Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
1.
Pathol Res Pract ; 230: 153764, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-35032831

RESUMO

MicroRNAs (miRNAs) act as oncogenes or tumor suppressors by suppressing the expression of target genes, some of which are engaged in angiogenic signaling pathways directly or indirectly. Tumor development and metastasis are dependent on angiogenesis, and it is the main reason for the poor prognosis of cancer patients. New blood vessels are formed from pre-existing vessels when angiogenesis occurs. Thus, it is essential to develop primary tumors and the spread of cancer to surrounding tissues. MicroRNAs (miRNAs) are small noncoding RNAs involved in various biological processes. They can bind to the 3'-UTR of their target genes and prevent them from expressing. MiRNAs control the activity of endothelial cells (ECs) through altering many biological pathways, which plays a key role in cancer progression and angiogenesis. Recent findings revealed that tumor-derived extracellular vesicles participated directly in the control of tumor angiogenesis by delivering miRNAs to ECs. miRNAs recently show great promise in cancer therapies to inhibit angiogenesis. In this study, we showed the miRNA-regulated signaling pathways in tumor angiogenesis with highlighting the anti-angiogenic therapy response and miRNA delivery methods that have been used to inhibit angiogenesis in both in vivo and in vitro studies.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Proteínas Angiogênicas/antagonistas & inibidores , Neoplasias da Mama/irrigação sanguínea , Neoplasias da Mama/terapia , Terapia Genética , MicroRNAs/uso terapêutico , Neovascularização Patológica , Proteínas Angiogênicas/genética , Proteínas Angiogênicas/metabolismo , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo , Terapia de Alvo Molecular , Transdução de Sinais
2.
Mol Neurobiol ; 57(5): 2461-2478, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32152825

RESUMO

Angiogenesis is the growth of new capillaries from the preexisting blood vessels. Glioblastoma (GBM) tumors are highly vascularized tumors, and glioma growth depends on the formation of new blood vessels. Angiogenesis is a complex process involving proliferation, migration, and differentiation of vascular endothelial cells (ECs) under the stimulation of specific signals. It is controlled by the balance between its promoting and inhibiting factors. Various angiogenic factors and genes have been identified that stimulate glioma angiogenesis. Therefore, attention has been directed to anti-angiogenesis therapy in which glioma proliferation is inhibited by inhibiting the formation of new tumor vessels using angiogenesis inhibitory factors and drugs. Here, in this review, we highlight and summarize the various molecular mediators that regulate GBM angiogenesis with focus on recent clinical research on the potential of exploiting angiogenic pathways as a strategy in the treatment of GBM patients.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Antineoplásicos/uso terapêutico , Neoplasias Encefálicas/irrigação sanguínea , Glioblastoma/irrigação sanguínea , Neovascularização Patológica/fisiopatologia , Adulto , Inibidores da Angiogênese/farmacologia , Proteínas Angiogênicas/antagonistas & inibidores , Proteínas Angiogênicas/fisiologia , Antineoplásicos/farmacologia , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/patologia , Diferenciação Celular , Hipóxia Celular , Ensaios Clínicos como Assunto , Glioblastoma/tratamento farmacológico , Glioblastoma/patologia , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Metaloproteinases da Matriz/fisiologia , Terapia de Alvo Molecular , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/fisiologia , Células-Tronco Neoplásicas/patologia , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/prevenção & controle , Neovascularização Fisiológica/fisiologia , Microambiente Tumoral , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Fator A de Crescimento do Endotélio Vascular/fisiologia
4.
J Transl Med ; 17(1): 71, 2019 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-30832671

RESUMO

BACKGROUND: Angiogenesis inhibition research is a cutting edge area in angiogenesis-dependent disease therapy, especially in cancer therapy. Recently, studies on anti-angiogenic peptides have provided promising results in the field of cancer treatment. METHODS: A non-redundant dataset of 135 anti-angiogenic peptides (positive instances) and 135 non anti-angiogenic peptides (negative instances) was used in this study. Also, 20% of each class were selected to construct an independent test dataset (see Additional files 1, 2). We proposed an effective machine learning based R package (AntAngioCOOL) to predict anti-angiogenic peptides. We have examined more than 200 different classifiers to build an efficient predictor. Also, more than 17,000 features were extracted to encode the peptides. RESULTS: Finally, more than 2000 informative features were selected to train the classifiers for detecting anti-angiogenic peptides. AntAngioCOOL includes three different models that can be selected by the user for different purposes; it is the most sensitive, most specific and most accurate. According to the obtained results AntAngioCOOL can effectively suggest anti-angiogenic peptides; this tool achieved sensitivity of 88%, specificity of 77% and accuracy of 75% on the independent test set. AntAngioCOOL can be accessed at https://cran.r-project.org/ . CONCLUSIONS: Only 2% of the extracted descriptors were used to build the predictor models. The results revealed that physico-chemical profile is the most important feature type in predicting anti-angiogenic peptides. Also, atomic profile and PseAAC are the other important features.


