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










Base de dados
Intervalo de ano de publicação
1.
Cancers (Basel) ; 12(10)2020 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-33076448

RESUMO

Neuroblastoma is the most common extra-cranial solid tumor in infants and children, which accounts for approximately 15% of all cancer-related deaths in the pediatric population. New therapeutic modalities are urgently needed. Antibody-Drug Conjugates (ADC)s-based therapy has been proposed as potential strategy to treat this pediatric malignancy. LGALS3BP is a highly glycosylated protein involved in tumor growth and progression. Studies have shown that LGALS3BP is enriched in extracellular vesicles (EV)s derived by most neuroblastoma cells, where it plays a critical role in preparing a favorable tumor microenvironment (TME) through direct cross talk between cancer and stroma cells. Here, we describe the development of a non-internalizing LGALS3BP ADC, named 1959-sss/DM3, which selectively targets LGALS3BP expressing neuroblastoma. 1959-sss/DM3 mediated potent therapeutic activity in different types of neuroblastoma models. Notably, we found that treatments were well tolerated at efficacious doses that were fully curative. These results offer preclinical proof-of-concept for an ADC targeting exosomal LGALS3BP approach for neuroblastomas.

2.
Sci Rep ; 9(1): 10505, 2019 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-31324853

RESUMO

P-glycoprotein (P-gp) largely influences the pharmacokinetics (PK) and toxicities of xenobiotics in a patient-specific manner so that personalized drug scheduling may lead to significant patient's benefit. This systems pharmacology study investigated P-gp activity in mice according to organ, sex, feeding status, and circadian time. Sex-specific circadian changes were found in P-gp ileum mRNA and protein levels, circadian amplitudes being larger in females as compared to males. Plasma, ileum and liver concentrations of talinolol, a pure P-gp substrate, significantly differed according to sex, feeding and circadian timing. A physiologically-based PK model was designed to recapitulate these datasets. Estimated mesors (rhythm-adjusted mean) of ileum and hepatic P-gp activity were higher in males as compared to females. Circadian amplitudes were consistently higher in females and circadian maxima varied by up to 10 h with respect to sex. Fasting increased P-gp activity mesor and dampened its rhythm. Ex-vivo bioluminescence recordings of ileum mucosae from transgenic mice revealed endogenous circadian rhythms of P-gp protein expression with a shorter period, larger amplitude, and phase delay in females as compared to males. Importantly, this study provided model structure and parameter estimates to refine PK models of any P-gp substrate to account for sex, feeding and circadian rhythms.


Assuntos
Subfamília B de Transportador de Cassetes de Ligação de ATP/metabolismo , Ritmo Circadiano , Citalopram/farmacocinética , Ingestão de Alimentos/fisiologia , Jejum/fisiologia , Propanolaminas/farmacocinética , Caracteres Sexuais , Subfamília B de Transportador de Cassetes de Ligação de ATP/genética , Animais , Transporte Biológico , Colo/metabolismo , Cruzamentos Genéticos , Feminino , Regulação da Expressão Gênica , Íleo/metabolismo , Mucosa Intestinal/metabolismo , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos , Propanolaminas/análise , RNA Mensageiro/biossíntese
3.
J Control Release ; 294: 176-184, 2019 01 28.
Artigo em Inglês | MEDLINE | ID: mdl-30553852

RESUMO

Galectin-3-binding protein (Gal-3BP) has been identified as a cancer and metastasis-associated, secreted protein that is expressed by the large majority of cancers. The present study describes a special type of non-internalizing antibody-drug-conjugates that specifically target Gal-3BP. Here, we show that the humanized 1959 antibody, which specifically recognizes secreted Gal-3BP, selectively localized around tumor but not normal cells. A site specific disulfide linkage with thiol-maytansinoids to unpaired cysteine residues of 1959, resulting in a drug-antibody ratio of 2, yielded an ADC product, which cured A375m melanoma bearing mice. ADC products based on the non-internalizing 1959 antibody may be useful for the treatment of several human malignancies, as the cognate antigen is abundantly expressed and secreted by several cancers, while being present at low levels in most normal adult tissues.


