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












Base de dados
Intervalo de ano de publicação
1.
Front Cell Dev Biol ; 11: 1076458, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37035245

RESUMO

All-trans-retinoic acid (ATRA)-based differentiation therapy of acute promyelocytic leukemia (APL) represents one of the most clinically effective examples of precision medicine and the first example of targeted oncoprotein degradation. The success of ATRA in APL, however, remains to be translated to non-APL acute myeloid leukemia (AML). We previously showed that aberrant histone modifications, including histone H3 lysine 4 (H3K4) and lysine 27 (H3K27) methylation, were associated with this lack of response and that epigenetic therapy with small molecule inhibitors of the H3K4 demethylase LSD1/KDM1A could reprogram AML cells to respond to ATRA. Serving as the enzymatic component of Polycomb Repressive Complex 2, EZH2/KMT6A methyltransferase plays a critical role in normal hematopoiesis by affecting the balance between self-renewal and differentiation. The canonical function of EZH2 is methylation of H3K27, although important non-canonical roles have recently been described. EZH2 mutation or deregulated expression has been conclusively demonstrated in the pathogenesis of AML and response to treatment, thus making it an attractive therapeutic target. In this study, we therefore investigated whether inhibition of EZH2 might also improve the response of non-APL AML cells to ATRA-based therapy. We focused on GSK-343, a pyridone-containing S-adenosyl-L-methionine cofactor-competitive EZH2 inhibitor that is representative of its class, and HKMTI-1-005, a substrate-competitive dual inhibitor targeting EZH2 and the closely related G9A/GLP H3K9 methyltransferases. We found that treatment with HKMTI-1-005 phenocopied EZH2 knockdown and was more effective in inducing differentiation than GSK-343, despite the efficacy of GSK-343 in terms of abolishing H3K27 trimethylation. Furthermore, transcriptomic analysis revealed that in contrast to treatment with GSK-343, HKMTI-1-005 upregulated the expression of differentiation pathway genes with and without ATRA, while downregulating genes associated with a hematopoietic stem cell phenotype. These results pointed to a non-canonical role for EZH2, which was supported by the finding that EZH2 associates with the master regulator of myeloid differentiation, RARα, in an ATRA-dependent manner that was enhanced by HKMTI-1-005, possibly playing a role in co-regulator complex exchange during transcriptional activation. In summary, our results strongly suggest that addition of HKMTI-1-005 to ATRA is a new therapeutic approach against AML that warrants further investigation.

2.
J Exp Clin Cancer Res ; 39(1): 78, 2020 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-32375866

RESUMO

The majority of high-risk neuroblastomas can be divided into three distinct molecular subgroups defined by the presence of MYCN amplification, upstream TERT rearrangements or alternative lengthening of telomeres (ALT). The common defining feature of all three subgroups is altered telomere maintenance; MYCN amplification and upstream TERT rearrangements drive high levels of telomerase expression whereas ALT is a telomerase independent telomere maintenance mechanism. As all three telomere maintenance mechanisms are independently associated with poor outcomes, the development of strategies to selectively target either telomerase expressing or ALT cells holds great promise as a therapeutic approach that is applicable to the majority of children with aggressive disease.Here we summarise the biology of telomere maintenance and the molecular drivers of aggressive neuroblastoma before describing the most promising therapeutic strategies to target both telomerase expressing and ALT cancers. For telomerase-expressing neuroblastoma the most promising targeted agent to date is 6-thio-2'-deoxyguanosine, however clinical development of this agent is required. In osteosarcoma cell lines with ALT, selective sensitivity to ATR inhibition has been reported. However, we present data showing that in fact ALT neuroblastoma cells are more resistant to the clinical ATR inhibitor AZD6738 compared to other neuroblastoma subtypes. More recently a number of additional candidate compounds have been shown to show selectivity for ALT cancers, such as Tetra-Pt (bpy), a compound targeting the telomeric G-quadruplex and pifithrin-α, a putative p53 inhibitor. Further pre-clinical evaluation of these compounds in neuroblastoma models is warranted.In summary, telomere maintenance targeting strategies offer a significant opportunity to develop effective new therapies, applicable to a large proportion of children with high-risk neuroblastoma. In parallel to clinical development, more pre-clinical research specifically for neuroblastoma is urgently needed, if we are to improve survival for this common poor outcome tumour of childhood.