Assuntos
Inibidores da Angiogênese/análise , Inibidores da Angiogênese/farmacologia , Proteínas Angiogênicas/antagonistas & inibidores , Biologia Computacional , Software , Humanos , Aprendizado de Máquina
5.
J Exp Clin Cancer Res ; 38(1): 114, 2019 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-30841931

RESUMO

BACKGROUND: Mast cells are being increasingly recognized as critical components in the tumor microenvironment. Protein Kinase D (PKD) is essential for the progression of prostate cancer, but its role in prostate cancer microenvironment remains poorly understood. METHODS: The expression of PKD, mast cells and microvessel density were examined by IHC. The clinical significance was determined by statistical analyses. The biological function of PKD and the underlying mechanisms were investigated using in vitro and in vivo models. RESULTS: PKD2/3 contributed to MCs recruitment and tumor angiogenesis in the prostate cancer microenvironment. Clinical data showed that increased activation of PKD at Ser744/748 in prostate cancer was correlated with mast cell infiltration and microvascular density. PKD2/3 silencing of prostate cancer cells markedly decreased MCs migration and tube formation of HUVEC cells. Moreover, PKD2/3 depletion not only reduced SCF, CCL5 and CCL11 expression in prostate cancer cells but also inhibited angiogenic factors in MCs. Conversely, exogenous SCF, CCL5 and CCL11 reversed the effect on MCs migration inhibited by PKD2/3 silencing. Mechanistically, PKD2/3 interacted with Erk1/2 and activated Erk1/2 or NF-κB signaling pathway, leading to AP-1 or NF-κB binding to the promoter of scf, ccl5 and ccl11. Finally, PKD-specific inhibitor significantly reduced tumor volume and tumor growth in mice bearing RM-1 prostate cancer cells, which was attributed to attenuation of mast cell recruitment and tumor angiogenesis. CONCLUSIONS: These results demonstrate a novel PKDs function that contributes to tumor angiogenesis and progression through mast cells recruitment in prostate cancer microenvironment.


Assuntos
Proteínas Angiogênicas/genética , Neovascularização Patológica/genética , Neoplasias da Próstata/genética , Proteína Quinase C/genética , Proteínas Angiogênicas/antagonistas & inibidores , Animais , Linhagem Celular Tumoral , Quimiocina CCL11/genética , Quimiocina CCL5/genética , Regulação Neoplásica da Expressão Gênica , Células Endoteliais da Veia Umbilical Humana , Humanos , Sistema de Sinalização das MAP Quinases/genética , Masculino , Mastócitos/metabolismo , Mastócitos/patologia , Camundongos , Neovascularização Patológica/patologia , Fosforilação , Regiões Promotoras Genéticas , Próstata/metabolismo , Próstata/patologia , Neoplasias da Próstata/patologia , Ligação Proteica/genética , Proteína Quinase C/antagonistas & inibidores , Fator de Células-Tronco/genética , Fator de Transcrição AP-1/genética , Fator de Transcrição RelA/genética , Microambiente Tumoral/genética , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Drug Resist Updat ; 36: 47-76, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29499837

RESUMO

Angiogenesis research in the past two decades has contributed significantly towards understanding the molecular pathophysiology of cancer progression and inspired target-oriented research and pharma industry for the development of novel anti-angiogenic agents. Currently, over eleven drugs targeting angiogenesis have been approved by the FDA for the treatment of various malignancies. Of the registered anti-angiogenic clinical trials until the end of 2017 (ClinicalTrials.gov), over 47% were completed, 10% were terminated, 3% withdrawn, over 0.5% were suspended and only 4 trials have culminated in FDA approval for marketing. On the one hand, the clinical benefits of anti-angiogenic drugs prompted the development of novel anti-angiogenic agents. On the other hand, however, a plethora of recent studies demonstrated the emergence of tumor drug resistance towards currently used anti-angiogenic therapeutics. Series of preclinical and clinical studies have highlighted the enigma of drug resistance with functional bypass pathways, and identified compensatory or alternative angiogenic mechanisms assuring tumor growth in the midst of an anti-angiogenic stress environment. In the present review the classical literature of such redundant angiogenic pathways in concert with the key angiogenic factors and specialized cells involved in anti-angiogenic escape mechanisms is described. A strategic discourse regarding increasing tumor drug resistance and future modalities for anti-angiogenic therapy is also discussed in view of recent advances.