Assuntos
Antígenos de Neoplasias/imunologia , Biomarcadores Tumorais/imunologia , Imunoconjugados/uso terapêutico , Neoplasias/terapia , Animais , Linhagem Celular Tumoral , Feminino , Humanos , Imunoconjugados/farmacocinética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias/imunologia , Neoplasias/metabolismo , Neoplasias/patologia , Coelhos
4.
Oncotarget ; 8(36): 60368-60377, 2017 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-28947977

RESUMO

The endosialin/CD248/TEM1 receptor is expressed on the cell surface of tumor-associated stroma cells as well as in sarcoma and neuroblastoma cells. This receptor is emerging as an attractive molecule in diagnostics and therapeutics because of its expression across the stroma of many human tumors, the low to absent expression in normal tissues and accessibility from the vascular circulation. In this study, we present evidence of the preclinical efficacy of a novel Antibody-Drug Conjugate (ENDOS/ADC). It consists of a humanized endosialin monoclonal antibody, named hMP-E-8.3, conjugated to a potent duocarmycin derivative. In endosialin expressing cancer cell lines, this ENDOS/ADC showed a powerful, specific and target-dependent killing activity. High expression levels of endosialin in cells correlated with efficient internalization and cytotoxic effects in vitro. Efficacy studies demonstrated that ENDOS/ADC treatment led to a long-lasting tumor growth inhibition of a cell line-based model of human osteosarcoma. Taken together, our results demonstrate that endosialin is an attractive target in sarcoma and suggest that ENDOS/ADC has the potential to be developed into a bio-therapeutic agent for these malignancies.

5.
J Transl Med ; 13: 248, 2015 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-26219351

RESUMO

BACKGROUND: A previous report has shown that LGALS3BP (also known as 90K or Mac-2 BP) has antitumor activity in colorectal cancer (CRC) via suppression of Wnt signalling with a novel mechanism of ISGylation-dependent ubiquitination of ß-catenin. The role of LGALS3BP in CRC prognosis was investigated. METHODS: The role of LGALS3BP on CRC progression and clinical prognosis was analyzed by combining cell cultures, in vitro assays, and immunohistochemistry. RESULTS: Silencing of LGALS3BP in HCT-116 human colon cancer cells resulted in enhanced ß-catenin expression that was reversed by addition of human recombinant LGALS3BP. Moreover, intra-tumor delivery of LGALS3BP reduced tumor growth of xenografts originating from LGALS3BP-silenced HCT-116 cells. Finally, in a series of 196 CRC patients, LGALS3BP expression in tumor tissue associated with clinical outcome. Patients with high LGALS3BP expression had lower risk of relapse and a longer overall survival time than those with low LGALS3BP expression. Multivariate analyses confirmed LGALS3BP expression status as the only independent prognostic factor of survival. CONCLUSIONS: These results provide evidence that low expression of LGALS3BP participates in malignant progression of CRC and implicates poor prognosis, highlighting its augmentation as a potential therapeutic approach.


Assuntos
Antígenos de Neoplasias/metabolismo , Biomarcadores Tumorais/metabolismo , Proteínas de Transporte/metabolismo , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Glicoproteínas/metabolismo , Animais , Proliferação de Células , Regulação para Baixo , Feminino , Técnicas de Silenciamento de Genes , Inativação Gênica , Células HCT116 , Células HEK293 , Humanos , Imuno-Histoquímica , Injeções Intralesionais , Estimativa de Kaplan-Meier , Camundongos Nus , Análise Multivariada , Prognóstico , Resultado do Tratamento , Ensaios Antitumorais Modelo de Xenoenxerto , beta Catenina/metabolismo
6.
Mol Cancer Ther ; 13(4): 916-25, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24552775

RESUMO

Accumulating evidence indicates that serum and tissue levels of lectin, galactoside-binding soluble 3 binding protein (LGALS3BP), a secreted glycoprotein, are elevated in human cancers. Recently, we have identified LGALS3BP as a factor capable of stimulating angiogenesis of microvascular endothelial cells in vitro as well as in vivo. However, the potential therapeutic implications of LGALS3BP function blockade have not been explored yet. Here, we tested the ability of an anti-LGALS3BP mouse monoclonal antibody, SP-2, to antagonize LGALS3BP-induced angiogenesis and tumor growth. The antibody was found to inhibit endothelial cell tubulogenesis induced by either conditioned medium of breast cancer and melanoma cells or human recombinant LGALS3BP. In addition, SP-2 inhibited phosphorylation of FAK and its recruitment to membrane sites as well as AKT and ERK phosphorylation promoted by LGALS3BP. When used in vivo, the antibody restrained LGALS3BP-stimulated angiogenesis and growth of tumor xenografts. Furthermore, the combination of SP-2 and low-dose bevacizumab was more effective than either agent alone. Taken together, these results lead to consideration of SP-2 as a promising candidate for LGALS3BP-targeted therapy.


