor prognosis with unknown mechanisms. Therefore, how loss of PTEN promotes GBM progression remains to be elucidated. Herein, we identified that ADP-ribosylation factor like-4C (ARL4C) was highly expressed in PTEN-deficient human GBM cells and tissues. Mechanistically, loss of PTEN stabilized ARL4C protein due to AKT/mTOR pathway-mediated inhibition of ARL4C ubiquitination. Functionally, ARL4C enhanced the progression of GBM cells in vitro and in vivo. Moreover, microarray profiling and GST pull-down assay identified that ARL4C accelerated tumor progression via RAC1-mediated filopodium formation. Importantly, targeting PTEN potently inhibited GBM tumor progression in vitro and in vivo, whereas overexpression of ARL4C reversed the tumor progression impaired by PTEN overexpression. Clinically, analyses with patients' specimens validated a negative correlation between PTEN and ARL4C expression. Elevated ARL4C expression but PTEN deficiency in tumor was associated with poorer disease-free survival and overall survival of GBM patients. Taken together, ARL4C is critical for PTEN-deficient GBM progression and acts as a novel prognostic biomarker and a potential therapeutic candidate. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Assuntos
Fatores de Ribosilação do ADP/metabolismo , Neoplasias Encefálicas/enzimologia , Glioblastoma/enzimologia , PTEN Fosfo-Hidrolase/deficiência , Proteínas Proto-Oncogênicas c-akt/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Fatores de Ribosilação do ADP/genética , Animais , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/terapia , Movimento Celular , Proliferação de Células , Intervalo Livre de Doença , Feminino , Regulação Neoplásica da Expressão Gênica , Glioblastoma/genética , Glioblastoma/patologia , Humanos , Camundongos Endogâmicos NOD , Camundongos SCID , Invasividade Neoplásica , PTEN Fosfo-Hidrolase/genética , Estabilidade Proteica , Pseudópodes/enzimologia , Pseudópodes/genética , Pseudópodes/patologia , Transdução de Sinais , Células Tumorais Cultivadas , Ubiquitinação , Proteínas rac1 de Ligação ao GTP/genética , Proteínas rac1 de Ligação ao GTP/metabolismo
4.
Clin Chim Acta ; 489: 254-260, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30194935

RESUMO

RAS oncogene mutations frequently occur in acute myeloid leukaemia (AML), but the prognostic significance of RAS mutations in AML is inconclusive. We searched the databases of PubMed, Web of Science, EMBASE, and Cochrane from 1990 to 2018. In this study, 24 eligible studies were included, and the meta-analysis was conducted with the Comprehensive Meta-Analysis Version 2 software program. The row hazard ratio (HR) was adjusted and re-evaluated when publication bias existed after detecting all the heterogeneities. A combined analysis showed that RAS mutations were not associated with a poor prognosis in general AML patients (HR: 0.96, 95% CI: 0.78-1.19, p = 0.70). To further verify the results, a subgroup analysis was conducted. Interestingly, in the analysis of age bracket, children with RAS mutations had an unfavourable survival (HR: 1.35, 95% CI: 1.05-1.75, p = 0.02) of AML, but the adults did not (HR: 0.87, 95% CI: 0.70-1.09, p = 0.21). Further analysis of the subgroup of children indicated that patients with NRAS mutations had an adverse prognosis (HR: 1.55, 95% CI: 1.13-2.12, p = 0.007), but not those with KRAS mutations (HR: 1.51, 95% CI: 0.34-6.73, p = 0.59). In conclusion, this study revealed that RAS mutations did not influence the over survival for adults with AML. However, NRAS mutations may be a key prognostic marker related with poor survival for children with AML.