Assuntos
Neuroblastoma/terapia , Telômero/fisiologia , Linhagem Celular Tumoral , Pré-Escolar , Feminino , Humanos , Lactente , Masculino , Neuroblastoma/tratamento farmacológico , Neuroblastoma/genética , Telômero/efeitos dos fármacos , Telômero/genética , Telômero/metabolismo
3.
Endocr Relat Cancer ; 26(3): 355-366, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30645190

RESUMO

Pharmacological inhibition of the sonic hedgehog (SHH) pathway can be beneficial against certain cancers but detrimental in others. Adamantinomatous craniopharyngioma (ACP) is a relevant pituitary tumour, affecting children and adults, that is associated with high morbidity and increased mortality in long-term follow-up. We have previously demonstrated overactivation of the SHH pathway in both human and mouse ACP. Here, we show that this activation is ligand dependent and induced by the expression of SHH protein in a small proportion of tumour cells. We investigate the functional relevance of SHH signalling in ACP through MRI-guided preclinical studies using an ACP mouse model. Treatment with vismodegib, a clinically approved SHH pathway inhibitor, results in a significant reduction in median survival due to premature development of highly proliferative and vascularised undifferentiated tumours. Reinforcing the mouse data, SHH pathway inhibition in human ACP leads to a significant increase in tumour cell proliferation both ex vivo, in explant cultures, and in vivo, in a patient-derived xenograft model. Together, our results demonstrate a protumourigenic effect of vismodegib-mediated SHH pathway inhibition in ACP.


Assuntos
Craniofaringioma/fisiopatologia , Proteínas Hedgehog/antagonistas & inibidores , Adolescente , Animais , Proliferação de Células , Criança , Pré-Escolar , Modelos Animais de Doenças , Humanos , Masculino , Camundongos , Neoplasias Hipofisárias , Transdução de Sinais
4.
Dis Model Mech ; 9(9): 941-52, 2016 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-27483357

RESUMO

The first-in-class inhibitor of ALK, c-MET and ROS1, crizotinib (Xalkori), has shown remarkable clinical efficacy in treatment of ALK-positive non-small cell lung cancer. However, in neuroblastoma, activating mutations in the ALK kinase domain are typically refractory to crizotinib treatment, highlighting the need for more potent inhibitors. The next-generation ALK inhibitor PF-06463922 is predicted to exhibit increased affinity for ALK mutants prevalent in neuroblastoma. We examined PF-06463922 activity in ALK-driven neuroblastoma models in vitro and in vivo In vitro kinase assays and cell-based experiments examining ALK mutations of increasing potency show that PF-06463922 is an effective inhibitor of ALK with greater activity towards ALK neuroblastoma mutants. In contrast to crizotinib, single agent administration of PF-06463922 caused dramatic tumor inhibition in both subcutaneous and orthotopic xenografts as well as a mouse model of high-risk neuroblastoma driven by Th-ALK(F1174L)/MYCN Taken together, our results suggest PF-06463922 is a potent inhibitor of crizotinib-resistant ALK mutations, and highlights an important new treatment option for neuroblastoma patients.


Assuntos
Lactamas Macrocíclicas/uso terapêutico , Proteína Proto-Oncogênica N-Myc/antagonistas & inibidores , Neuroblastoma/tratamento farmacológico , Inibidores de Proteínas Quinases/uso terapêutico , Receptores Proteína Tirosina Quinases/antagonistas & inibidores , Aminopiridinas , Quinase do Linfoma Anaplásico , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Ensaios Clínicos como Assunto , Crizotinibe , Lactamas , Lactamas Macrocíclicas/farmacologia , Camundongos Endogâmicos BALB C , Camundongos Nus , Mutação/genética , Proteína Proto-Oncogênica N-Myc/metabolismo , Neuroblastoma/patologia , Células PC12 , Inibidores de Proteínas Quinases/farmacologia , Pirazóis/farmacologia , Pirazóis/uso terapêutico , Piridinas/farmacologia , Piridinas/uso terapêutico , Ratos , Receptores Proteína Tirosina Quinases/genética , Receptores Proteína Tirosina Quinases/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Cancer Res ; 59(9): 2142-9, 1999 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-10232601