Assuntos
Anticorpos Monoclonais/farmacologia , Antineoplásicos/farmacologia , Biomarcadores Tumorais/antagonistas & inibidores , Proteínas de Transporte/antagonistas & inibidores , Glicoproteínas/antagonistas & inibidores , Neoplasias/tratamento farmacológico , Animais , Anticorpos Monoclonais Humanizados/administração & dosagem , Antígenos de Neoplasias , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Bevacizumab , Linhagem Celular Tumoral , Sinergismo Farmacológico , Feminino , Células Endoteliais da Veia Umbilical Humana , Humanos , Neoplasias Mamárias Experimentais , Camundongos , Camundongos Nus , Neoplasias/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Med Hypotheses ; 80(1): 75-82, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23177570

RESUMO

Metastasis is the leading cause of cancer death, yet it is mechanistically considered a very inefficient process suggesting the presence of some sort of (e.g. systemic) routes for fuelling the process. The pre-metastatic niche formation is described as one such metastasis promoting route. Now, the emerging potentials of tumor-derived microvesicles (TDMVs), not only in formulating the pre-metastatic niche, but also conferring neoplastic phenotypes onto normal cells, has integrated new concepts into the field. Here, we note as an ancillary proposition that, exerting functional disturbances in other sites, TDMVs (we have termed them metastasomes) may aid foundation of the secondary lesions via two seemingly interrelated models: (i) tumor-organ-training (TOTr), training a proper niche for the growth of the disseminated tumor cells; (ii) tumor-organ-targeting (TOTa), contribution to the propagation of the transformed phenotype via direct or indirect (TOTr-mediated disturbed stroma) transformation and/or heightened growth/survival states of the normal resident cells in the secondary organs. Respecting the high content of the RNA molecules (particularly microRNAs) identified in the secretory MVs, they may play crucial parts in such "malignant trait" spreading system. That is, the interactions between tumor tissue-specific RNA signatures, being transferred via metastasomes, and the cell-type/tissue-specific RNA stockrooms in other areas may settle a unique outcome in each organ. Thus, serving as tumor-organ matchmakers, the RNA molecules may also play substantial roles in the seeding and tropism of the process.


Assuntos
Transformação Celular Neoplásica , Modelos Biológicos , Metástase Neoplásica/fisiopatologia , Vesículas Transportadoras/fisiologia , Microambiente Tumoral/fisiologia , Humanos , MicroRNAs/metabolismo
8.
J Mol Med (Berl) ; 91(1): 83-94, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22864925

RESUMO

Elevated serum or tissue levels of lectin galactoside-binding soluble 3 binding protein (LGALS3BP) have been associated with short survival and development of metastasis in a variety of human cancers. However, the role of LGALS3BP, particularly in the context of tumor-host relationships, is still missing. Here, we show that LGALS3BP knockdown in MDA-MB-231 human breast cancer cells leads to a decreased adhesion to fibronectin, a reduced transendothelial migration and, more importantly, a reduced expression of vascular endothelial growth factor (VEGF). Production of VEGF, that was restored by exposure of silenced cells to recombinant LGALS3BP, required an intact PI3k/Akt signaling. Furthermore, we show that LGALS3BP was able to directly stimulate HUVEC tubulogenesis in a VEGF-independent, galectin-3-dependent manner. Immunohistochemical analysis of human breast cancer tissues revealed a correlation among LGALS3BP expression, VEGF expression, and blood vessel density. We propose that in addition to its prometastatic role, LGALS3BP secreted by breast cancer cells functions critically as a pro-angiogenic factor through a dual mechanism, i.e by induction of tumor VEGF and stimulation of endothelial cell tubulogenesis.


Assuntos
Antígenos de Neoplasias/genética , Biomarcadores Tumorais/genética , Neoplasias da Mama/irrigação sanguínea , Carcinoma Ductal de Mama/irrigação sanguínea , Carcinoma Lobular/irrigação sanguínea , Proteínas de Transporte/genética , Regulação Neoplásica da Expressão Gênica , Glicoproteínas/genética , Fator A de Crescimento do Endotélio Vascular/genética , Antígenos de Neoplasias/metabolismo , Biomarcadores Tumorais/antagonistas & inibidores , Biomarcadores Tumorais/metabolismo , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Carcinoma Ductal de Mama/genética , Carcinoma Ductal de Mama/metabolismo , Carcinoma Ductal de Mama/patologia , Carcinoma Lobular/genética , Carcinoma Lobular/metabolismo , Carcinoma Lobular/patologia , Proteínas de Transporte/antagonistas & inibidores , Proteínas de Transporte/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Técnicas de Cocultura , Feminino , Galectina 3/metabolismo , Técnicas de Silenciamento de Genes , Glicoproteínas/antagonistas & inibidores , Glicoproteínas/metabolismo , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Neovascularização Patológica , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Cultura Primária de Células , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Interferente Pequeno/genética , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/metabolismo
9.
Transl Oncol ; 6(6): 676-84, 2013 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-24466370