Assuntos
Genes ras/genética , Leucemia Mieloide Aguda/genética , Mutação , Humanos
5.
Cancer Res ; 78(11): 3041-3053, 2018 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-29549164

RESUMO

Potassium ion channels are emerging as promalignant factors involved in cancer progression. In this study, we found that invading human gastric cancer cells express high levels of inwardly rectifying potassium channel 2.1 (Kir2.1). Silencing Kir2.1 markedly reduced the invasive and metastatic capabilities as well as the epithelial-mesenchymal transition (EMT) of gastric cancer cells. The promalignant nature of Kir2.1 in gastric cancer cells was independent of potassium permeation but relied on its interaction with serine/threonine-protein kinase 38 (Stk38) to inhibit ubiquitination and degradation of mitogen-activated protein kinase kinase kinase 2 (MEKK2). Degradation of MEKK2 was mediated by small mothers against decapentaplegic-specific E3 ubiquitin protein ligase 1 (Smurf1), which resulted in activation of the MEK1/2-ERK1/2-Snail pathway in gastric cancer cells. In human gastric cancer tissues, expression was high and positively correlated with invasion depth and metastatic status of the tumors as well as poor overall patient survival. Cox regression analysis identified Kir2.1 as an independent prognostic indicator for patients with gastric cancer. Our results suggest that Kir2.1 is an important regulator of gastric cancer malignancy and acts as a novel prognostic marker and a therapeutic target for gastric cancer.Significance: Kir2.1 contributes to invasion and metastasis by a noncanonical ion permeation-independent signaling pathway and may act as a novel prognostic marker and therapeutic target for gastric cancer. Cancer Res; 78(11); 3041-53. ©2018 AACR.


Assuntos
MAP Quinase Quinase 1/genética , MAP Quinase Quinase 2/genética , MAP Quinase Quinase Quinases/genética , Sistema de Sinalização das MAP Quinases/genética , Canais de Potássio Corretores do Fluxo de Internalização/genética , Proteínas Serina-Treonina Quinases/genética , Neoplasias Gástricas/genética , Animais , Linhagem Celular Tumoral , Transição Epitelial-Mesenquimal/genética , Humanos , MAP Quinase Quinase Quinase 2 , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Prognóstico , Transdução de Sinais/genética , Neoplasias Gástricas/patologia , Ubiquitina-Proteína Ligases/genética , Ubiquitinação/genética
6.
J Pathol ; 243(3): 376-389, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28802057

RESUMO

Cancer stem cells/cancer-initiating cells (CICs) and their microenvironmental niche play a vital role in malignant tumour recurrence and metastasis. Cancer-associated fibroblasts (CAFs) are major components of the niche of breast cancer-initiating cells (BCICs), and their interactions may profoundly affect breast cancer progression. Autophagy has been considered to be a critical process for CIC maintenance, but whether it is involved in the cross-talk between CAFs and CICs to affect tumourigenesis and pathological significance has not been determined. In this study, we found that the presence of CAFs containing high levels of microtubule-associated protein 1 light chain 3 (LC3II), a marker of autophagosomes, was associated with more aggressive luminal human breast cancer. CAFs in human luminal breast cancer tissues with high autophagy activity enriched BCICs with increased tumourigenicity. Mechanistically, autophagic CAFs released high-mobility group box 1 (HMGB1), which activated its receptor, Toll-like receptor (TLR) 4, expressed by luminal breast cancer cells, to enhance their stemness and tumourigenicity. Furthermore, immunohistochemistry of 180 luminal breast cancers revealed that high LC3II/TLR4 levels predicted an increased relapse rate and a poorer prognosis. Our findings demonstrate that autophagic CAFs play a critical role in promoting the progression of luminal breast cancer through an HMGB1-TLR4 axis, and that both autophagy in CAFs and TLR4 on breast cancer cells constitute potential therapeutic targets. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Assuntos
Neoplasias da Mama/metabolismo , Fibroblastos Associados a Câncer/metabolismo , Fibroblastos Associados a Câncer/patologia , Transformação Celular Neoplásica/patologia , Proteína HMGB1/metabolismo , Recidiva Local de Neoplasia/metabolismo , Células-Tronco Neoplásicas/metabolismo , Idoso , Idoso de 80 Anos ou mais , Autofagia , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Transformação Celular Neoplásica/metabolismo , Feminino , Humanos , Proteínas Associadas aos Microtúbulos/metabolismo , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/patologia , Células-Tronco Neoplásicas/patologia , Receptor 4 Toll-Like/metabolismo , Microambiente Tumoral/fisiologia
7.
Autophagy ; 13(9): 1528-1542, 2017 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-28812437

