Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
1.
Rev Neurol (Paris) ; 179(5): 490-501, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-36964121

RESUMO

Glioblastoma (GB) is the most common and aggressive primary brain tumor, with poor patient survival and lack of effective therapies. Late advances trying to decipher the composition of the GB tumor microenvironment (TME) emphasized its role in tumor progression and potentialized it as a therapeutic target. Many components participate critically to tumor development and expansion such as blood vessels, immune cells or components of the nervous system. Dysmorphic tumor vasculature brings challenges to optimal delivery of cytotoxic agents currently used in clinics. Also, massive infiltration of immunosuppressive myeloid cells and limited recruitment of T cells limits the success of conventional immunotherapies. Neuronal input seems also be required for tumor expansion. In this review, we provide a comprehensive report of vascular and immune component of the GB TME and their cross talk during GB progression.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Humanos , Glioblastoma/tratamento farmacológico , Neoplasias Encefálicas/terapia , Microambiente Tumoral
2.
Sci Adv ; 5(6): eaau6562, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31206014

RESUMO

Most achievements to engineer blood vessels are based on multiple-step manipulations such as manual sheet rolling or sequential cell seeding followed by scaffold degradation. Here, we propose a one-step strategy using a microfluidic coextrusion device to produce mature functional blood vessels. A hollow alginate hydrogel tube is internally coated with extracellular matrix to direct the self-assembly of a mixture of endothelial cells (ECs) and smooth muscle cells (SMCs). The resulting vascular structure has the correct configuration of lumen, an inner lining of ECs, and outer sheath of SMCs. These "vesseloids" reach homeostasis within a day and exhibit the following properties expected for functional vessels (i) quiescence, (ii) perfusability, and (iii) contractility in response to vasoconstrictor agents. Together, these findings provide an original and simple strategy to generate functional artificial vessels and pave the way for further developments in vascular graft and tissue engineering and for deciphering the angiogenesis process.


Assuntos
Vasos Sanguíneos/citologia , Endotélio Vascular/citologia , Células Endoteliais da Veia Umbilical Humana/citologia , Modelos Cardiovasculares , Miócitos de Músculo Liso/citologia , Engenharia Tecidual/métodos , Alginatos/química , Vasos Sanguíneos/efeitos dos fármacos , Vasos Sanguíneos/fisiologia , Linhagem Celular , Técnicas de Cocultura , Colágeno/química , Combinação de Medicamentos , Endotelina-1/farmacologia , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/fisiologia , Matriz Extracelular/química , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/fisiologia , Humanos , Hidrogéis/química , Laminina/química , Técnicas Analíticas Microfluídicas , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/fisiologia , Proteoglicanas/química , Alicerces Teciduais , Vasoconstrição/efeitos dos fármacos , Vasoconstritores/farmacologia
3.
Sci Rep ; 7(1): 10703, 2017 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-28878333

RESUMO

The chemokine receptor CXCR3 plays important roles in angiogenesis, inflammation and cancer. Activation studies and biological functions of CXCR3 are complex due to the presence of spliced isoforms. CXCR3-A is known as a pro-tumor receptor whereas CXCR3-B exhibits anti-tumor properties. Here, we focused on the conformational change of CXCR3-A and CXCR3-B after agonist or antagonist binding using Plasmon Waveguide Resonance (PWR). Agonist stimulation induced an anisotropic response with very distinct conformational changes for the two isoforms. The CXCR3 agonist bound CXCR3-A with higher affinity than CXCR3-B. Using various concentrations of SCH546738, a CXCR3 specific inhibitor, we demonstrated that low SCH546738 concentrations (≤1 nM) efficiently inhibited CXCR3-A but not CXCR3-B's conformational change and activation. This was confirmed by both, biophysical and biological methods. Taken together, our study demonstrates differences in the behavior of CXCR3-A and CXCR3-B upon ligand activation and antagonist inhibition which may be of relevance for further studies aimed at specifically inhibiting the CXCR3A isoform.


Assuntos
Conformação Proteica , Receptores CXCR3/química , Cálcio/metabolismo , Linhagem Celular , Descoberta de Drogas , Expressão Gênica , Humanos , Ligantes , Ligação Proteica , Conformação Proteica/efeitos dos fármacos , Isoformas de Proteínas , Relação Quantitativa Estrutura-Atividade , Receptores CXCR3/genética , Receptores CXCR3/metabolismo , Transdução de Sinais
4.
Ukr Biochem J ; 87(2): 76-87, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26255341