Assuntos
Inibidores da Angiogênese/farmacologia , Proteínas Angiogênicas/antagonistas & inibidores , Neoplasias/tratamento farmacológico , Neovascularização Patológica/tratamento farmacológico , Transdução de Sinais/efeitos dos fármacos , Inibidores da Angiogênese/uso terapêutico , Proteínas Angiogênicas/metabolismo , Animais , Ensaios Clínicos como Assunto , Progressão da Doença , Resistencia a Medicamentos Antineoplásicos , Humanos , Neoplasias/irrigação sanguínea , Neoplasias/patologia , Neovascularização Patológica/patologia , Resultado do Tratamento
8.
Proc Natl Acad Sci U S A ; 114(23): E4641-E4647, 2017 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-28533390

RESUMO

Epithelial growth factor-like 7 (EGFL7) is a protein that is secreted by endothelial cells and plays an important role in angiogenesis. Although EGFL7 is aberrantly overexpressed in solid tumors, its role in leukemia has not been evaluated. Here, we report that levels of both EGFL7 mRNA and EGFL7 protein are increased in blasts of patients with acute myeloid leukemia (AML) compared with normal bone marrow cells. High EGFL7 mRNA expression associates with lower complete remission rates, and shorter event-free and overall survival in older (age ≥60 y) and younger (age <60 y) patients with cytogenetically normal AML. We further show that AML blasts secrete EGFL7 protein and that higher levels of EGFL7 protein are found in the sera from AML patients than in sera from healthy controls. Treatment of patient AML blasts with recombinant EGFL7 in vitro leads to increases in leukemic blast cell growth and levels of phosphorylated AKT. EGFL7 blockade with an anti-EGFL7 antibody reduced the growth potential and viability of AML cells. Our findings demonstrate that increased EGFL7 expression and secretion is an autocrine mechanism supporting growth of leukemic blasts in patients with AML.


Assuntos
Fatores de Crescimento Endotelial/genética , Fatores de Crescimento Endotelial/metabolismo , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Adulto , Fatores Etários , Idoso , Idoso de 80 Anos ou mais , Proteínas Angiogênicas/antagonistas & inibidores , Proteínas Angiogênicas/genética , Proteínas Angiogênicas/metabolismo , Animais , Anticorpos Bloqueadores/farmacologia , Proteínas de Ligação ao Cálcio , Estudos de Casos e Controles , Linhagem Celular Tumoral , Proliferação de Células , Intervalo Livre de Doença , Família de Proteínas EGF , Fatores de Crescimento Endotelial/antagonistas & inibidores , Feminino , Humanos , Leucemia Mieloide Aguda/terapia , Masculino , Camundongos , MicroRNAs/genética , MicroRNAs/metabolismo , Pessoa de Meia-Idade , Prognóstico , Proteínas/metabolismo , Proteínas/farmacologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Fatores de Risco , Regulação para Cima , Adulto Jovem
9.
Vascul Pharmacol ; 92: 6-15, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-26196302

RESUMO

Diabetic retinopathy is an ocular complication associated with the chronic endocrine disorder of diabetes mellitus. Angiogenesis is adjudged as a prime modulatory event in this complication. The formation of new blood vessels on the pre-existing vasculature gives rise to an abundance of anatomical and physiological alterations which ultimately results in vision loss. The drastic consequences of this complication prompt the obligation of developing effective therapies for its cure. The existing therapy mainly includes destructive techniques such as laser photocoagulation. Owing to the various drawbacks associated with this technique, there is a need to develop alternative therapies which could halt the progression of diabetic retinopathy without causing considerable damage to the retinal cells. One such possible alternative treatment being researched upon is the antiangiogenic therapy. Since angiogenesis is a critical event during the progression of this disorder, targeting this event may perhaps prove effective in its treatment. Amongst several antiangiogenic agents, thalidomide holds a reputable position due to its effectiveness in terminating angiogenesis during various pathological conditions. This review focuses on the diverse molecular mechanisms proposed to explain the antiangiogenic properties of thalidomide and their applicability in diabetic retinopathy.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Retinopatia Diabética/tratamento farmacológico , Neovascularização Patológica , Neovascularização Retiniana , Vasos Retinianos/efeitos dos fármacos , Talidomida/uso terapêutico , Inibidores da Angiogênese/efeitos adversos , Proteínas Angiogênicas/antagonistas & inibidores , Proteínas Angiogênicas/metabolismo , Animais , Retinopatia Diabética/metabolismo , Retinopatia Diabética/fisiopatologia , Humanos , Vasos Retinianos/metabolismo , Vasos Retinianos/fisiopatologia , Transdução de Sinais/efeitos dos fármacos , Talidomida/efeitos adversos
10.
Cancer Sci ; 108(3): 512-519, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28032401