RESUMO

ErbB-3 (HER-3) receptor is involved in tumor progression and resistance to therapy. Development of specific inhibitors impairing the activity of ErbB-3 is an attractive tool for cancer therapeutics. MP-RM-1, a murine monoclonal antibody targeting human ErbB-3, has shown anticancer activity in preclinical models. With the aim to provide novel candidates for clinical use, we have successfully generated a humanized version of MP-RM-1. The humanized antibody, named EV20, abrogates both ligand-dependent and ligand-independent receptor signaling of several tumor cell types, strongly promotes ErbB-3 down-regulation, and efficiently and rapidly internalizes into tumor cells. Furthermore, treatment with EV20 significantly inhibits growth of xenografts originating from prostatic, ovarian, and pancreatic cancers as well as melanoma in nude mice. In conclusion, we provide a novel candidate for ErbB-3-targeted cancer therapy.

10.
PLoS One ; 6(6): e20393, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21674030

RESUMO

BACKGROUND: ATP-binding cassette transporter abcc2 is involved in the cellular efflux of irinotecan. The drug is toxic for mouse ileum, where abcc2 is highly expressed. Here, we investigate whether circadian changes in local abcc2 expression participate in the circadian rhythm of irinotecan toxicity for ileum mucosa, and further assess whether genetic background or sex modify this relation. METHODOLOGY/PRINCIPAL FINDINGS: Ileum mucosa was obtained every 3-4 h for 24 h in male and female B6D2F(1) and B6CBAF(1) mice synchronized with light from Zeitgeber Time (ZT)0 to ZT12 alternating with 12 h of darkness. Irinotecan (50 mg/kg i.v. daily for 4 days) was administered at the sex- and strain-specific times corresponding to least (ZT11-15) or largest drug-induced body weight loss (ZT23-03-07). Abcc2 expression was determined with qRT-PCR for mRNA and with immunohistochemistry and confocal microscopy for protein. Histopathologic lesions were graded in ileum tissues obtained 2, 4 or 6 days after treatment. Two- to six-fold circadian changes were demonstrated for mRNA and protein mean expressions of abcc2 in mouse ileum (p<0.05). ZT12 corresponded to high mRNA and protein expressions, with circadian waveforms differing according to genetic background and sex. The proportion of mice spared from ileum lesions varied three-fold according to irinotecan timing, with best tolerability at ZT11-15 (p = 0.00003). Irinotecan was also best tolerated in males (p = 0.05) and in B6CBAF(1) (p = 0.0006). CONCLUSIONS/SIGNIFICANCE: Strain- and sex-dependent circadian patterns in abcc2 expressions displayed robust relations with the chronotolerance of ileum mucosa for irinotecan. This finding has strong potential implications for improving the intestinal tolerability of anticancer drugs through circadian delivery.


Assuntos
Camptotecina/análogos & derivados , Ritmo Circadiano , Regulação da Expressão Gênica , Íleo/efeitos dos fármacos , Íleo/metabolismo , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo , Caracteres Sexuais , Animais , Antineoplásicos/efeitos adversos , Camptotecina/efeitos adversos , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Íleo/fisiologia , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/fisiologia , Irinotecano , Masculino , Camundongos , Proteína 2 Associada à Farmacorresistência Múltipla , Proteínas Associadas à Resistência a Múltiplos Medicamentos/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Especificidade da Espécie
11.
Int J Cancer ; 126(1): 41-52, 2010 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-19544526

RESUMO

Starting from an experimental model that accounts for the 2 most important adverse processes to successful therapy of Ewing's sarcoma (EWS), chemoresistance and the presence of metastasis at the time of diagnosis, we defined a molecular signature of potential prognostic value. Functional annotation of differentially regulated genes revealed 3 major networks related to cell cycle, cell-to-cell interactions and cellular development. The prognostic impact of 8 genes, representative of these 3 networks, was validated in 56 EWS patients. High mRNA expression levels of HINT1, IFITM2, LGALS3BP, STOML2 and c-MYC were associated with reduced risk to death and lower risk to develop metastasis. At multivariate analysis, LGALS3BP, a matricellular protein with a role in tumor progression and metastasis, was the most important predictor of event-free survival and overall survival. The association between LGALS3BP and prognosis was confirmed at protein level, when expression of the molecule was determined in tumor tissues but not in serum, indicating a role for the protein at local tumor microenvironment. Engineered enhancement of LGALS3BP expression in EWS cells resulted in inhibition of anchorage independent cell growth and reduction of cell migration and metastasis. Silencing of LGALS3BP expression reverted cell behavior with respect to in vitro parameters, thus providing further functional validation of genetic data obtained in clinical samples. Thus, we propose LGALS3BP as a novel reliable indicator of prognosis, and we offer genetic signatures to the scientific communities for cross-validation and meta-analysis, which are indispensable tools for a rare tumor such as EWS.