RESUMO

Hypoxia as well as the endoplasmic reticulum stress are important factors of malignant tumor growth and control of the expression of genes, which regulate numerous metabolic processes and cell proliferation. Furthermore, blockade of ERN1 (endoplasmic reticulum to nucleus 1) suppresses cell proliferation and tumor growth. We studied the effect of hypoxia on the expression of genes encoding the transcription factors such as E2F8 (E2F transcription factor 8), EPAS1 (endothelial PAS domain protein 1), TBX3 (T-box 3), ATF3 (activating transcription factor 3), FOXF1 (forkhead box F), and HOXC6 (homeobox C6) in U87 glioma cells with and without ERN1 signaling enzyme function. We have established that hypoxia enhances the expression of HOXC6, E2F8, ATF3, and EPAS1 genes but does not change TBX3 and FOXF1 gene expression in glioma cells with ERNI function. At the same time, the expression level of all studied genes is strongly decreased, except for TBX3 gene, in glioma cells without ERN1 function. Moreover, the inhibition of ERN1 signaling enzyme function significantly modifies the effect of hypoxia on the expression of these transcription factor genes. removes or introduces this regulation as well as changes a direction or magnitude of hypoxic regulation. Present study demonstrates that fine-tuning of the expression of proliferation related genes depends upon hypoxia and ERN1-mediated endoplasmic reticulum stress signaling and correlates with slower proliferation rate of glioma cells without ERN1 function.


Assuntos
Endorribonucleases/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Neuroglia/efeitos dos fármacos , Oxigênio/farmacologia , Proteínas Serina-Treonina Quinases/genética , Transdução de Sinais/efeitos dos fármacos , Fator 3 Ativador da Transcrição/genética , Fator 3 Ativador da Transcrição/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Hipóxia Celular/genética , Linhagem Celular Tumoral , Proliferação de Células , Estresse do Retículo Endoplasmático/genética , Endorribonucleases/antagonistas & inibidores , Endorribonucleases/metabolismo , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Neuroglia/metabolismo , Neuroglia/patologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Proteínas com Domínio T/genética , Proteínas com Domínio T/metabolismo , Transfecção
5.
Microvasc Res ; 89: 25-33, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23747987

RESUMO

Anti-angiogenic and anti-lymphangiogenic drugs slow tumor progression and dissemination. However, an important difficulty is that a tumor reacts and compensates to obtain the blood supply needed for tumor growth and lymphatic vessels to escape to distant loci. Therefore, there is a growing consensus on the requirement of multiple anti-(lymph)angiogenic molecules to stop cell invasion efficiently. Here we studied the cooperation between endogenous anti-angiogenic molecules, endostatin and fibstatin, and a chemokine, the Platelet Factor-4 variant 1, CXCL4L1. Anti-angiogenic factors were co-expressed by IRES-based bicistronic vectors and their cooperation was analyzed either by local delivery following transduction of pancreatic adenocarcinoma cells with lentivectors, or by distant delivery resulting from intramuscular administration in vivo of adeno-associated virus derived vectors followed by tumor subcutaneous injection. In this study, fibstatin and CXCL4L1 cooperate to inhibit endothelial cell proliferation, migration and tubulogenesis in vitro. No synergistic effect was found for fibstatin-endostatin combination. Importantly, we demonstrated for the first time that fibstatin and CXCL4L1 not only inhibit in vivo angiogenesis, but also lymphangiogenesis and tumor spread to the lymph nodes, whereas no beneficial effect was found on tumor growth inhibition using molecule combinations compared to molecules alone. These data reveal the synergy of CXCL4L1 and fibstatin in inhibition of tumor angiogenesis, lymphangiogenesis and metastasis and highlight the potential of IRES-based vectors to develop anti-metastasis combined gene therapies.


Assuntos
Proteínas de Transporte/metabolismo , Regulação Neoplásica da Expressão Gênica , Linfangiogênese/fisiologia , Proteínas de Membrana/metabolismo , Neovascularização Patológica , Fator Plaquetário 4/metabolismo , Animais , Movimento Celular , Proliferação de Células , Colágeno/química , DNA Complementar/metabolismo , Progressão da Doença , Combinação de Medicamentos , Endostatinas/metabolismo , Células Endoteliais da Veia Umbilical Humana , Humanos , Laminina/química , Camundongos , Invasividade Neoplásica , Metástase Neoplásica , Transplante de Neoplasias , Neoplasias/irrigação sanguínea , Proteoglicanas/química , Proteínas Recombinantes/metabolismo
6.
Oncogene ; 32(17): 2150-60, 2013 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-22733133