RESUMO

There are two members of the vasohibin (VASH) family, VASH1 and VASH2. VASH1 is expressed mainly in endothelial cells to inhibit angiogenesis, whereas VASH2 is expressed mainly in cancer cells to stimulate tumor growth. The aim of the present study was to establish neutralizing monoclonal antibody (mAb) against human VASH2 and apply it as an anti-cancer treatment. We previously raised mAb against several synthetic peptides of hVASH1, and found that one of them exhibited neutralizing activity against hVASH1. Because of the similarity in the amino acid sequences between VASH1 and VASH2, we hypothesized that they shared the bioactive center. When we mutated four amino acids within the region, the mutant VASH2 lost its pro-angiogenic activity. Therefore, we raised mAb against a synthetic peptide overlapping the mutated amino acids of hVASH2, and isolated one clone (1760) that almost completely inhibited the stimulatory effect of hVASH2 on the migration of and tube formation by endothelial cells. When we used this clone 1760 antibody for cancer treatment, the peritoneal injection of it inhibited both tumor growth and angiogenesis in a mouse xenograft model of human cancer cells. In terms of anti-tumor activity, 25 mg/kg of clone 1760 was equivalent to 5 mg/kg of bevacizmab. From these results, we propose the targeting of human VASH2 with neutralizing mAb as a new strategy for cancer treatment.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Proteínas Angiogênicas/antagonistas & inibidores , Anticorpos Monoclonais/uso terapêutico , Anticorpos Neutralizantes/uso terapêutico , Neovascularização Patológica/prevenção & controle , Neoplasias Ovarianas/tratamento farmacológico , Proteínas Angiogênicas/imunologia , Animais , Anticorpos Monoclonais/genética , Anticorpos Monoclonais/imunologia , Anticorpos Neutralizantes/genética , Anticorpos Neutralizantes/imunologia , Bevacizumab/uso terapêutico , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Células Endoteliais/metabolismo , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neovascularização Patológica/imunologia , Neoplasias Ovarianas/irrigação sanguínea , Neoplasias Ovarianas/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Life Sci ; 148: 1-8, 2016 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-26860892

RESUMO

AIMS: An abnormal ratio of vasohibin-2/vasohibin-1 may be involved in the abnormal angiogenesis and vascular remodeling during pulmonary arterial hypertension (PAH). MAIN METHODS: To evaluate the pharmacological actions of Ponatinib (AP) in experimental model of PAH, the effects of AP on TGF-ß1-mediated endothelial-mesenchymal transition (EndoMT) in human pulmonary microvascular endothelial cells (HPMEC), and the hypoxic human pulmonary artery smooth muscle cells (HPASMC) proliferation and HPMEC in vitro, and on bleomycin (BLM)-induced PAH in vivo were investigated. KEY FINDINGS: AP treatment resulted in a reduction of EndoMT in HPMECs with a decrease of vimentin, whereas an increase of VE-cadherin, reduction of fibroblast growth factor (FGF-2), vascular endothelial growth factor (VEGF) and vasohibin-2 (VASH-2), whereas an increase of vasohibin-1 (VASH-1) in the hypoxic HPMEC, a reduction of the HPASMC proliferation with decreases of wnt5a, ß-catenin and cyclin D1 expression. AP ameliorated BLM-induced PAH in rats with reductions of FGF-2, VEGF, von Willebrand factor (vWF) and VASH-2 expression, whereas an increase of VASH-1 expression. AP ameliorated BLM-induced PAH in rats with reductions of the pathological score and the collagen deposition. In addition, AP ameliorated hemodynamics and right ventricular hypertrophy. SIGNIFICANCES: Our results identified a therapeutic potential of AP in PAH therapy might be modulated VASH-2/VASH-1 and the Wnt signaling.


Assuntos
Proteínas Angiogênicas/metabolismo , Proteínas de Ciclo Celular/metabolismo , Hipertensão Pulmonar/metabolismo , Hipertensão Pulmonar/prevenção & controle , Imidazóis/uso terapêutico , Piridazinas/uso terapêutico , Via de Sinalização Wnt/fisiologia , Proteínas Angiogênicas/antagonistas & inibidores , Animais , Proteínas de Ciclo Celular/antagonistas & inibidores , Células Cultivadas , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Humanos , Imidazóis/farmacologia , Masculino , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Piridazinas/farmacologia , Ratos , Ratos Sprague-Dawley , Via de Sinalização Wnt/efeitos dos fármacos
12.
Cancer Lett ; 381(1): 201-10, 2016 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-26723874