Assuntos
Proteínas de Transporte/fisiologia , Glicoproteínas/fisiologia , Sarcoma de Ewing/patologia , Animais , Antígenos de Neoplasias , Biomarcadores Tumorais , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Adesão Celular , Ciclo Celular , Linhagem Celular Tumoral , Ensaio de Imunoadsorção Enzimática , Inativação Gênica , Glicoproteínas/genética , Glicoproteínas/metabolismo , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Nus , Metástase Neoplásica , Análise de Sequência com Séries de Oligonucleotídeos , Prognóstico , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sarcoma de Ewing/metabolismo , Transdução de Sinais
12.
Anticancer Res ; 29(1): 403-10, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19331179

RESUMO

BACKGROUND: Galectins have emerged as critical regulators of tumor progression and metastasis, by modulating different biological events including homotypic cell aggregation, apoptosis, migration, angiogenesis and immune escape. Therefore, galectin inhibitors might represent novel therapeutic agents for cancer. MATERIALS AND METHODS: A series of structural analogs of the disaccharide methyl beta-lactosaminide were screened as potential galectin inhibitors by examining their capability to block binding of galectin-1 and/or galectin-3 to LGalS3BP in solid-phase assays. To demonstrate any functional role in vitro, oligosaccharides were characterized by their ability to regulate tumor cell apoptosis and LGalS3BP-induced homotypic cell aggregation. RESULTS: Oligosaccharides differentially inhibited binding of each galectin to LGalS3BP. Compounds containing longer oligosaccharide chains were found to be potent inhibitors of both galectins under static conditions. Strikingly, the most active compound in inhibiting homotypic cell aggregation and tumor cell apoptosis was found to be allyl lactoside, which paradoxically exhibited a modest inhibitory capacity for blocking galectin-1 and -3 binding to LGalS3BP. CONCLUSION: Allyl lactoside represents a novel powerful inhibitor of tumor-associated homotypic cell aggregation and apoptosis. Further investigations are required to remodel selective and potent inhibitors capable of specifically modulating the activity of different members of the galectin family.


Assuntos
Galectina 1/antagonistas & inibidores , Galectina 3/antagonistas & inibidores , Melanoma/tratamento farmacológico , Oligossacarídeos/farmacologia , Antígenos de Neoplasias , Apoptose/efeitos dos fármacos , Biomarcadores Tumorais , Sequência de Carboidratos , Proteínas de Transporte/metabolismo , Agregação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Sinergismo Farmacológico , Galectina 1/metabolismo , Galectina 3/metabolismo , Glicoproteínas/metabolismo , Humanos , Melanoma/metabolismo , Melanoma/patologia , Dados de Sequência Molecular
13.
Methods Enzymol ; 434: 187-232, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17954249

RESUMO

Many and various experimental techniques have been developed to fully analyze the intracellular signaling pathways of membrane phosphoinositides and their water-soluble derivatives. This chapter concentrates mainly on the range of lipid-derived, water-soluble signaling molecules that can be produced in cells from these membrane phosphoinositides, for which we and others have proposed biological roles. These include lysophosphatidylinositol, produced via phospholipase A(1/2) activities on phosphatidylinositol; cyclic inositol phosphates, produced via phosphatidylinositol/lysophosphatidylinositol-specific phospholipase C activities; and glycerophosphoinositols, produced via lysophospholipase A(2/1) activities on their corresponding lysophosphoinositides. While the methodologies described in this chapter are aimed more specifically at the separation, identification, and quantification of monophosphorylated glyceropho sphoinositols and other similarly charged inositol-containing products of the membrane phosphoinositides in cell extracts, they can be equally well applied to the full range of lysophosphoinositides, glycerophosphoinositols, inositol phosphates, and further inositol-containing water-soluble products of the phosphoinositides (e.g., cyclic inositol phosphates, methylphosphoinositol phosphates).