RESUMO

Many tumor cells or their secreted products suppress the function of tumor-infiltrating macrophages. Tumor cells often produce abundant transforming growth factor beta1 (TGF-beta1), which in addition to other immunosuppressive actions suppresses the inducible isoform of NO synthase. TGF-beta1 is secreted in a latent form, which consists of TGF-beta1 noncovalently associated with latency-associated peptide (LAP) and which can be activated efficiently by exposure to reactive oxygen species. Coculture of the human lung adenocarcinoma cell line A549 and ANA-1 macrophages activated with IFN-gamma plus lipopolysaccharide resulted in increased synthesis and activation of latent TGF-beta1 protein by both A549 and ANA-1 cells, whereas unstimulated cultures of either cell type alone expressed only latent TGF-beta1. We investigated whether exposure of tumor cells to NO influences the production, activation, or activity of TGF-beta1.A549 human lung adenocarcinoma cells exposed to the chemical NO donor diethylamine-NONOate showed increased immunoreactivity of cell-associated latent and active TGF-beta1 in a time- and dose-dependent fashion at 24-48 h after treatment. Exposure of latent TGF-beta1 to solution sources of NO neither led to recombinant latent TGF-beta1 activation nor modified recombinant TGF-beta1 activity. A novel mechanism was observed, however: treatment of recombinant LAP with NO resulted in its nitrosylation and interfered with its ability to neutralize active TGF-beta1. These results provide the first evidence that nitrosative stress influences the regulation of TGF-beta1 and raise the possibility that NO production may augment TGF-beta1 activity by modifying a naturally occurring neutralizing peptide.


Assuntos
Precursores Enzimáticos/metabolismo , Regulação Neoplásica da Expressão Gênica , Proteínas de Neoplasias/biossíntese , Óxido Nítrico/fisiologia , Fragmentos de Peptídeos , Precursores de Proteínas , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Proteínas/metabolismo , Fator de Crescimento Transformador beta/biossíntese , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Animais , Técnicas de Cocultura , Indução Enzimática , Precursores Enzimáticos/genética , Humanos , Hidrazinas/farmacologia , Processamento de Imagem Assistida por Computador , Interferon gama/farmacologia , Lipopolissacarídeos/farmacologia , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Ativação de Macrófagos/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Macrófagos/fisiologia , Camundongos , Proteínas de Neoplasias/genética , Óxido Nítrico/farmacologia , Doadores de Óxido Nítrico/farmacologia , Óxido Nítrico Sintase/biossíntese , Óxido Nítrico Sintase/fisiologia , Óxido Nítrico Sintase Tipo II , Óxidos de Nitrogênio , Estresse Oxidativo , Proteínas Recombinantes de Fusão/biossíntese , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo , Fator de Crescimento Transformador beta1
7.
J Exp Med ; 184(4): 1425-33, 1996 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-8879214

RESUMO

In Alzheimer's disease (AD), affected neurons accumulate beta amyloid protein, components of which can induce mouse microglia to express the high-output isoform of nitric oxide synthase (NOS2) in vitro. Products of NOS2 can be neurotoxic. In mice, NOS2 is normally suppressed by transforming growth factor beta 1 (TGF-beta 1). Expression of TGF-beta 1 is decreased in brains from AD patients, a situation that might be permissive for accumulation of NOS2. Accordingly, we investigated the expression of NOS2 in patients with AD, using three monospecific antibodies: a previously described polyclonal and two new monoclonal antibodies. Neurofibrillary tangle-bearing neurons and neuropil threads contained NOS2 in brains from each of 11 AD patients ranging in age from 47 to 81 years. NOS2 was undetectable in brains from 6 control subjects aged 23-72 years, but was expressed in small amounts in 3 control subjects aged 77-87 years. Thus, human neurons can express NOS2 in vivo. The high-output pathway of NO production may contribute to pathogenesis in AD.