RESUMO

The tumour suppressor p53, involved in DNA repair, cell cycle arrest and apoptosis, also inhibits blood vessel formation, that is, angiogenesis, a process strongly contributing to tumour development. The p53 gene expresses 12 different proteins (isoforms), including TAp53 (p53 (or p53α), p53ß and p53γ) and Δ133p53 isoforms (Δ133p53α, Δ133p53ß and Δ133p53γ). The Δ133p53α isoform was shown to modulate p53 transcriptional activity and is overexpressed in various human tumours. However, its role in tumour progression is still unexplored. In the present study, we examined the involvement of Δ133p53 isoforms in tumoural angiogenesis and tumour growth in the highly angiogenic human glioblastoma U87. Our data show that conditioned media from U87 cells depleted for Δ133p53 isoforms block endothelial cell migration and tubulogenesis without affecting endothelial cell proliferation in vitro. The Δ133p53 depletion in U2OS osteosarcoma cells resulted in a similar angiogenesis blockade. Furthermore, using conditioned media from U87 cells ectopically expressing each Δ133p53 isoform, we determined that Δ133p53α and Δ133p53γ but not Δ133p53ß, stimulate angiogenesis. Our in vivo data using the chicken chorio-allantoic membrane and mice xenografts establish that angiogenesis and growth of glioblastoma U87 tumours are inhibited upon depletion of Δ133p53 isoforms. By TaqMan low-density array, we show that alteration of expression ratio of Δ133p53 and TAp53 isoforms differentially regulates angiogenic gene expression with Δ133p53 isoforms inducing pro-angiogenic gene expression and repressing anti-angiogenic gene expression.


Assuntos
Neoplasias Encefálicas/irrigação sanguínea , Glioblastoma/irrigação sanguínea , Neovascularização Patológica/metabolismo , Proteína Supressora de Tumor p53/genética , Proteínas Angiogênicas/genética , Proteínas Angiogênicas/metabolismo , Animais , Neoplasias Encefálicas/patologia , Bovinos , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Embrião de Galinha , Membrana Corioalantoide/irrigação sanguínea , Membrana Corioalantoide/metabolismo , Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Glioblastoma/patologia , Células Endoteliais da Veia Umbilical Humana/metabolismo , Células Endoteliais da Veia Umbilical Humana/fisiologia , Humanos , Camundongos , Camundongos Nus , Transplante de Neoplasias , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Carga Tumoral , Proteína Supressora de Tumor p53/metabolismo
7.
BMC Med ; 8: 19, 2010 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-20334641

RESUMO

BACKGROUND: Amifostine (WR-2721, delivered as Ethyol) is a phosphorylated aminothiol compound clinically used in addition to cis-platinum to reduce the toxic side effects of therapeutic treatment on normal cells without reducing their efficacy on tumour cells. Its mechanism of action is attributed to the free radical scavenging properties of its active dephosphorylated metabolite WR-1065. However, amifostine has also been described as a potent hypoxia-mimetic compound and as a strong p53 inducer; both effects are known to potently modulate vascular endothelial growth factor (VEGF-A) expression. The angiogenic properties of this drug have not been clearly defined. METHODS: Cancer cell lines and endothelial cells were used in culture and treated with Amifostine in order to study (i) the expression of angiogenesis related genes and proteins and (ii) the effects of the drug on VEGF-A induced in vitro angiogenesis. RESULTS: We demonstrated that the treatment of several human cancer cell lines with therapeutical doses of WR-1065 led to a strong induction of different VEGF-A mRNA isoforms independently of HIF-1alpha. VEGF-A induction by WR-1065 depends on the activation of the eIF2alpha/ATF4 pathway. This up-regulation of VEGF-A mRNA was accompanied by an increased secretion of VEGF-A proteins fully active in stimulating vascular endothelial cells (EC). Nevertheless, direct treatment of EC with amifostine impaired their ability to respond to exogenous VEGF-A, an effect that correlated to the down-regulation of VEGFR-2 expression, to the reduction in cell surface binding of VEGF-A and to the decreased phosphorylation of the downstream p42/44 kinases. CONCLUSIONS: Taken together, our results indicate that amifostine treatment modulates tumour angiogenesis by two apparently opposite mechanisms - the increased VEGF-A expression by tumour cells and the inhibition of EC capacity to respond to VEGF-A stimulation.


Assuntos
Amifostina/farmacologia , Moduladores da Angiogênese/farmacologia , Sequestradores de Radicais Livres/farmacologia , Expressão Gênica/efeitos dos fármacos , Ativação Transcricional/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/biossíntese , Fator A de Crescimento do Endotélio Vascular/metabolismo , Linhagem Celular , Células Cultivadas , Humanos , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/biossíntese
8.
Oncogene ; 28(2): 243-56, 2009 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-18850004

RESUMO

Cancer cells have complex, unique characteristics that distinguish them from normal cells, such as increased growth rates and evasion of anti-proliferative signals. Global inhibition of class I and II histone deacetylases (HDACs) stops cancer cell proliferation in vitro and has proven effective against cancer in clinical trials, at least in part, through transcriptional reactivation of the p21(WAF1/Cip1)gene. The HDACs that regulate p21(WAF1/Cip1) are not fully identified. Using small interfering RNAs, we found that HDAC4 participates in the repression of p21(WAF1/Cip1) through Sp1/Sp3-, but not p53-binding sites. HDAC4 interacts with Sp1, binds and reduces histone H3 acetylation at the Sp1/Sp3 binding site-rich p21(WAF1/Cip1) proximal promoter, suggesting a key role for Sp1 in HDAC4-mediated repression of p21(WAF1/Cip1). Induction of p21(WAF1/Cip1) mediated by silencing of HDAC4 arrested cancer cell growth in vitro and inhibited tumor growth in an in vivo human glioblastoma model. Thus, HDAC4 could be a useful target for new anti-cancer therapies based on selective inhibition of specific HDACs.