RESUMO

The importance of angiogenesis in pancreatic ductal adenocarcinoma (PDAC) and its therapeutic potential have been explored in both pre-clinical and clinical studies. Human PDACs overexpress a number of angiogenic factors and their cognate high-affinity receptors, and anti-angiogenic agents reduce tumor volume, metastasis, and microvessel density (MVD), and improve survival in subcutaneous and orthotopic pre-clinical models. Nonetheless, clinical trials using anti-angiogenic therapy have been overwhelmingly unsuccessful. This review will focus on these pre-clinical and clinical studies, the potential reasons for failure in the clinical setting, and ways these shortcomings could be addressed in future investigations of angiogenic mechanisms in PDAC.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Proteínas Angiogênicas/antagonistas & inibidores , Carcinoma Ductal Pancreático/tratamento farmacológico , Neovascularização Patológica , Neoplasias Pancreáticas/tratamento farmacológico , Inibidores da Angiogênese/efeitos adversos , Proteínas Angiogênicas/metabolismo , Animais , Carcinoma Ductal Pancreático/irrigação sanguínea , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patologia , Resistencia a Medicamentos Antineoplásicos , Humanos , Terapia de Alvo Molecular , Neoplasias Pancreáticas/irrigação sanguínea , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Receptores de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Receptores de Fatores de Crescimento do Endotélio Vascular/metabolismo , Transdução de Sinais/efeitos dos fármacos , Microambiente Tumoral , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Fator A de Crescimento do Endotélio Vascular/metabolismo
13.
Cancer Lett ; 380(2): 586-597, 2016 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-26639195

RESUMO

Bone morphogenetic protein (BMP) belongs to transforming growth factor-ß superfamily. To date, more than 20 BMPs have been identified in humans. BMPs play a critical role in embryonic and postnatal development, and also in maintaining homeostasis in different organs and tissues by regulating cell differentiation, proliferation, survival and motility. They play important roles in the development and progression of certain malignancies, including prostate cancer, breast cancer, lung cancer, etc. Recently, more evidence shows that BMPs are also involved in tumour associated angiogenesis. For example BMP can either directly regulate the functions of vascular endothelial cells or indirectly influence the angiogenesis via regulation of angiogenic factors, such as vascular endothelial growth factor (VEGF). Such crosstalk can also be reflected in the interaction with other angiogenic factors, like hepatocyte growth factor (HGF) and basic fibroblast growth factor (bFGF). All these factors are involved in the orchestration of the angiogenic process during tumour development and progression. Review of the relevant studies will provide a comprehensive prospective on current understanding and shed light on the corresponding therapeutic opportunity.


Assuntos
Proteínas Angiogênicas/metabolismo , Proteínas Morfogenéticas Ósseas/metabolismo , Neoplasias/irrigação sanguínea , Neoplasias/metabolismo , Neovascularização Patológica , Inibidores da Angiogênese/uso terapêutico , Proteínas Angiogênicas/antagonistas & inibidores , Animais , Proteínas Morfogenéticas Ósseas/antagonistas & inibidores , Humanos , Metástase Neoplásica , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Transdução de Sinais , Microambiente Tumoral
14.
Cancer Lett ; 380(1): 216-26, 2016 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-26238184

RESUMO

Angiogenesis is sustained by classical and non-classical proangiogenic factors (PFs) acting in tumor microenvironment and these factors are also potential targets of antiangiogenic therapies. All PFs induce the overexpression of several signaling pathways that lead to migration and proliferation of endothelial cells contributing to tumor angiogenesis and survival of cancer cells. In this review, we have analyzed each PF with its specific receptor/s and we have summarized the available antiangiogenic drugs (e.g. monoclonal antibodies) targeting these PFs, some of these agents have already been approved, others are currently in development for the treatment of several human malignancies.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Proteínas Angiogênicas/antagonistas & inibidores , Células Endoteliais/efeitos dos fármacos , Neoplasias/tratamento farmacológico , Neovascularização Patológica , Microambiente Tumoral , Proteínas Angiogênicas/metabolismo , Animais , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Humanos , Terapia de Alvo Molecular , Neoplasias/irrigação sanguínea , Neoplasias/metabolismo , Neoplasias/patologia , Transdução de Sinais/efeitos dos fármacos
15.
Korean J Radiol ; 16(4): 776-82, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26175576