Assuntos
Doença de Alzheimer/enzimologia , Encéfalo/enzimologia , Isoenzimas/isolamento & purificação , Neurônios/enzimologia , Óxido Nítrico Sintase/isolamento & purificação , Adulto , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/patologia , Sequência de Aminoácidos , Anticorpos Monoclonais , Especificidade de Anticorpos , Encéfalo/patologia , Indução Enzimática , Feminino , Humanos , Immunoblotting , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Dados de Sequência Molecular , Neurônios/patologia
8.
J Leukoc Biol ; 60(2): 261-70, 1996 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-8773588

RESUMO

Both in vivo and in vitro experiments demonstrate that transforming growth factor-beta1 (TGF-beta1) suppresses expression of the inducible form of nitric oxide synthase (iNOS). In this study, we examined the effects of exogenous and endogenous TGF-beta1 on retinal pigment epithelial (RPE) cells and resident peritoneal macrophages ex vivo using cells from TGF-beta1 null (TGF-beta1-/-) mice or age-matched wild-type (TGF-beta1+/+) or heterozygous (TGF-beta1+/-) littermates. RPE cells from both TGF-beta1-/- mice and TGF-beta1+/+ littermates produced NO and were immunocytochemically positive for iNOS protein only following treatment with interferon-gamma (IFN-gamma) and bacterial lipopolysaccharide (LPS); however, RPE cells from TGF-beta1-/- mice produced 40% more NO than cells from TGF-beta1+/+ mice. In contrast, resident peritoneal macrophages from both TGF-beta1+/+ and TGF-beta1-/- mice expressed iNOS protein without stimulation and in the absence of detectable production of NO. The expression of iNOS was increased by treatment with IFN-gamma, resulting in detectable levels of NO. Macrophages from TGF-beta1+/+ mice appeared to produce NO in a manner inversely proportional to the serum content of NO2- and NO3- of the mice from which the cells were obtained; no such correlation existed in TGF-beta1+/- or TGF-beta1-/- mice. Treatment of RPE cells or macrophages from both TGF-beta1+/+ and TGF-beta1-/- mice with exogenous TGF-beta1 decreased both iNOS protein and NO production. These findings demonstrate a novel role of endogenous TGF-beta1 in coupling systemic NO production to the production of NO by macrophages, and demonstrate that endogenous and exogenous TGF-beta1 can act differently to suppress NO production.


Assuntos
Macrófagos Peritoneais/metabolismo , Óxido Nítrico/biossíntese , Óxido Nítrico/fisiologia , Epitélio Pigmentado Ocular/metabolismo , Fator de Crescimento Transformador beta/fisiologia , Animais , Células Cultivadas , Imunossupressores/farmacologia , Macrófagos Peritoneais/efeitos dos fármacos , Camundongos , Óxido Nítrico Sintase/metabolismo , Epitélio Pigmentado Ocular/efeitos dos fármacos , Polienos/farmacologia , Sirolimo , Fator de Crescimento Transformador beta/farmacologia
9.
J Clin Invest ; 98(3): 671-9, 1996 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-8698858

RESUMO

Captopril, an inhibitor of angiotensin converting enzyme, is widely used clinically to manage hypertension and congestive heart failure. Here captopril is shown to be an inhibitor of angiogenesis able to block neovascularization induced in the rat cornea. Captopril acted directly and specifically on capillary endothelial cells, inhibiting their chemotaxis with a biphasic dose-response curve showing an initial decrease at clinically achievable doses under 10 microM and a further slow decline in the millimolar range. Captopril inhibition of endothelial cell migration was not mediated by angiotensin converting enzyme inhibition, but was suppressed by zinc. Direct inhibition by captopril of zinc-dependent endothelial cell-derived 72-and 92-kD metalloproteinases known to be essential for angiogenesis was also seen. When used systemically on rats captopril inhibited corneal neovascularization and showed the antitumor activity expected of an inhibitor of angiogenesis, decreasing the number of mitoses present in carcinogen-induced foci of preneoplastic liver cells and slowing the growth rate of an experimental fibrosarcoma whose cells were resistant to captopril in vitro. These data define this widely used drug as a new inhibitor of neovascularization and raise the possibility that patients on long term captopril therapy may derive unexpected benefits from its antiangiogenic activities.


Assuntos
Inibidores da Enzima Conversora de Angiotensina/farmacologia , Captopril/farmacologia , Neoplasias Experimentais/tratamento farmacológico , Neovascularização Patológica/prevenção & controle , Animais , Bovinos , Movimento Celular/efeitos dos fármacos , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Feminino , Metaloendopeptidases/antagonistas & inibidores , Ratos , Ratos Endogâmicos F344
10.
J Exp Med ; 183(5): 2337-42, 1996 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-8642342

RESUMO

Transforming growth factor beta 1 null mice (TGF-beta 1-/-) suffer from multifocal inflammation and die by 3-4 wk of age. In these mice, levels of nitric oxide (NO) reaction products in serum are elevated approximately fourfold over levels in controls, peaking at 15-17 d of life. Shortterm treatment of TGF-beta 1-/- mice with NG-monomethyl-L-arginine suppressed this elevated production of NO. Expression of inducible NO synthase (iNOS) mRNA and protein is increased in the kidney and heart of TGF-beta 1-/- mice. These findings demonstrate that TGF-beta 1 negatively regulates iNOS expression in vivo, as had been inferred from mechanistic studies on the control of iNOS expression by TGF-beta 1 in vitro.


Assuntos
Arginina/análogos & derivados , Expressão Gênica/efeitos dos fármacos , Óxido Nítrico Sintase/biossíntese , Óxido Nítrico/biossíntese , Fator de Crescimento Transformador beta/deficiência , Sequência de Aminoácidos , Animais , Arginina/farmacologia , Northern Blotting , Indução Enzimática , Genótipo , Imuno-Histoquímica , Isoenzimas/análise , Isoenzimas/antagonistas & inibidores , Isoenzimas/biossíntese , Rim/enzimologia , Rim/patologia , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Miocárdio/enzimologia , NG-Nitroarginina Metil Éster , Nitratos/sangue , Óxido Nítrico Sintase/análise , Óxido Nítrico Sintase/antagonistas & inibidores , Nitritos/sangue , Fragmentos de Peptídeos/imunologia , Reação em Cadeia da Polimerase , RNA Mensageiro/análise , RNA Mensageiro/biossíntese , Valores de Referência , Fator de Crescimento Transformador beta/genética , ômega-N-Metilarginina
11.
Blood ; 86(12): 4506-15, 1995 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-8541540

RESUMO

Tissue inhibitor of metalloproteinases-1 (TIMP-1), the major physiological matrix metalloproteinase inhibitor and a potent antimetastatic factor, also stimulates the growth of erythroid progenitors (erythroid-potentiating activity). We analyzed the relationship between the growth factor activity and protease inhibition by preparing purified TIMP-1 "knockout" proteins lacking in vitro antiproteolytic activity. The growth-stimulatory effect of these N-terminal TIMP-1 point mutants, as tested in an in vitro assay using erythroid precursors (erythroid burst-forming units) was equal to that of unmutated TIMP-1. A fully antiproteolytic C-terminal TIMP-1 truncation also stimulated growth in the erythroid burst-forming unit assay. The results indicate that the influence of TIMP-1 on erythroid precursor growth is independent of its ability to inhibit metalloproteinases. TIMP-1 is analogous to proteins that have both proteolytic and growth factor activity, such as plasmin, thrombin, and urokinase. However, TIMP-1 is novel in this regard because it is a metalloproteinase inhibitor. We show that the antiproteolytic and growth factor activities of the TIMP-1 molecule are physically and functionally distinct.


Assuntos
Eritropoese/fisiologia , Glicoproteínas/fisiologia , Metaloendopeptidases/antagonistas & inibidores , Sequência de Aminoácidos , Animais , Sequência de Bases , Linhagem Celular Transformada , Células Cultivadas , Chlorocebus aethiops , DNA Complementar/genética , Células Precursoras Eritroides/efeitos dos fármacos , Eritropoese/efeitos dos fármacos , Genes myc , Glicoproteínas/química , Glicoproteínas/genética , Glicoproteínas/farmacologia , Humanos , Metaloproteinase 3 da Matriz , Metaloendopeptidases/metabolismo , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Mutação Puntual , Ligação Proteica , Conformação Proteica , Proteínas Recombinantes de Fusão/isolamento & purificação , Proteínas Recombinantes de Fusão/farmacologia , Relação Estrutura-Atividade , Inibidores Teciduais de Metaloproteinases
12.
Cancer Res ; 55(4): 721-6, 1995 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-7850780

RESUMO

Multiple copies of N-myc proto-oncogene are only rarely detected in localized neuroblastomas (NBs), and the prognostic relevance of amplification in this subset of patients is not clear. We analyzed a series of 850 children with NB admitted to a Pediatric Oncology Group NB Biology Study and identified six patients with localized NBs harboring N-myc gene amplification. Three patients whose tumors showed favorable histology by Shimada classification and low-risk histological features according to the Joshi classification have remained disease-free, whereas two of three patients with unfavorable histology tumors have developed recurrent disease. Although earlier studies have indicated that N-myc amplification is associated with diploid DNA content, flow cytometric analysis revealed that only two of the localized tumors contained stem lines with diploid DNA content. Loss of chromosome 1p was not detected by fluorescence in situ hybridization in the two tumors examined. N-myc protein was detected by immunohistochemical studies in four of the five NBs analyzed. However, N-myc protein was not visualized in one of the tumors with stroma-rich histology, and Western blot analysis revealed only low levels of N-myc protein expression in another NB with favorable histology. These studies indicate that the presence of N-myc amplification in localized NBs does not necessarily portend an adverse outcome. Furthermore, the biological features of this subset of N-myc-amplified NBs appear to differ from those of more advanced N-myc-amplified tumors.


Assuntos
Amplificação de Genes , Genes myc , Neuroblastoma/genética , Criança , Pré-Escolar , Cromossomos Humanos Par 1 , Sondas de DNA , Humanos , Imuno-Histoquímica , Lactente , Estadiamento de Neoplasias , Neuroblastoma/patologia , Ploidias , Prognóstico , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas/genética , Receptores Proteína Tirosina Quinases/genética
13.
J Cell Physiol ; 160(1): 194-202, 1994 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-7517405

RESUMO

Matrix proteases play a critical role in cell invasion and migration, including the process of angiogenesis. The ability of specific factors to induce angiogenic responses correlates with their stimulation of matrix protease synthesis and release. Using an in vivo angiogenesis assay, the endothelial cell response to known angiogenic factors, basic fibroblast growth factor (bFGF) and adipocyte conditioned medium, was blocked by an inhibitor of matrix metalloproteinase activity, TIMP-1. The TIMP effect was mediated, at least in part, through the inhibition of endothelial cell migration, as determined by the ability of TIMP to block chemotaxis in a Boyden chamber assay. These results indicate that the inhibition of migration is a direct effect on the endothelial cells and does not require accessory cells. An additional observation was that the RNA levels for TIMP were significantly reduced in differentiated adipocytes, compared to undifferentiated F442A controls. Therefore, the acquisition of an angiogenic phenotype may involve not only the induction of positive factors, but also the suppression of angiogenesis inhibitors.


Assuntos
Glicoproteínas/farmacologia , Neovascularização Patológica/genética , Adipócitos/citologia , Adipócitos/metabolismo , Adipócitos/fisiologia , Animais , Northern Blotting , Bovinos , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Movimento Celular/efeitos dos fármacos , Movimento Celular/fisiologia , Células Cultivadas , Meios de Cultivo Condicionados/análise , Meios de Cultivo Condicionados/farmacologia , Endotélio Vascular/citologia , Endotélio Vascular/fisiologia , Fator 2 de Crescimento de Fibroblastos/antagonistas & inibidores , Fator 2 de Crescimento de Fibroblastos/farmacologia , Glicerídeos/análise , Glicerídeos/antagonistas & inibidores , Glicerídeos/farmacologia , Humanos , Técnicas In Vitro , Inibidores de Metaloproteinases de Matriz , Fenótipo , Ratos , Proteínas Recombinantes/farmacologia , Inibidores Teciduais de Metaloproteinases
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...