Assuntos
Inibidor de Quinase Dependente de Ciclina p21/biossíntese , Regulação Neoplásica da Expressão Gênica , Histona Desacetilases/fisiologia , Proteínas de Neoplasias/fisiologia , Proteínas Repressoras/fisiologia , Fator de Transcrição Sp1/fisiologia , Acetilação , Animais , Sítios de Ligação , Neoplasias Ósseas/patologia , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral/metabolismo , Embrião de Galinha , Inibidor de Quinase Dependente de Ciclina p21/genética , Epigênese Genética , Técnicas de Silenciamento de Genes , Glioblastoma/tratamento farmacológico , Glioblastoma/patologia , Células HeLa/efeitos dos fármacos , Células HeLa/metabolismo , Inibidores de Histona Desacetilases , Histonas/metabolismo , Humanos , Proteínas de Neoplasias/antagonistas & inibidores , Osteossarcoma/patologia , Processamento de Proteína Pós-Traducional , RNA Mensageiro/biossíntese , RNA Neoplásico/biossíntese , Proteínas Repressoras/antagonistas & inibidores , Proteína Supressora de Tumor p53/antagonistas & inibidores , Proteína Supressora de Tumor p53/fisiologia
9.
Ukr Biokhim Zh (1999) ; 80(1): 19-25, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18710022

RESUMO

Expression of 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase-3 (PFKFB-3) mRNA alternative splice variants was studied in different mouse tissues in hypoxic conditions in vivo. Significant increase of the expression of PFKFB-3 mRNA was observed in the mouse lungs, testes and brain in hypoxia. Several new alternative splice variants of PFKFB-3 mRNA were identified in the lung, testis, brain and skeletal muscle. They have different length and amino acid sequence of C-terminal regulatory part. However, 6-phosphofructo-2-kinase and fructose-2,6-bisphosphatase catalytic domains were identical. Moreover, the expression of different alternative splice variants of PFKFB-3 mRNA has shown tissue specificity and different levels of induction in hypoxic conditions in vivo. Results of this investigation indicate a possible role of PFKFB-3 splice isoform in cell adaptation to hypoxic conditions.


Assuntos
Adaptação Fisiológica/genética , Processamento Alternativo , Expressão Gênica , Hipóxia , Fosfofrutoquinase-2/genética , RNA Mensageiro/genética , Processamento Alternativo/fisiologia , Sequência de Aminoácidos , Animais , Encéfalo/enzimologia , Expressão Gênica/fisiologia , Hipóxia/enzimologia , Hipóxia/genética , Hipóxia/fisiopatologia , Isoenzimas/genética , Pulmão/enzimologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Músculo Esquelético/enzimologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Testículo/enzimologia
10.
Oncogene ; 27(37): 4973-85, 2008 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-18469859

RESUMO

The lysosomal cysteine proteinase cathepsin L is involved in proteolytic processing of internalized proteins. In transformed cells, where it is frequently overexpressed, its intracellular localization and functions can be altered. Previously, we reported that treatment of highly metastatic, murine carcinoma H-59 cells with small molecule cysteine proteinase inhibitors altered the responsiveness of the type I insulin-like growth factor (IGF-I) receptor and consequently reduced cell invasion and metastasis. To assess more specifically the role of cathepsin L in IGF-I-induced signaling and tumorigenicity, we generated H-59 subclones with reduced cathepsin L expression levels. These clonal lines showed an altered responsiveness to IGF-I in vitro, as evidenced by (i) loss of IGF-I-induced receptor phosphorylation and Shc recruitment, (ii) reduced IGF-I (but not IGF-II)-induced cellular proliferation and migration, (iii) decreased anchorage-independent growth and (iv) reduced plasma membrane levels of IGF-IR. These changes resulted in increased apoptosis in vivo and an impaired ability of the cells to form liver metastases. The results demonstrate that cathepsin L expression levels regulate cell responsiveness to IGF-I and thereby identify a novel function for cathepsin L in the control of the tumorigenic/metastatic phenotype.