RESUMO

OBJECTIVE: To evaluate whether suppression of tumor microvasculature by double anti-angiogenic protein (DAAP) treatment could increase the extent of radiofrequency ablation (RFA)-induced coagulation in a murine renal cell carcinoma model. MATERIALS AND METHODS: Renal cell carcinoma cell lines were implanted subcutaneously into 10 nude mice. Four mice received adenoviral DAAP treatment and 6 mice received sterile 0.9% saline solution as DAAP-untreated group. The effect of DAAP was evaluated according to the vascularity by contrast-enhanced ultrasound (CEUS) using microbubbles. Four DAAP-treated mice and 4 DAAP-untreated mice were then treated with RFA, resulting in 3 groups: no-therapy (n = 2), RFA only (n = 4), and RFA combined with DAAP treatment (n = 4). Immediately after RFA, the size of coagulation necrosis and mitochondrial enzyme activity were compared between the groups using analysis of variance (ANOVA) and post hoc test. RESULTS: The contrast enhancement ratio for tumor vascularization on CEUS was significantly lower in the DAAP treated group than in DAAP-untreated group (30.2 ± 9.9% vs. 77.4 ± 17.3%; p = 0.021). After RFA, the mean coagulation diameter was 0 mm for no-therapy group, 6.7 ± 0.7 mm for the RFA only group and 8.5 ± 0.4 mm for the RFA with DAAP group (ANOVA, p < 0.001). The area of viable mitochondria within the tumor was 27.9 ± 3.9% in no-therapy group, 10.3 ± 4.5% in the RFA only group, and 2.1 ± 0.7% in the RFA with DAAP group (ANOVA, p < 0.001). CONCLUSION: Our results suggest the potential value of combining RFA with anti-angiogenic therapy.


Assuntos
Proteínas Angiogênicas/antagonistas & inibidores , Carcinoma de Células Renais/terapia , Ablação por Cateter/métodos , Neoplasias Renais/terapia , Neovascularização Patológica/terapia , Adenoviridae , Animais , Carcinoma de Células Renais/irrigação sanguínea , Carcinoma de Células Renais/cirurgia , Terapia Combinada , Meios de Contraste , Neoplasias Renais/irrigação sanguínea , Neoplasias Renais/cirurgia , Masculino , Camundongos , Camundongos Nus , Microbolhas , Neovascularização Patológica/diagnóstico por imagem , Neovascularização Patológica/cirurgia , Proteínas Recombinantes , Ultrassonografia
16.
J Pathol ; 237(2): 190-202, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25988668

RESUMO

Recent studies reveal that chemotherapy can enhance metastasis due to host responses, such as augmented expression of adhesion molecules in endothelial cells and increased populations of myeloid cells. However, it is still unclear how tumour cells contribute to this process. Here, we observed that paclitaxel and carboplatin accelerated lung metastasis in tumour-bearing mice, while doxorubicin and fluorouracil did not. Mechanistically, paclitaxel and carboplatin induced similar changes in cytokine and angiogenic factors. Increased levels of CXCR2, CXCR4, S1P/S1PR1, PlGF and PDGF-BB were identified in the serum or primary tumour tissues of tumour-bearing mice treated by paclitaxel. The serum levels of CXCL1 and PDGF-BB and the tissue level of CXCR4 were also elevated by carboplatin. On the other hand, doxorubicin and fluorouracil did not induce such changes. The chemotherapy-induced cytokine and angiogenic factor changes were also confirmed in gene expression datasets from human patients following chemotherapy treatment. These chemotherapy-enhanced cytokines and angiogenic factors further induced angiogenesis, destabilized vascular integrity, recruited BMDCs to metastatic organs and mediated the proliferation, migration and epithelial-to-mesenchymal transition of tumour cells. Interestingly, inhibitors of these factors counteracted chemotherapy-enhanced metastasis in both tumour-bearing mice and normal mice injected intravenously with B16F10-GFP cells. In particular, blockade of the SDF-1α-CXCR4 or S1P-S1PR1 axes not only compromised chemotherapy-induced metastasis but also prolonged the median survival time by 33.9% and 40.3%, respectively. The current study delineates the mechanism of chemotherapy-induced metastasis and provides novel therapeutic strategies to counterbalance pro-metastatic effects of chemo-drugs via combination treatment with anti-cytokine/anti-angiogenic therapy.


Assuntos
Proteínas Angiogênicas/metabolismo , Antineoplásicos/toxicidade , Carboplatina/toxicidade , Citocinas/metabolismo , Neoplasias Pulmonares/secundário , Melanoma Experimental/secundário , Paclitaxel/toxicidade , Transdução de Sinais/efeitos dos fármacos , Células Estromais/efeitos dos fármacos , Inibidores da Angiogênese/farmacologia , Proteínas Angiogênicas/antagonistas & inibidores , Proteínas Angiogênicas/sangue , Animais , Becaplermina , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Citocinas/antagonistas & inibidores , Citocinas/sangue , Relação Dose-Resposta a Droga , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Feminino , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/prevenção & controle , Lisofosfolipídeos/metabolismo , Melanoma Experimental/metabolismo , Melanoma Experimental/prevenção & controle , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Nus , Fator de Crescimento Placentário , Proteínas da Gravidez/metabolismo , Proteínas Proto-Oncogênicas c-sis/metabolismo , Receptores CXCR4/metabolismo , Receptores de Interleucina-8B/metabolismo , Receptores de Lisoesfingolipídeo/metabolismo , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Receptores de Esfingosina-1-Fosfato , Células Estromais/metabolismo , Células Estromais/patologia , Fatores de Tempo , Regulação para Cima , Ensaios Antitumorais Modelo de Xenoenxerto
17.
Arch Pharm (Weinheim) ; 348(5): 338-46, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25846009

RESUMO

Pyridazinones are widely recognized as versatile scaffolds with a wide spectrum of biological activities. In the present work, a series of new 4-chloro-2-(3-chloro-4-fluorophenyl)-5-(aliphatic/cyclic saturated amino)pyridazin-3(2H)-one derivatives 4a-i were synthesized and characterized by spectral techniques. The inhibitory effects of the synthesized compounds 4a-i on the viability of three human cancer cell lines, HEP3BPN 11 (liver), MDA 453 (breast), and HL 60 (leukemia), were assessed using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) reduction assay. Among the compounds 4a-i screened, 4g and 4i exhibited inhibitory activity very close to the standard methotrexate; therefore, these lead compounds were further tested for their potential to inhibit the proangiogenic cytokines involved in tumor progression. Compound 4g was found to be a potent antiangiogenic agent against TNFα, VEGF, FGFb, and TGFß, whereas 4i showed potent antiangiogenic activity against TNFα, VEGF, FGFb, and leptin. All the compounds 4a-i were screened for their antioxidant activities using 2,2-diphenyl-1-picryl hydrazine (DPPH), OH, and superoxide anion radicals. Compound 4f showed better OH radical scavenging activity than the standard ascorbic acid.


Assuntos
Inibidores da Angiogênese/síntese química , Inibidores da Angiogênese/farmacologia , Antioxidantes/síntese química , Antioxidantes/farmacologia , Neoplasias/tratamento farmacológico , Piridonas/síntese química , Piridonas/farmacologia , Proteínas Angiogênicas/antagonistas & inibidores , Proteínas Angiogênicas/metabolismo , Compostos de Bifenilo/química , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Desenho de Fármacos , Feminino , Células HL-60 , Humanos , Estrutura Molecular , Neoplasias/patologia , Picratos/química , Relação Estrutura-Atividade , Superóxidos/química
18.
Artigo em Inglês | WPRIM (Pacífico Ocidental) | ID: wpr-22493

RESUMO

OBJECTIVE: To evaluate whether suppression of tumor microvasculature by double anti-angiogenic protein (DAAP) treatment could increase the extent of radiofrequency ablation (RFA)-induced coagulation in a murine renal cell carcinoma model. MATERIALS AND METHODS: Renal cell carcinoma cell lines were implanted subcutaneously into 10 nude mice. Four mice received adenoviral DAAP treatment and 6 mice received sterile 0.9% saline solution as DAAP-untreated group. The effect of DAAP was evaluated according to the vascularity by contrast-enhanced ultrasound (CEUS) using microbubbles. Four DAAP-treated mice and 4 DAAP-untreated mice were then treated with RFA, resulting in 3 groups: no-therapy (n = 2), RFA only (n = 4), and RFA combined with DAAP treatment (n = 4). Immediately after RFA, the size of coagulation necrosis and mitochondrial enzyme activity were compared between the groups using analysis of variance (ANOVA) and post hoc test. RESULTS: The contrast enhancement ratio for tumor vascularization on CEUS was significantly lower in the DAAP treated group than in DAAP-untreated group (30.2 +/- 9.9% vs. 77.4 +/- 17.3%; p = 0.021). After RFA, the mean coagulation diameter was 0 mm for no-therapy group, 6.7 +/- 0.7 mm for the RFA only group and 8.5 +/- 0.4 mm for the RFA with DAAP group (ANOVA, p < 0.001). The area of viable mitochondria within the tumor was 27.9 +/- 3.9% in no-therapy group, 10.3 +/- 4.5% in the RFA only group, and 2.1 +/- 0.7% in the RFA with DAAP group (ANOVA, p < 0.001). CONCLUSION: Our results suggest the potential value of combining RFA with anti-angiogenic therapy.


Assuntos
Animais , Masculino , Camundongos , Adenoviridae , Proteínas Angiogênicas/antagonistas & inibidores , Carcinoma de Células Renais/irrigação sanguínea , Ablação por Cateter/métodos , Terapia Combinada , Meios de Contraste , Neoplasias Renais/irrigação sanguínea , Camundongos Nus , Microbolhas , Neovascularização Patológica/cirurgia , Proteínas Recombinantes
19.
World J Gastroenterol ; 20(27): 8910-20, 2014 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-25083063

RESUMO

Mast cells (MCs), located ubiquitously near blood vessels, are descended from CD34(+) hematopoietic stem cells. Initially, although their role has been well defined in hypersensitivity reactions, the discovery of their sharing in both innate and adaptive immunity has allowed to redefine their crucial interplay on the regulatory function between inflammatory and tumor cells through the release of mediators granule-associated (mainly tryptase and vascular endothelial growth factor). In particular, in several animal and human malignancies it has been well demonstrated that activated c-Kit receptor (c-KitR) and tryptase (an agonist of the proteinase-activated receptor-2) take pivotal part in tumor angiogenesis after the MCs activation, contributing to tumor cells invasion and metastasis. In this review, we focused on crucial MCs density (MCD) role in colorectal cancer (CRC) development and progression angiogenesis-mediated; then, we will analyze the principal studies that have focused on MCD as possible prognostic factor. Finally, we will consider a possible role of MCD as novel therapeutic target mainly by c-KitR tyrosine kinase inhibitors (imatinib, masitinib) and tryptase inhibitors (gabexate and nafamostat mesylate) with the aim to prevent CRC progression.


Assuntos
Proteínas Angiogênicas/metabolismo , Neoplasias Colorretais/irrigação sanguínea , Neoplasias Colorretais/metabolismo , Mastócitos/metabolismo , Neovascularização Patológica , Inibidores da Angiogênese/uso terapêutico , Proteínas Angiogênicas/antagonistas & inibidores , Animais , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/imunologia , Neoplasias Colorretais/patologia , Desenho de Fármacos , Humanos , Mastócitos/efeitos dos fármacos , Mastócitos/imunologia , Mastócitos/patologia , Terapia de Alvo Molecular , Prognóstico , Transdução de Sinais
20.
Mol Med Rep ; 10(2): 663-9, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24920244

RESUMO

Vasohibin­2 (VASH2) is an angiogenic factor, and has been previously reported to be a cancer­related gene, with cytoplasmic and karyotypic forms. In the current study VASH2 expression in human breast cancer tissue and adjacent non­cancerous tissue was investigated with immunohistochemistry. MCF­7 and BT474 human breast cancer cells were transfected with lentiviral constructs to generate in vitro VASH2 overexpression and knockdown models. In addition, BALB/cA nude mice were inoculated subcutaneously with transfected cells to generate in vivo models of VASH2 overexpression and knockdown. The effect of VASH2 on cell proliferation was investigated using a bromodeoxyuridine assay in vitro and immunohistochemistry of Ki67 in xenograft tumors. Growth factors were investigated using a human growth factor array, and certain factors were further confirmed by an immunoblot. The results indicated that the expression level of cytoplasmic VASH2 was higher in breast cancer tissues with a Ki67 (a proliferation marker) level of ≥14%, compared with tissues with a Ki67 level of <14%. VASH2 induced proliferation in vitro and in vivo. Four growth factors activated by VASH2 were identified as follows: Fibroblast growth factor 2 (FGF2), growth/differentiation factor­15 (GDF15), insulin­like growth factor­binding protein (IGFBP)3 and IGFBP6. FGF2 and GDF15 may contribute to VASH2­induced proliferation. The current study identified a novel role for VASH2 in human breast cancer, and this knowledge suggests that VASH2 may be a novel target in breast cancer treatment.


Assuntos
Proteínas Angiogênicas/metabolismo , Neoplasias da Mama/patologia , Fator 2 de Crescimento de Fibroblastos/metabolismo , Fator 15 de Diferenciação de Crescimento/metabolismo , Adulto , Proteínas Angiogênicas/antagonistas & inibidores , Proteínas Angiogênicas/genética , Animais , Neoplasias da Mama/metabolismo , Proliferação de Células , Feminino , Humanos , Imuno-Histoquímica , Proteína 3 de Ligação a Fator de Crescimento Semelhante à Insulina/metabolismo , Proteína 6 de Ligação a Fator de Crescimento Semelhante à Insulina/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Antígeno Ki-67/metabolismo , Células MCF-7 , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Pessoa de Meia-Idade , Análise Serial de Proteínas , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Transplante Heterólogo , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...