RESUMO

Antiangiogenesis with bevacizumab, an antibody against vascular endothelial growth factor (VEGF), has been used for devascularization to limit the growth of malignant glioma. However, the benefits are transient due to elusive mechanisms underlying resistance to the antiangiogenic therapy. Glioma stem cells (GSCs) are capable of forming vasculogenic mimicry (VM), an alternative microvascular circulation independent of VEGF-driven angiogenesis. Herein, we report that the formation of VM was promoted by bevacizumab-induced macroautophagy/autophagy in GSCs, which was associated with tumor resistance to antiangiogenic therapy. We established a 3-dimensional collagen scaffold to examine the formation of VM and autophagy by GSCs, and found that rapamycin increased the number of VM and enhanced KDR/VEGFR-2 phosphorylation. Treatment with chloroquine, or knockdown of the autophagy gene ATG5, inhibited the formation of VM and KDR phosphorylation in GSCs. Notably, neutralization of GSCs-produced VEGF with bevacizumab failed to recapitulate the effect of chloroquine treatment and ATG5 knockdown, suggesting that autophagy-promoted formation of VM was independent of tumor cell-derived VEGF. ROS was elevated when autophagy was induced in GSCs and activated KDR phosphorylation through the phosphoinositide 3-kinase (PI3K)-AKT pathway. A ROS inhibitor, N-acetylcysteine, abolished KDR phosphorylation and the formation of VM by GSCs. By examination of the specimens from 95 patients with glioblastoma, we found that ATG5 and p-KDR expression was strongly associated with the density of VM in tumors and poor clinical outcome. Our results thus demonstrate a crucial role of autophagy in the formation of VM by GSCs, which may serve as a therapeutic target in drug-resistant glioma.


Assuntos
Autofagia , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Glioma/metabolismo , Glioma/patologia , Células-Tronco Neoplásicas/patologia , Neovascularização Patológica/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Animais , Proteína 5 Relacionada à Autofagia/metabolismo , Bevacizumab/farmacologia , Bevacizumab/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/ultraestrutura , Linhagem Celular Tumoral , Proliferação de Células , Cloroquina/farmacologia , Feminino , Técnicas de Silenciamento de Genes , Glioma/tratamento farmacológico , Glioma/ultraestrutura , Humanos , Camundongos , Camundongos SCID , Modelos Biológicos , Neovascularização Patológica/tratamento farmacológico , Fosforilação/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Sirolimo/farmacologia , Análise de Sobrevida , Alicerces Teciduais/química , Fator A de Crescimento do Endotélio Vascular/metabolismo
8.
Am J Cancer Res ; 7(4): 955-961, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28469967

RESUMO

Achaete scute-like 2 (ASCL2) is a member of the basic helix-loop-helix (bHLH) transcription factors, and is expressed mainly in intestinal stem cells under normal conditions. Recently, aberrantly elevated ASCL2 was detected in cancer tissues, but the clinical relevance of ASCL2 in breast cancers remains to be decided. In this study, we evaluated the expression of ASCL2 and its relationship to cancer progression in specimens from 191 cases of breast cancer patients with follow-up information. The results indicated that ASCL2 was highly expressed in cancer cells while it was undetectable in normal epithelial cells. Moreover, the expression of ASCL2 was positively correlated with breast tumor size, lymphatic metastasis and the active growth of tumor cells as shown by increased expression of Ki67. Kaplan-Meier analysis revealed that patients with higher levels of ASCL2 suffered higher tumor recurrent rate. Multivariable Cox-regression analysis showed that elevated expression of ASCL2 was an independent and unfavorable indicator of tumor relapse in breast cancer patients. Altogether, our study suggests that ASCL2 defines a subgroup of highly progressive breast cancer and serves as a marker to evaluate the risk of cancer relapse.

9.
Tumour Biol ; 37(10): 13479-13487, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27465552

RESUMO

Metastasis is a complicated, multistep process and remains the major cause of cancer-related mortality. Exploring the molecular mechanisms underlying tumor metastasis is crucial for development of new strategies for cancer prevention and treatment. In this study, we found that protein tyrosine phosphatase 1B (PTP1B) promoted breast cancer metastasis by regulating phosphatase and tensin homolog (PTEN) but not epithelial-mesenchymal transition (EMT). By detecting PTP1B expression of the specimens from 128 breast cancer cases, we found that the level of PTP1B was higher in breast cancer tissues than the corresponding adjacent normal tissues. Notably, PTP1B was positively associated with lymph node metastasis (LNM) and estrogen receptor (ER) status. In vitro, disturbing PTP1B expression obviously attenuated cell migration and invasion. On the contrary, PTP1B overexpression significantly increased migration and invasion of breast cancer cells. Mechanistically, PTP1B knockdown upregulated PTEN, accompanied with an abatement of AKT phosphorylation and the expression of matrix metalloproteinase 2 (MMP2) and MMP7. Conversely, forced expression of PTP1B reduced PTEN and increased AKT phosphorylation as well as the expression of MMP2 and MMP7. Notably, neither EMT nor stemness of breast cancer cells was regulated by PTP1B. We also found that PTP1B acted as an independent prognostic factor and predicted poor prognosis in ER-positive breast cancer patients. Taken together, our findings provide advantageous evidence for the development of PTP1B as a potential therapeutic target for breast cancer, especially for ER-positive breast cancer patients.


Assuntos
Biomarcadores Tumorais/metabolismo , Neoplasias da Mama/patologia , Movimento Celular , Transição Epitelial-Mesenquimal , Regulação Neoplásica da Expressão Gênica , PTEN Fosfo-Hidrolase/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 1/metabolismo , Apoptose , Western Blotting , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Proliferação de Células , Feminino , Humanos , Técnicas Imunoenzimáticas , Metástase Linfática , Pessoa de Meia-Idade , Gradação de Tumores , Estadiamento de Neoplasias , Fosforilação , Prognóstico , Transdução de Sinais , Células Tumorais Cultivadas
10.
Cancer Lett ; 377(1): 105-15, 2016 07 10.
Artigo em Inglês | MEDLINE | ID: mdl-27091400

RESUMO

Fluorescence-activated cell sorting (FACS) based on the surface marker CD133 is the most common method for isolating glioma stem cells (GSCs) from heterogeneous glioma cell populations. Optimization of this method will have profound implications for the future of GSC research. Five commonly used digestion reagents, Liberase-TL, trypsin, TrypLE, Accutase, and non-enzymatic cell dissociation solution (NECDS), were used to dissociate glioma tumorspheres derived from two primary glioma specimens (091214 and 090116) and the cell lines U87 and T98G. The dissociation time, cell viability, retention of CD133, and stemness capacity were assessed. The results showed that single cells derived from the Liberase-TL (200 µg/ml) group exhibited high viability and less damage to the antigen CD133. However, the efficiency of NECDS for dissociating the tumorspheres into single cells was fairly low. Meanwhile, the use of this digestion reagent resulted in obvious cellular and antigenic impairments. Taken together, Liberase-TL (200 µg/ml) is an ideal reagent for isolating GSCs from tumorspheres. In contrast, the use of NECDS for such a protocol should be carefully considered.


Assuntos
Antígeno AC133/metabolismo , Biomarcadores Tumorais/metabolismo , Separação Celular/métodos , Colagenases/metabolismo , Citometria de Fluxo , Glioma/metabolismo , Células-Tronco Multipotentes/metabolismo , Células-Tronco Neoplásicas/metabolismo , Termolisina/metabolismo , Animais , Linhagem Celular Tumoral , Autorrenovação Celular , Sobrevivência Celular , Feminino , Glioma/patologia , Humanos , Camundongos Nus , Células-Tronco Multipotentes/patologia , Células-Tronco Neoplásicas/patologia , Peptídeo Hidrolases/metabolismo , Fenótipo , Esferoides Celulares , Fatores de Tempo , Tripsina/metabolismo , Carga Tumoral
11.
J Clin Pathol ; 69(4): 313-8, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26483561

RESUMO

AIMS: ASCL2, a basic helix-loop-helix (bHLH) transcription factor, is putatively involved in tumour progression. This study aimed to evaluate ASCL2 expression level in non-small-cell carcinoma (NSCLC) and assess its prognostic value for patients. METHODS: ASCL2 protein expression was detected by immunohistochemistry (IHC cohort) in 79 cases of squamous cell carcinoma (SCC) and 67 cases of adenocarcinoma (AC). Kaplan-Meier analysis and Cox regression analysis were performed to evaluate the prognostic significance of ASCL2. The same analyses were conducted in a cohort (n=790) from The Cancer Genome Atlas database (TCGA) to validate the expression pattern and prognostic value of ASCL2. RESULTS: ASCL2 expression levels were significantly increased in SCC compared with normal lung tissue (p<0.001) and AC (p=0.008). High ASCL2 expression was associated with advanced tumour-node-metastasis (TNM) stage (p=0.023) and worse differentiation status (p=0.001) in SCC, but a positive correlation between ASCL2 expression level and advanced TNM stage (p=0.016) was observed in AC. Kaplan-Meier analysis showed that ASCL2 was prognostic in SCC (p=0.004) but not in AC (p=0.183). Multivariable Cox regression analysis indicated that elevated expression of ASCL2 was an independent prognostic factor (HR 2.764; p=0.030) in SCC patients. The expression pattern and prognostic significance of ASCL2 in SCC and AC were validated using the TCGA cohort. CONCLUSIONS: Elevated expression of ASCL2 may identify an aggressive subgroup in SCC and serve as an independent prognostic indicator in these patients.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/biossíntese , Biomarcadores Tumorais/análise , Carcinoma de Células Escamosas/patologia , Neoplasias Pulmonares/patologia , Adenocarcinoma/mortalidade , Adenocarcinoma/patologia , Adulto , Idoso , Fatores de Transcrição Hélice-Alça-Hélice Básicos/análise , Carcinoma de Células Escamosas/mortalidade , Feminino , Humanos , Imuno-Histoquímica , Estimativa de Kaplan-Meier , Neoplasias Pulmonares/mortalidade , Masculino , Prognóstico , Modelos de Riscos Proporcionais , Análise Serial de Tecidos
12.
Cancer Biol Ther ; 14(10): 937-48, 2013 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-23917399

RESUMO

Endoglin (CD105, END) is upregulated in proliferating endothelial cells, suggesting potential therapeutic properties. However, it is not clear whether endoglin mediates an enhanced proliferative rate or may be upregulated as part of a negative feedback loop. To gain insights into context-dependent and cell type-dependent regulatory effects of endoglin, we studied its role properties in human ovarian carcinoma-derived endothelial cells (ODMECs). We isolated and cultured primary ODMECs from epithelial ovarian carcinoma tissue. ODMECs had higher expression of endoglin and VEGFR-2, and also exhibited enhanced spontaneous formation of vessel-like structures in vitro. Transfection of siRNA targeting endoglin in ODMECs cells resulted in the reduction of the proliferation and tube formation. These results indicate that a subset of ODMECs display abnormal angiogenic properties and this phenotype was blocked by decreasing endoglin levels, suggesting endoglin is essential for stimulating angiogenesis, and targeting it may be an attractive approach to anti-angiogenesis therapy for ovarian carcinoma.


Assuntos
Antígenos CD/metabolismo , Células Endoteliais/metabolismo , Neoplasias Epiteliais e Glandulares/patologia , Neovascularização Patológica/metabolismo , Neoplasias Ovarianas/patologia , Receptores de Superfície Celular/metabolismo , Antígenos CD/genética , Movimento Celular , Proliferação de Células , Forma Celular , Endoglina , Células Endoteliais/fisiologia , Feminino , Técnicas de Silenciamento de Genes , Humanos , Microvasos/patologia , Neoplasias Epiteliais e Glandulares/irrigação sanguínea , Neoplasias Ovarianas/irrigação sanguínea , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Cultura Primária de Células , RNA Interferente Pequeno/genética , Receptores de Superfície Celular/genética , Células Tumorais Cultivadas , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Fator de von Willebrand/metabolismo
14.
Zhonghua Bing Li Xue Za Zhi ; 37(2): 103-8, 2008 Feb.
Artigo em Chinês | MEDLINE | ID: mdl-18681321

RESUMO

OBJECTIVE: To study the expression level and significance of glucose transporter 1 (Glut-1) in normal breast tissue, adenosis, adenoma and breast carcinoma. METHODS: A total of 147 cases of female breast tissue samples, including 92 cases of invasive ductal carcinoma, 26 cases of breast fibroadenoma, 24 cases of breast adenosis and 5 cases of normal breast tissues, were collected for quantitative detection of the expression of Glut-1 protein by immunohistochemistry (EnVision method) and Western blot, and its mRNA by reverse transcriptase-polymerase chain reaction (RT-PCR). RESULTS: In normal breast tissue and benign lesions of the breast, Glut-1 was undetectable or only weakly detectable in cytoplasm of ductal and acinar epithelia. In contrast, the intensity of Glut-1 staining was significantly higher in invasive ductal carcinomas (P = 0.0002) with protein expression predominantly in cellular membrane and lesser in cytoplasm. Western blot and RT-PCR analyses showed that the expression of Glut-1 protein and mRNA were significantly increased in invasive ductal carcinoma than fibroadenoma (P =0.001 for protein; P <0.05 for mRNA) and adenosis (P =0.001 for protein; P < 0.05 for mRNA). There was a significant difference among groups (P = 0.0002 for protein; P = 0.0001 for mRNA). CONCLUSIONS: Glucose transport activity, as indicated by Glut-1 protein and its mRNA expression, significantly increases in breast carcinoma than non-cancerous lesions. The over-expression of Glut-1 in breast carcinoma is tightly coupled with tumor cell proliferation, invasion and metastasis, implying that Glut-1 may serve as a new marker in the early diagnosis and prognostication of breast malignancy as well as a new therapeutic target.


Assuntos
Neoplasias da Mama/metabolismo , Carcinoma Ductal de Mama/metabolismo , Regulação Neoplásica da Expressão Gênica , Transportador de Glucose Tipo 1/metabolismo , Glucose/fisiologia , Feminino , Proteínas Facilitadoras de Transporte de Glucose/genética , Proteínas Facilitadoras de Transporte de Glucose/metabolismo , Transportador de Glucose Tipo 1/genética , Humanos , Prognóstico
15.
Artigo em Inglês | MEDLINE | ID: mdl-12215792

RESUMO

The inhibition of glutamate uptake by superoxide anion radical (O(-)(2).) in rat cortical synaptosomes and the protective effect of Ebselen was studied by radioisotope method. The exposure to xanthine/xanthine oxidase, a O(-)(2).-generating system, resulted in a marked decrease of high-affinity glutamate transport in the synaptosomes. In parallel, the Na(+),K(+)-ATPase activity was also damaged, while the lactate dehydrogenase activity and the TBARS contents in synaptic culture were not affected. It suggests that the inhibition of glutamate uptake is related to the damage of Na(+), K(+)-ATPase by O(-)(2). Ebselen was showed to have a blocking effect on the glutamate uptake inhibition by O(-)(2)., probably through protecting the Na(+),K(+)-ATPase.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...