Assuntos
Carcinoma/patologia , Catepsinas/metabolismo , Cisteína Endopeptidases/metabolismo , Fator de Crescimento Insulin-Like I/uso terapêutico , Neoplasias Hepáticas/prevenção & controle , Neoplasias Hepáticas/secundário , Neoplasias Pulmonares/patologia , Animais , Carcinoma/metabolismo , Catepsina L , Catepsinas/antagonistas & inibidores , Adesão Celular/efeitos dos fármacos , Regulação para Baixo , Fator de Crescimento Insulin-Like I/farmacologia , Neoplasias Hepáticas/metabolismo , Neoplasias Pulmonares/metabolismo , Camundongos , Modelos Biológicos , Invasividade Neoplásica , Transplante de Neoplasias , RNA Interferente Pequeno/farmacologia , Células Tumorais Cultivadas
11.
Curr Pharm Biotechnol ; 8(1): 3-9, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17311548

RESUMO

Angiogenesis is a developmental process that also plays the central role in adults during the female menstruation cycle, wound healing and neoplastic growth and metastasis. Ideally, blocking neovessel growth starves the developing tumor and induces tumor regression. Restricting the vascular ingrowth into the tumor might have adverse effect on drugs targeting the tumor. Nevertheless, anti-VEGF treatment of the neoplastic diseases when combined with chemotherapy significantly increases median survival in treated patients. This suggests alternative mechanisms of anti-angiogenesis therapy. A number of molecules that are in current clinical trials have been identified using angiogenesis models. However, current angiogenesis models have advantages and inconvenience and conclusion drawn upon their use should be interpreted with caution. Thus, it is necessary to optimize existing models and to develop new ones that take into account the complexity of the angiogenic process as it happens in many angiogenesis-related diseases and in particular in cancer.


Assuntos
Inibidores da Angiogênese/administração & dosagem , Proteínas Angiogênicas/metabolismo , Modelos Biológicos , Neoplasias/irrigação sanguínea , Neoplasias/fisiopatologia , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/fisiopatologia , Animais , Humanos , Neoplasias/tratamento farmacológico
13.
J Neurosurg Sci ; 47(2): 69-78, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-14618134

RESUMO

AIM: The growth of gliomas depends on the balance of factors stimulating or inhibiting angiogenesis, tumor cell invasion and proliferation. The administration of endogenous inhibitors to experimental human gliomas in animal models resulted in a significant inhibition of tumor growth. It is becoming apparent that resistance can develop over time to many types of endogenous inhibitors and seems to be influenced by the tumor type and system of delivery. METHODS: We recently isolated a potent endogenous inhibitor, called human PEX, from human glioma cells in culture. Human PEX is a potent inhibitor of angiogenesis, tumor and endothelial cell proliferation and migration. In this paper, we investigated the ability of human PEX to sustain inhibition of glioma growth for a prolonged period of time. We initially developed a recombinant form of the inhibitor and showed that this form had similar in vitro and in vivo activities to the natural one. Human PEX was then administered to nude mice intracranial human glioma model, in combination with metronomic chemotherapy, for a period of 185 days, starting 15 days after tumor cells implantation. RESULTS: Our data showed that the systemic administration of human PEX mantained a very prolonged inhibition of glioma growth (50% survival of animals treated with 2 mg/kg/days was 160 days vs 24 days of the control) and had a synergistic effect with low dose chemotherapy. Histological analysis of tumors, showed that treatment with PEX was associated with a decrease of vascularity, cell proliferation, and increase in apoptosis. CONCLUSION: These data indicate that human PEX controls tumor growth by separate mechanisms. In addition, treatment with PEX produced well delineated tumors, indicating the persistence of a direct anti-invasive effect of the molecule even after a prolonged period of treatment.


Assuntos
Inibidores da Angiogênese/farmacologia , Células Endoteliais/efeitos dos fármacos , Glioma/tratamento farmacológico , Metaloproteinase 2 da Matriz/farmacologia , Fragmentos de Peptídeos/farmacologia , Animais , Apoptose/efeitos dos fármacos , Western Blotting , Divisão Celular/efeitos dos fármacos , Modelos Animais de Doenças , Glioma/patologia , Humanos , Masculino , Camundongos , Camundongos Nus , Transplante de Neoplasias , Reação em Cadeia da Polimerase , Proteínas Recombinantes/farmacologia , Fatores de Tempo , Células Tumorais Cultivadas
14.
Cancer Res ; 61(24): 8730-6, 2001 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-11751392

RESUMO

Angiogenesis, tumor cell proliferation, and migration are the hallmarks of solid tumors, such as gliomas. This study demonstrates that a fragment derived from the autocatalytic digestion of matrix metalloproteinase (MMP)-2, called PEX, acts simultaneously as an inhibitor of glioma angiogenesis, cell proliferation, and migration. PEX is detected in the cultured medium of various human glioma, endothelial, breast, and prostate carcinoma cell lines. PEX is purified from the medium of glioma cell lines by chromatography, where PEX is constitutively expressed as a free and a TIMP-2-bound form. In human glioma tissue, PEX expression correlates with histological subtype and grade and with alpha v beta 3 integrin expression to which it is bound. Systemic administration of PEX to s.c. and intracranial human glioma xenografts results in a 99% suppression of tumor growth with no signs of toxicity. Thus, PEX is a very promising candidate for the treatment of human malignant gliomas.