Assuntos
Fosfatidilinositóis/metabolismo , Fosfolipases A/metabolismo , Cromatografia Líquida de Alta Pressão/métodos , Cromatografia em Camada Fina/métodos , Lisofosfolipídeos/isolamento & purificação , Lisofosfolipídeos/metabolismo , Fosfatidilinositóis/análise , Solubilidade
14.
Glycobiology ; 16(3): 210-20, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16282605

RESUMO

Galectins, a family of structurally related carbohydrate-binding proteins, contribute to different events associated with cancer biology, including apoptosis, homotypic cell aggregation, angiogenesis and tumor-immune escape. To interfere with galectin-carbohydrate interactions during tumor progression, a current challenge is the design of specific galectin inhibitors for therapeutic purposes. Here, we report the synthesis of three novel low molecular weight synthetic lactulose amines (SLA): (1) N-lactulose-octamethylenediamine (LDO), (2) N,N'-dilactulose-octamethylenediamine (D-LDO), and (3) N,N'-dilactulose-dodecamethylenediamine (D-LDD). These compounds showed a differential ability to inhibit binding of galectin-1 and/or galectin-3 to the highly glycosylated protein 90K in solid-phase assays. In addition, each compound demonstrated selective regulatory effects in different events linked to tumor progression including tumor-cell apoptosis, homotypic cell aggregation, and endothelial cell morphogenesis. Our results suggest that galectin inhibitors with subtle differences in their carbohydrate structures may be potentially used to specifically block different steps of tumor growth and metastasis.


Assuntos
Aminas/síntese química , Aminas/farmacologia , Antineoplásicos/síntese química , Apoptose/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Galectinas/antagonistas & inibidores , Lactulose/química , Aminas/sangue , Aminas/química , Antineoplásicos/química , Antineoplásicos/classificação , Antineoplásicos/farmacologia , Agregação Celular/efeitos dos fármacos , Forma Celular/efeitos dos fármacos , Células Cultivadas , Células Endoteliais/citologia , Galectinas/farmacologia , Glicosilação , Humanos , Estrutura Molecular
15.
Cancer Res ; 65(18): 8339-49, 2005 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-16166311

RESUMO

The purpose of this study was to investigate the antiangiogenic and in vivo properties of the recently identified phosphatidylinositol 3-kinase (PI3K)/Akt inhibitor Inositol(1,3,4,5,6) pentakisphosphate [Ins(1,3,4,5,6)P5]. Because activation of the PI3K/Akt pathway is a crucial step in some of the events leading to angiogenesis, the effect of Ins(1,3,4,5,6)P5 on basic fibroblast growth factor (FGF-2)-induced Akt phosphorylation, cell survival, motility, and tubulogenesis in vitro was tested in human umbilical vein endothelial cells (HUVEC). The effect of Ins(1,3,4,5,6)P5 on FGF-2-induced angiogenesis in vivo was evaluated using s.c. implanted Matrigel in mice. In addition, the effect of Ins(1,3,4,5,6)P5 on growth of ovarian carcinoma SKOV-3 xenograft was tested. Here, we show that FGF-2 induces Akt phosphorylation in HUVEC resulting in antiapoptotic effect in serum-deprived cells and increase in cellular motility. Ins(1,3,4,5,6)P5 blocks FGF-2-mediated Akt phosphorylation and inhibits both survival and migration in HUVEC. Moreover, Ins(1,3,4,5,6)P5 inhibits the FGF-2-mediated capillary tube formation of HUVEC plated on Matrigel and the FGF-2-induced angiogenic reaction in BALB/c mice. Finally, Ins(1,3,4,5,6)P5 blocks the s.c. growth of SKOV-3 xenografted in nude mice to the same extent than cisplatin and it completely inhibits Akt phosphorylation in vivo. These data definitively identify the Akt inhibitor Ins(1,3,4,5,6)P5 as a specific antiangiogenic and antitumor factor. Inappropriate activation of the PI3K/Akt pathway has been linked to the development of several diseases, including cancer, making this pathway an attractive target for therapeutic strategies. In this respect, Ins(1,3,4,5,6)P5, a water-soluble, natural compound with specific proapoptotic and antiangiogenic properties, might result in successful anticancer therapeutic strategies.


Assuntos
Fosfatos de Inositol/farmacologia , Inibidores de Fosfoinositídeo-3 Quinase , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Inibidores da Angiogênese/farmacologia , Animais , Antineoplásicos/farmacologia , Processos de Crescimento Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Colágeno , Combinação de Medicamentos , Interações Medicamentosas , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Feminino , Fator 2 de Crescimento de Fibroblastos/antagonistas & inibidores , Fator 2 de Crescimento de Fibroblastos/farmacologia , Humanos , Fosfatos de Inositol/metabolismo , Fosfatos de Inositol/farmacocinética , Laminina , Camundongos , Camundongos Endogâmicos BALB C , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação/efeitos dos fármacos , Proteoglicanas , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
16.
Oncogene ; 23(9): 1754-65, 2004 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-14755253

RESUMO

Phosphoinositide 3-kinase (PI 3-K) is implicated in a wide array of biological and pathophysiological responses, including tumorigenesis, invasion and metastasis, therefore specific inhibitors of the kinase may prove useful in cancer therapy. We propose that specific inositol polyphosphates have the potential to antagonize the activation of PI 3-K pathways by competing with the binding of PtdIns(3,4,5)P3 to pleckstrin homology (PH) domains. Here we show that Ins(1,3,4,5,6)P5 inhibits the serine phosphorylation and the kinase activity of Akt/PKB. As a consequence of this inhibition, Ins(1,3,4,5,6)P5 induces apoptosis in ovarian, lung and breast cancer cells. Overexpression of constitutively active Akt protects SKBR-3 cells from Ins(1,3,4,5,6)P5-induced apoptosis. Furthermore, Ins(1,3,4,5,6)P5 enhances the proapoptotic effect of cisplatin and etoposide in ovarian and lung cancer cells, respectively. These results support a role for Ins(1,3,4,5,6)P5 as a specific inhibitor of the PI 3-K/Akt signalling pathway, that may sensitize cancer cells to the action of commonly used anticancer drugs.


Assuntos
Apoptose/efeitos dos fármacos , Fosfatos de Inositol/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Serina-Treonina Quinases , Proteínas Proto-Oncogênicas/metabolismo , Transdução de Sinais/efeitos dos fármacos , Antineoplásicos/farmacologia , Cromonas/farmacologia , Cisplatino/farmacologia , Feminino , Fibronectinas/metabolismo , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Morfolinas/farmacologia , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt , Células Tumorais Cultivadas
17.
EMBO J ; 22(16): 4178-89, 2003 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-12912916

RESUMO

Phosphatidylinositol-3-phosphate (PtdIns-3-P) is considered as a lipid constitutively present on endosomes; it does not seem to have a dynamic role in signalling. In contrast, phosphatidylinositol-3,4,5-trisphosphate (PtdIns-3,4,5-P(3)) plays a crucial role in different signalling pathways including translocation of the glucose transporter protein GLUT4 to the plasma membrane upon insulin receptor activation. GLUT4 translocation requires activation of two distinct pathways involving phosphatidylinositol 3-kinase (PI 3-K) and the small GTP-binding protein TC10, respectively. The contribution of each pathway remains to be elucidated. Here we show that insulin specifically induces the formation of PtdIns-3-P in insulin- responsive cells. The insulin-mediated formation of PtdIns-3-P occurs through the activation of TC10 at the lipid rafts subdomain of the plasma membrane. Exogenous PtdIns-3-P induces the plasma membrane translocation of both overexpressed and endogenous GLUT4. These data indicate that PtdIns-3-P is specifically produced downstream from insulin-mediated activation of TC10 to promote the plasma membrane translocation of GLUT4. These results give a new insight into the intracellular role of PtdIns-3-P and shed light on some aspects of insulin signalling so far not completely understood.


Assuntos
Hipoglicemiantes/farmacologia , Insulina/farmacologia , Proteínas Musculares , Fosfatos de Fosfatidilinositol/biossíntese , Proteínas rho de Ligação ao GTP/metabolismo , Células 3T3 , Adipócitos/citologia , Adipócitos/efeitos dos fármacos , Adipócitos/metabolismo , Androstadienos/farmacologia , Animais , Linhagem Celular , Cromonas/farmacologia , Desoxiglucose/farmacocinética , Inibidores Enzimáticos/farmacologia , Transportador de Glucose Tipo 4 , Microdomínios da Membrana/metabolismo , Camundongos , Proteínas de Transporte de Monossacarídeos/metabolismo , Morfolinas/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Fosfatos de Fosfatidilinositol/antagonistas & inibidores , Fosfatos de Fosfatidilinositol/farmacologia , Fator de Crescimento Derivado de Plaquetas/farmacologia , Proteínas Recombinantes de Fusão/metabolismo , Sistemas do Segundo Mensageiro , Wortmanina
18.
Mol Biol Cell ; 14(2): 503-15, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12589050

RESUMO

Glycerophosphoinositol 4-phosphate (GroPIns-4P) is a biologically active, water-soluble phospholipase A metabolite derived from phosphatidylinositol 4-phosphate, whose cellular concentrations have been reported to increase in Ras-transformed cells. It is therefore important to understand its biological activities. Herein, we have examined whether GroPIns-4P can regulate the organization of the actin cytoskeleton, because this could be a Ras-related function involved in cell motility and metastatic invasion. We find that in serum-starved Swiss 3T3 cells, exogenously added GroPIns-4P rapidly and potently induces the formation of membrane ruffles, and, later, the formation of stress fibers. These actin structures can be regulated by the small GTPases Cdc42, Rac, and Rho. To analyze the mechanism of action of GroPIns-4P, we selectively inactivated each of these GTPases. GroPIns-4P requires active Rac and Rho, but not Cdc42, for ruffle and stress fiber formation, respectively. Moreover, GroPIns-4P induces a rapid translocation of the green fluorescent protein-tagged Rac into ruffles, and increases the fraction of GTP-bound Rac, in intact cells. The activation of Rac by GroPIns-4P was near maximal and long-lasting. Interestingly, this feature seems to be critical in the induction of actin ruffles by GroPIns-4P.


Assuntos
Actinas/metabolismo , Citoesqueleto/metabolismo , Fosfatos de Inositol/metabolismo , Fosfatidilinositóis/química , Células 3T3 , Animais , Linhagem Celular Transformada , Movimento Celular , Relação Dose-Resposta a Droga , GTP Fosfo-Hidrolases/química , Proteínas de Fluorescência Verde , Substâncias de Crescimento/metabolismo , Proteínas Luminescentes/metabolismo , Camundongos , Microscopia de Fluorescência , Invasividade Neoplásica , Metástase Neoplásica , Proteínas Recombinantes/metabolismo , Fatores de Tempo , Transfecção , Proteína cdc42 de Ligação ao GTP/metabolismo
19.
Oncogene ; 21(42): 6520-9, 2002 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-12226755

RESUMO

Activation of the enzyme phospholipase C (PLC) leads to the formation of second messengers inositol 1,4,5-trisphosphate and diacylglycerol. Tyrosine kinase receptors activate this reaction through PLCgamma isoenzymes. PLCgamma activity involves its activation with, and phosphorylation by, receptor tyrosine kinases. Recently, it has been shown that phosphoinositide 3-kinase (PI 3-K) may regulate PLCgamma activity through the interaction of the PI 3-K product phosphatidylinositol 3,4,5-trisphosphate (PtdIns-3,4,5-P(3)) and the PLCgamma pleckstrin homology (PH) domain. In an effort to understand the signalling pathway that involves PI 3-K regulation of PLCgamma, we found that EGF induces a PI 3-K-dependent translocation of PLCgamma1 at the leading edge of migrating cells in a wound healing assay. Similarly, the isolated PH, but not the Src-homology (SH) domains, N-SH2 or SH3, of PLCgamma1, translocates at the leading edge. Our experiments also showed that stable PH PLCgamma1 expression blocks epidermal growth factor (EGF)- and serum-induced cell motility and increases cell adhesion in MDA-MB-231 cells. This may suggest that influence of PI 3-K on PLCgamma1 could be relevant in cell migration, where PLCgamma1 seems to play a key role by modulating a series of events involved in actin polymerization.


Assuntos
Neoplasias da Mama/enzimologia , Movimento Celular/fisiologia , Fator de Crescimento Epidérmico/farmacologia , Isoenzimas/metabolismo , Células Tumorais Cultivadas/patologia , Fosfolipases Tipo C/metabolismo , Neoplasias da Mama/patologia , Adesão Celular/fisiologia , Regulação da Expressão Gênica , Proteínas de Fluorescência Verde , Humanos , Concentração de Íons de Hidrogênio , Proteínas Luminescentes/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Fosfolipase C gama , Fosforilação , Transporte Proteico , Transdução de Sinais , Células Tumorais Cultivadas/metabolismo , Domínios de Homologia de src/fisiologia
20.
Glycoconj J ; 19(7-9): 551-6, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-14758079

RESUMO

Galectins and their ligands have been implicated in cell transformation and cancer metastasis, and found to have prognostic value. Mac-2 BP, also known as 90K, is a highly glycosylated, secreted protein extensively studied in human cancer, which binds galectin-1, galectin-3 and galectin-7. High expression levels of 90K are associated with a shorter survival, the occurrence of metastasis or a reduced response to chemotherapy in patients with different types of malignancy. The mechanisms underlying the prognostic significance of 90K and galectins in cancer are far from being understood, although they may be related to the ability of these proteins to interact and, to some extent, modulate cell-cell and cell-matrix adhesion and apoptosis. The resulting scenario is even more complex, as data have been presented that all these proteins might be associated with either a positive or a negative outcome of the patients. It is hypothesised that different galectins and galectin ligands with overlapping or opposite functions, expressed in different tumors during the different steps of the metastatic cascade might play a crucial role in tumor progression.


Assuntos
Galectinas/metabolismo , Lipoproteínas/metabolismo , Metástase Neoplásica , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Neoplasias/patologia , Antígenos de Neoplasias , Biomarcadores Tumorais , Proteínas de Transporte , Progressão da Doença , Glicoproteínas , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...