Assuntos
Neoplasias Encefálicas/irrigação sanguínea , Neoplasias Encefálicas/tratamento farmacológico , Glioma/irrigação sanguínea , Glioma/tratamento farmacológico , Metaloproteinase 2 da Matriz/farmacologia , Neovascularização Patológica/tratamento farmacológico , Fragmentos de Peptídeos/farmacologia , Adulto , Idoso , Animais , Apoptose/efeitos dos fármacos , Neoplasias Encefálicas/enzimologia , Neoplasias Encefálicas/patologia , Adesão Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Meios de Cultura , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Feminino , Fator 2 de Crescimento de Fibroblastos/metabolismo , Glioma/enzimologia , Glioma/patologia , Humanos , Masculino , Metaloproteinase 2 da Matriz/biossíntese , Metaloproteinase 2 da Matriz/isolamento & purificação , Camundongos , Camundongos Nus , Pessoa de Meia-Idade , Invasividade Neoplásica , Fragmentos de Peptídeos/biossíntese , Fragmentos de Peptídeos/isolamento & purificação , Receptores de Vitronectina/biossíntese , Receptores de Vitronectina/metabolismo , Vitronectina/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
15.
Cancer Res ; 61(20): 7501-6, 2001 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-11606386

RESUMO

This study evaluates the efficacy of the combination of an antiangiogenic drug and conventional chemotherapeutics for the treatment of experimental human gliomas. As an antiangiogenic, we used recombinant human PEX, a fragment of matrix metalloproteinase-2 that we have previously shown to have a significant antimitotic, anti-invasive, and antiangiogenic properties against human glioblastoma in vitro and in vivo. We used carboplatin and etoposide as the two chemotherapeutic drugs routinely used in our institution (Ospedale Maggiore de Milano) for the treatment of malignant gliomas. Conventional chemotherapeutic drugs were administered at high dose or at a low and semicontinuous regimen. Combined treatment of high-dose chemotherapy and PEX did not produce an improvement of survival in comparison with chemotherapy alone, but it was associated with a decrease in tumor volume, vascularity, and proliferative index and an increased apoptosis. All of these animals experienced severe side effects. The longest survival was documented in animals submitted to low and semicontinuous chemotherapy and antiangiogenic treatment. This regimen was associated with no side effects, marked decrease in tumor volume, vascularity, and proliferative index, and an increased apoptosis. Our data suggest that low-dose chemotherapy in combination with PEX can be successfully used against human malignant glioma in vivo.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Neoplasias Encefálicas/tratamento farmacológico , Glioblastoma/tratamento farmacológico , Neovascularização Patológica/tratamento farmacológico , Inibidores da Angiogênese/administração & dosagem , Inibidores da Angiogênese/efeitos adversos , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Neoplasias Encefálicas/irrigação sanguínea , Neoplasias Encefálicas/patologia , Carboplatina/administração & dosagem , Carboplatina/efeitos adversos , Divisão Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Etoposídeo/administração & dosagem , Etoposídeo/efeitos adversos , Glioblastoma/irrigação sanguínea , Glioblastoma/patologia , Humanos , Masculino , Metaloproteinase 2 da Matriz/administração & dosagem , Camundongos , Camundongos Nus , Fragmentos de Peptídeos/administração & dosagem , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/efeitos adversos , Ensaios Antitumorais Modelo de Xenoenxerto
16.
J Biol Chem ; 276(38): 35723-34, 2001 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-11423536

RESUMO

Platelet factor-4 is a protein belonging to the family of ELR-negative CXC chemokines which binds to fibroblast growth factor and inhibits its mitogenic activity. Platelet factor-4 also inhibits tumor growth by mechanisms involving antiangiogenesis. Antiangiogenic activity in vitro has also been shown for the 24-residue C-terminal fragment of the protein, which decreases the affinity between basic fibroblast growth factor and its cell-surface receptor. In this study, the preferential conformation of this fragment in solution has been determined and has been found to be composed of two helical subdomains. In addition, we show that the fragment forms a specific 1:1 complex with acidic and basic fibroblast growth factors and that both subdomains are probably required for inhibition of fibroblast growth factor-driven mitogenesis. Finally, we show that the binding of the fragment alters the structure of the fibroblast growth factors, although some of such alterations do not seem related with the inhibition of mitogenic activity. Since this fragment has recently been shown to inhibit fibroblast growth factor-induced angiogenesis in vivo when injected intraperitoneally, these results are relevant for developing new antiangiogenic treatments.


Assuntos
Inibidores da Angiogênese/metabolismo , Fator 1 de Crescimento de Fibroblastos/metabolismo , Fator 2 de Crescimento de Fibroblastos/metabolismo , Fator Plaquetário 4/metabolismo , Sequência de Aminoácidos , Inibidores da Angiogênese/química , Inibidores da Angiogênese/farmacologia , Fluorescência , Humanos , Dados de Sequência Molecular , Ressonância Magnética Nuclear Biomolecular , Fator Plaquetário 4/química , Fator Plaquetário 4/farmacologia , Estrutura Secundária de Proteína , Soluções
17.
Cancer Res ; 61(4): 1717-26, 2001 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-11245488

RESUMO

We undertook a series of systematic studies to address the role of fibroblast growth factor/fibroblast growth factor receptor (FGF/FGFR) activity in tumor growth and angiogenesis. We expressed dominant-negative FGFR2 (FGFR2-DN) or FGFR1 (FGFR1-DN) in glioma C6 cells by using constitutive or tetracycline-regulated expression systems. Anchorage-dependent or independent growth was inhibited in FGFR-DN-expressing cells. Tumor development after xenografting FGFR-DN-expressing cells in immunodeficient mice or after transplantation in rat brain was strongly inhibited. Quantification of microvessels demonstrated a significant decrease in vessel density in tumors derived from FGFR-DN-expressing cells. Furthermore, in a rabbit corneal assay, the angiogenic response after implantation of FGFR-DN-expressing cells was decreased. In tumors expressing FGFR-DN, vascular endothelial growth factor expression was strongly inhibited as compared with control tumor. These results indicate that inhibition of FGF activity may constitute a dominant therapeutic strategy in the treatment of FGF-producing cerebral malignancies and may disrupt both angiogenesis-dependent and -independent signals required for glioma growth and invasion.


Assuntos
Neoplasias Encefálicas/irrigação sanguínea , Neoplasias Encefálicas/patologia , Fatores de Crescimento de Fibroblastos/antagonistas & inibidores , Glioma/irrigação sanguínea , Glioma/patologia , Neovascularização Patológica/patologia , Receptores de Fatores de Crescimento de Fibroblastos/antagonistas & inibidores , Animais , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Divisão Celular/fisiologia , Fatores de Crescimento Endotelial/biossíntese , Fatores de Crescimento Endotelial/genética , Fator 2 de Crescimento de Fibroblastos/biossíntese , Fator 2 de Crescimento de Fibroblastos/genética , Fator 4 de Crescimento de Fibroblastos , Fatores de Crescimento de Fibroblastos/biossíntese , Fatores de Crescimento de Fibroblastos/genética , Fatores de Crescimento de Fibroblastos/fisiologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glioma/genética , Glioma/metabolismo , Linfocinas/biossíntese , Linfocinas/genética , Masculino , Camundongos , Neovascularização Patológica/metabolismo , Fenótipo , Proteínas Proto-Oncogênicas/biossíntese , Proteínas Proto-Oncogênicas/genética , Coelhos , Ratos , Ratos Sprague-Dawley , Receptores Proteína Tirosina Quinases/biossíntese , Receptores Proteína Tirosina Quinases/genética , Receptores Proteína Tirosina Quinases/fisiologia , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos , Receptores de Fatores de Crescimento de Fibroblastos/biossíntese , Receptores de Fatores de Crescimento de Fibroblastos/genética , Receptores de Fatores de Crescimento de Fibroblastos/fisiologia , Tetraciclina/farmacologia , Transfecção , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
18.
FASEB J ; 15(3): 550-2, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11259363

RESUMO

Platelet factor 4 (PF-4) is a CXC-chemokine with strong anti-angiogenic properties. We have shown previously that PF-4 inhibits angiogenesis by associating directly with fibroblast growth factor 2 (FGF-2), inhibiting its dimerization, and blocking FGF-2 binding to endothelial cells. We now have characterized a small peptide domain (PF-447-70) derived from the C-terminus of PF-4, which conserves anti-angiogenic effects of the parent protein. PF-447-70 inhibited internalization of 125I-FGF-2 by endothelial cells in a time-dependent manner. The peptide reduced FGF-2-stimulated cell migration to control levels in wounded monolayers of bovine capillary endothelial cells. PF-447-70 also reduced FGF-2 induced phosphorylation of MAP kinases ERK-1 and ERK-2, which are essential for migration and survival of endothelial cells. In a serum-free ex vivo angiogenesis assay, the peptide blocked microvessel outgrowth by 89%. A single amino acid substitution within PF-447-70 abolished all inhibitory activities. To simulate a real anti-angiogenic treatment situation, we administered PF-447-70 systemically to mice implanted subcutaneously with FGF-2 containing gelatin sponges with the result of sparse, scattered, and immature vessel growth. The small peptide fragment derived from the angio-inhibitory CXC-chemokine PF-4 might be used as a starting point to develop anti-angiogenic designer drugs for angiogenesis-dependent pathologies such as cancer, diabetic retinopathy, and rheumatoid arthritis.


Assuntos
Endotélio Vascular/citologia , Fator 2 de Crescimento de Fibroblastos/metabolismo , Neovascularização Fisiológica/efeitos dos fármacos , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/farmacologia , Fator Plaquetário 4/química , Fator Plaquetário 4/farmacologia , Sequência de Aminoácidos , Animais , Aorta , Divisão Celular , Movimento Celular , Células Cultivadas , Meios de Cultura Livres de Soro , Técnicas de Cultura , Endotélio Vascular/efeitos dos fármacos , Ativação Enzimática , Humanos , Camundongos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Modelos Biológicos , Dados de Sequência Molecular , Fragmentos de Peptídeos/genética , Fator Plaquetário 4/genética , Estrutura Terciária de Proteína , Ratos , Receptores Proteína Tirosina Quinases/metabolismo , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo
19.
Exp Eye Res ; 71(4): 395-404, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10995560

RESUMO

Fibroblast growth factors (FGF) are multipotent cytokines with demonstrated mitogenic, neurotrophic and angiogenic properties. There is evidence that they have multiple functions during and after development of the vertebrate eye. Amongst these, the role of FGF receptor mediated signaling in the retinal pigmented epithelium (RPE) is not yet well understood. FGF-2 is produced in RPE cells and may play a role in photoreceptor development and/or survival in vivo. It may also stimulate growth of melanocytes and angiogenesis in the choroid. To address these questions, we have specifically disrupted FGF signaling by generating lines of transgenic mice expressing dominant-negative FGF receptor 1 (FGFR-1) in the pigmented cells. Histological analysis of the eyes were conducted on hemizygous and homozygous mice at different ages. In homozygotes, eye growth is strongly impaired during embryogenesis leading to massive eye degeneration seen in the early post-natal stages. In hemizygotes, the choroid is thinned and the finger-like junctions between RPE cells and photoreceptors are disrupted. Scanning electron microscopy of the choroid vasculature showed that choriocapillary density, diameter and branching are strongly affected. As mice age, they develop progressive retinal degeneration as evidenced by photoreceptor cell loss. Our results are in agreement with the hypothesis that FGF signaling in the RPE participates in photoreceptor survival in vivo. Our model provides evidence that FGF signaling is also involved in choroidal angiogenesis by a process that could relate to induction of terminal branching.


Assuntos
Anormalidades do Olho/genética , Epitélio Pigmentado Ocular/fisiologia , Receptores de Fatores de Crescimento de Fibroblastos/fisiologia , Animais , Comunicação Celular/fisiologia , Sobrevivência Celular , Olho/irrigação sanguínea , Camundongos , Camundongos Transgênicos , Microscopia Eletrônica de Varredura , Neovascularização Fisiológica , Células Fotorreceptoras de Vertebrados/fisiologia , Epitélio Pigmentado Ocular/citologia , Degeneração Retiniana/genética , Degeneração Retiniana/patologia
20.
Crit Rev Oncol Hematol ; 34(2): 89-110, 2000 May.
Artigo em Inglês | MEDLINE | ID: mdl-10799835

RESUMO

There is growing evidence that anti-angiogenic drugs will improve future therapies of diseases like cancer, rheumatoid arthritis and ocular neovascularisation. However, it is still uncertain which kind of substance, out of the large number of angiogenesis inhibitors, will prove to be a suitable agent to treat these human diseases. There are currently more than 30 angiogenesis inhibitors in clinical trials and a multitude of promising new candidates are under investigation in vitro and in animal models. Important therapeutic strategies are: suppression of activity of the major angiogenic regulators like vascular endothelial growth factor (VEGF) and fibroblast growth factor (FGF); inhibition of function of alphav-integrins and matrix metalloproteinases (MMPs); the exploitation of endogenous anti-angiogenic molecules like angiostatin, endostatin or thrombospondin. Given the wide spectrum of diseases which could be treated by anti-angiogenic compounds, it is important for today's clinicians to understand their essential mode of action at a cellular and molecular level. Here we give an in-depth overview of the basic pathophysiological mechanisms underlying the different anti-angiogenic approaches used to date based on the most recent fundamental and clinical research data. The angiogenesis inhibitors in clinical trials are presented and promising future drug candidates are discussed.


Assuntos
Inibidores da Angiogênese/farmacologia , Inibidores da Angiogênese/uso terapêutico , Endotélio Vascular/efeitos dos fármacos , Animais , Ensaios Clínicos como Assunto , Endotélio Vascular/citologia , Endotélio Vascular/fisiologia , Substâncias de Crescimento/farmacologia , Humanos , Projetos de Pesquisa
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA