Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
3.
Oncogene ; 36(41): 5734-5744, 2017 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-28581517

RESUMO

With very similar 3D structures, the widely expressed ß-arrestin isoforms 1 and 2 play at times identical, distinct or even opposing roles in regulating various aspects of G protein-coupled receptors (GPCR) expression and signalling. Recent evidence recognizes the ß-arrestin system as a key regulator of not only GPCRs, but also receptor tyrosine kinases, including the highly cancer relevant insulin-like growth factor type 1 receptor (IGF-1R). Binding of ß-arrestin1 to IGF-1R leads to ligand-dependent degradation of the receptor and generates additional MAPK/ERK signalling, protecting cancer cells against anti-IGF-1R therapy. Because the interplay between ß-arrestin isoforms governs the biological effects for most GPCRs, as yet unexplored for the IGF-1R, we sought to investigate specifically the regulatory roles of the ß-arrestin2 isoform on expression and function of the IGF-1R. Results from controlled expression of either ß-arrestin isoform demonstrate that ß-arrestin2 acts in an opposite manner to ß-arrestin1 by promoting degradation of an unstimulated IGF-1R, but protecting the receptor against agonist-induced degradation. Although both isoforms co-immunoprecipitate with IGF-1R, the ligand-occupied receptor has greater affinity for ß-arrestin1; this association lasts longer, sustains MAPK/ERK signalling and mitigates p53 activation. Conversely, ß-arrestin2 has greater affinity for the ligand-unoccupied receptor; this interaction is transient, triggers receptor ubiquitination and degradation without signalling activation, and leads to a lack of responsiveness to IGF-1, cell cycle arrest and decreased viability of cancer cells. This study reveals contrasting abilities of IGF-1R to interact with each ß-arrestin isoform, depending on the presence of the ligand and demonstrates the antagonism between the two ß-arrestin isoforms in controlling IGF-1R expression and function, which could be developed into a practical anti-IGF-1R strategy for cancer therapy.


Assuntos
Neoplasias/genética , Receptor IGF Tipo 1/genética , beta-Arrestinas/genética , Animais , Regulação Neoplásica da Expressão Gênica , Células HEK293 , Humanos , Camundongos , Neoplasias/patologia , Fosforilação , Ligação Proteica , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteólise , Receptor IGF Tipo 1/metabolismo , Transdução de Sinais/genética , Proteína Supressora de Tumor p53/genética , beta-Arrestinas/metabolismo
4.
Oncogene ; 36(23): 3274-3286, 2017 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-28092675

RESUMO

Melanoma tumors usually retain wild-type p53; however, its tumor-suppressor activity is functionally disabled, most commonly through an inactivating interaction with mouse double-minute 2 homolog (Mdm2), indicating p53 release from this complex as a potential therapeutic approach. P53 and the tumor-promoter insulin-like growth factor type 1 receptor (IGF-1R) compete as substrates for the E3 ubiquitin ligase Mdm2, making their relative abundance intricately linked. Hence we investigated the effects of pharmacological Mdm2 release from the Mdm2/p53 complex on the expression and function of the IGF-1R. Nutlin-3 treatment increased IGF-1R/Mdm2 association with enhanced IGF-1R ubiquitination and a dual functional outcome: receptor downregulation and selective downstream signaling activation confined to the mitogen-activated protein kinase/extracellular signal-regulated kinase pathway. This Nutlin-3 functional selectivity translated into IGF-1-mediated bioactivities with biphasic effects on the proliferative and metastatic phenotype: an early increase and late decrease in the number of proliferative and migratory cells, while the invasiveness was completely inhibited following Nutlin-3 treatment through an impaired IGF-1-mediated matrix metalloproteinases type 2 activation mechanism. Taken together, these experiments reveal the biased agonistic properties of Nutlin-3 for the mitogen-activated protein kinase pathway, mediated by Mdm2 through IGF-1R ubiquitination and provide fundamental insights into destabilizing p53/Mdm2/IGF-1R circuitry that could be developed for therapeutic gain.


Assuntos
Biomarcadores Tumorais/metabolismo , Fator de Crescimento Insulin-Like I/metabolismo , Melanoma/patologia , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Receptor IGF Tipo 1/metabolismo , Neoplasias Cutâneas/patologia , Proteína Supressora de Tumor p53/metabolismo , Apoptose , Biomarcadores Tumorais/genética , Ciclo Celular , Proliferação de Células , Humanos , Melanoma/genética , Melanoma/metabolismo , Invasividade Neoplásica , Fenótipo , Proteínas Proto-Oncogênicas c-mdm2/genética , Receptor IGF Tipo 1/genética , Transdução de Sinais , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/metabolismo , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/genética
5.
Prog Mol Biol Transl Sci ; 141: 277-311, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27378760

RESUMO

Cell-surface receptors govern the critical information passage from outside to inside the cell and hence control important cellular decisions such as survival, growth, and differentiation. These receptors, structurally grouped into different families, utilize common intracellular signaling-proteins and pathways, yet promote divergent biological consequences. In rapid processing of extracellular signals to biological outcomes, posttranslational modifications offer a repertoire of protein processing options. Protein ubiquitination was originally identified as a signal for protein degradation through the proteasome system. It is now becoming increasingly recognized that both ubiquitin and ubiquitin-like proteins, all evolved from a common ubiquitin structural superfold, are used extensively by the cell and encompass signal tags for many different cellular fates. In this chapter we examine the current understanding of the ubiquitin regulation surrounding the insulin-like growth factor and insulin signaling systems, major members of the larger family of receptor tyrosine kinases (RTKs) and key regulators of fundamental physiological and pathological states.


Assuntos
Receptor IGF Tipo 1/metabolismo , Transdução de Sinais , Ubiquitinação , Animais , Humanos , Modelos Biológicos , Fosforilação , Receptor de Insulina/metabolismo
6.
Oncogene ; 31(3): 352-65, 2012 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-21685939

RESUMO

The human cathelicidin antimicrobial protein-18 and its C terminal peptide, LL-37, displays broad antimicrobial activity that is mediated through direct contact with the microbial cell membrane. In addition, recent studies reveal that LL-37 is involved in diverse biological processes such as immunomodulation, apoptosis, angiogenesis and wound healing. An intriguing role for LL-37 in carcinogenesis is also beginning to emerge and the aim of this paper was to explore if and how LL-37 contributes to the signaling involved in tumor development. To this end, we investigated the putative interaction between LL-37 and growth factor receptors known to be involved in tumor growth and progression. Among several receptors tested, LL-37 bound with the highest affinity to insulin-like growth factor 1 receptor (IGF-1R), a receptor that is strongly linked to malignant cellular transformation. Furthermore, this interaction resulted in a dose-dependent phosphorylation and ubiquitination of IGF-1R, with downstream signaling confined to the mitogen-activated protein kinase/extracellular signal-regulated kinase (ERK)-pathway but not affecting phosphatidylinositol 3 kinase/Akt signaling. We found that signaling induced by LL-37 was dependent on the recruitment of ß-arrestin to the fully functional IGF-1R and by using mutant receptors we demonstrated that LL-37 signaling is dependent on ß-arrestin-1 binding to the C-terminus of IGF-1R. When analyzing the biological consequences of increased ERK activation induced by LL-37, we found that it resulted in enhanced migration and invasion of malignant cells in an IGF-1R/ß-arrestin manner, but did not affect cell proliferation. These results indicate that LL-37 may act as a partial agonist for IGF-1R, with subsequent intra-cellular signaling activation driven by the binding of ß-arrestin-1 to the IGF-1R. Functional experiments show that LL-37-dependent activation of the IGF-1R signaling resulted in increased migratory and invasive potential of malignant cells.


Assuntos
Peptídeos Catiônicos Antimicrobianos/metabolismo , Transformação Celular Neoplásica/metabolismo , Receptor IGF Tipo 1/agonistas , Peptídeos Catiônicos Antimicrobianos/química , Arrestinas/metabolismo , Linhagem Celular , Movimento Celular , Proliferação de Células , Humanos , Sistema de Sinalização das MAP Quinases , Invasividade Neoplásica/fisiopatologia , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor IGF Tipo 1/metabolismo , Ubiquitinação , beta-Arrestina 1 , beta-Arrestinas , Catelicidinas
7.
Oncogene ; 27(11): 1629-38, 2008 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-17828296

RESUMO

The insulin-like growth factor 1 receptor (IGF-1R) is crucial for growth and survival of malignant cells. Experience in targeting IGF-1R in cancer models has shown that strategies promoting downregulation of the receptor are much more efficient in inducing apoptosis than those inhibiting the IGF-1R activity. Recently, we found that the cyclolignan picropodophyllin (PPP) inhibits phosphorylation of IGF-1R and activation of downstream signaling without interfering with the highly homologous insulin receptor (IR). Furthermore, PPP treatment caused strong regression of tumor grafts and prolonged survival of animals with systemic tumor disease. Here we demonstrate that PPP also downregulates the IGF-1R, whereas the IR and several other receptors were not affected. PPP-induced IGF-1R downregulation required expression of the MDM2 E3 ligase, which recently was found to ubiquitinate and cause degradation of the IGF-1R. In addition knockdown of beta-arrestin1, the adaptor molecule known to bridges MDM2 and IGF-1R, prevented downregulation of the receptor and significantly decreased PPP-induced cell death. All together these data suggest that PPP downregulates IGF-1R by interfering with the action of beta-arrestin1/MDM2 as well as the achieved receptor downregulation contributes to the apoptotic effect of PPP.


Assuntos
Arrestinas/metabolismo , Regulação para Baixo , Neoplasias/metabolismo , Podofilotoxina/análogos & derivados , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Receptor IGF Tipo 1/metabolismo , Animais , Aorta/metabolismo , Aorta/patologia , Apoptose , Arrestinas/genética , Western Blotting , Proliferação de Células , Células Cultivadas , Citometria de Fluxo , Humanos , Imunoprecipitação , Fator de Crescimento Insulin-Like I/metabolismo , Camundongos , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Podofilotoxina/farmacologia , Proteínas Proto-Oncogênicas c-mdm2/genética , Receptor de Insulina/metabolismo , Suínos , Ubiquitina/metabolismo , beta-Arrestinas
8.
Br J Cancer ; 96 Suppl: R2-6, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17393577

RESUMO

The insulin-like growth factor (IGF-1) signalling is highly implicated in cancer. In this signalling the IGF-1 receptor (IGF-1R) is unquestionable, the predominating single factor. IGF-1R is crucial for tumour transformation and survival of malignant cell, but is only partially involved in normal cell growth. This is in part due to the interactions with oncogenes. Recent findings suggest a close interplay with the p53/MDM2 pathway. Disturbances in components in the p53/MDM2/IGF-1R network may cause IGF-1R upregulation and growth advantage for the cancer cell. Targeting of IGF-1R is more and more seen as a promising option for future cancer therapy. Single chain antibodies and small molecules with selective effects on IGF-1R dependent malignant growth are of particular interest. Forthcoming clinical trials are welcome and will indeed be the only way to evaluate the impact of IGF-1R targeting in human cancer.

9.
Oncogene ; 25(22): 3186-95, 2006 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-16407828

RESUMO

The cyclolignan PPP was recently demonstrated to inhibit the activity of insulin-like growth factor-1 receptor (IGF-1R), without affecting the highly homologous insulin receptor. In addition, PPP caused complete regression of xenografts derived from various types of cancer. These data highlight the use of this compound in cancer treatment. However, a general concern with antitumor agents is development of resistance. In light of this problem, we aimed to investigate whether malignant cells may develop serious resistance to PPP. After trying to select 10 malignant cell lines, with documented IGF-1R expression and apoptotic responsiveness to PPP treatment (IC50s less than 0.1 microM), only two survived an 80-week selection but could only tolerate maximal PPP doses of 0.2 and 0.5 microM, respectively. Any further increase in the PPP dose resulted in massive cell death. These two cell lines were demonstrated not to acquire any essential alteration in responsiveness to PPP regarding IGF-1-induced IGF-1R phosphorylation. Neither did they exhibit any increase in expression of the multidrug resistance proteins MDR1 or MRP1. Consistently, they did not exhibit decreased sensitivity to conventional cytostatic drugs. Rather, the sensitivity was increased. During the first half of the selection period, both cell lines responded with a temporary and moderate increase in IGF-1R expression, which appeared to be because of an increased transcription of the IGF-1R gene. This increase in IGF-1R might be necessary to make cells competent for further selection but only up to a PPP concentration of 0.2 and 0.5 microM. In conclusion, malignant cells develop no or remarkably weak resistance to the IGF-1R inhibitor PPP.


Assuntos
Resistencia a Medicamentos Antineoplásicos , Neoplasias/tratamento farmacológico , Podofilotoxina/análogos & derivados , Receptor IGF Tipo 1/antagonistas & inibidores , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Apoptose/efeitos dos fármacos , Relação Dose-Resposta a Droga , Humanos , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo , Neoplasias/metabolismo , Fosforilação , Podofilotoxina/uso terapêutico , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptor IGF Tipo 1/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Tumorais Cultivadas
10.
Br J Cancer ; 92(12): 2097-101, 2005 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-15956962

RESUMO

The insulin-like growth factor (IGF-1) signalling is highly implicated in cancer. In this signalling the IGF-1 receptor (IGF-1R) is unquestionable, the predominating single factor. IGF-1R is crucial for tumour transformation and survival of malignant cell, but is only partially involved in normal cell growth. This is in part due to the interactions with oncogenes. Recent findings suggest a close interplay with the p53/MDM2 pathway. Disturbances in components in the p53/MDM2/IGF-1R network may cause IGF-1R upregulation and growth advantage for the cancer cell. Targeting of IGF-1R is more and more seen as a promising option for future cancer therapy. Single chain antibodies and small molecules with selective effects on IGF-1R dependent malignant growth are of particular interest. Forthcoming clinical trials are welcome and will indeed be the only way to evaluate the impact of IGF-1R targeting in human cancer.


Assuntos
Neoplasias/fisiopatologia , Receptor IGF Tipo 1/fisiologia , Antineoplásicos/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Genes Supressores de Tumor/efeitos dos fármacos , Genes Supressores de Tumor/fisiologia , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/genética , Proto-Oncogenes/efeitos dos fármacos , Proto-Oncogenes/fisiologia , Receptor IGF Tipo 1/antagonistas & inibidores , Transdução de Sinais/fisiologia
11.
Melanoma Res ; 13(4): 325-37, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12883358

RESUMO

Adhesion between the CD44s receptor and hyaluronic acid plays an important role in cell migration, tumour growth and progression. Although the alternative splicing of CD44 variant exons represents the principal regulatory mechanism of CD44-mediated functions, CD44v spliced variants are scantily expressed in melanoma cells. For this reason, we have investigated the possibility that post-translational modifications of the CD44 standard receptor could play a pivotal role in regulating CD44-mediated functions in melanoma. Using metabolic inhibitors of N- and O-glycosylation, as well as melanoma transfectants expressing CD44s O-glycosylation site-specific mutants, we performed structural and functional analysis of N- and O-deglycosylated CD44s molecules expressed in melanoma cells. We discovered that complete N- and O-glycosylation is not required by CD44s to be correctly expressed on the melanoma cell surface. Indeed, variably glycosylated and functionally different CD44s molecules were constitutively expressed in primary and metastatic lesions. Furthermore, we observed that changes in N- and O-glycosylation of CD44s could modulate its cleavage. In fact, spontaneous CD44s shedding was dependent on the presence of partial or complete O-glycosylation of four serine-glycine motifs localized in the membrane-proximal CD44 ectodomain. Mutation of these serine residues, as well as an extensive metabolic O-deglycosylation, strongly impaired spontaneous CD44 shedding. Furthermore, an O-glycosylation-independent mechanism of CD44 cleavage has been identified. This alternative mechanism of receptor cleavage is phorbol 12-myristate-13-acetate (PMA) inducible, mediated by metalloproteinase and requires the presence of N-linked sugar residues. Our findings demonstrate that the post-translational modification of CD44s represents the principal regulatory mechanism of CD44s-mediated functions in melanoma.


Assuntos
Receptores de Hialuronatos/metabolismo , Melanoma/metabolismo , Proteínas de Neoplasias/metabolismo , Processamento de Proteína Pós-Traducional/fisiologia , Neoplasias Cutâneas/metabolismo , Antimetabólitos/farmacologia , Regulação para Baixo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glicosilação/efeitos dos fármacos , Humanos , Receptores de Hialuronatos/efeitos dos fármacos , Receptores de Hialuronatos/genética , Ácido Hialurônico/metabolismo , Melanoma/genética , Metaloproteases/efeitos dos fármacos , Metaloproteases/metabolismo , Mutagênese Sítio-Dirigida , Proteínas de Neoplasias/efeitos dos fármacos , Proteínas de Neoplasias/genética , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/fisiologia , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Neoplasias Cutâneas/genética , Acetato de Tetradecanoilforbol/farmacologia , Células Tumorais Cultivadas
12.
Cancer Res ; 60(18): 5278-83, 2000 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-11016658

RESUMO

We investigated the functional impact of p53 on insulin-like growth factor I receptor (IGF-IR) expression in malignant cells. Using the BL-41tsp53-2 cell line, a transfectant carrying temperature-sensitive (ts) p53 and endogenous mutant p53 (codon 248), we demonstrated a drastic down-regulation of plasma membrane-bound IGF-IRs on induction of wild-type p53. However, a similar response was obtained by treatment of BL-41tsp53-2 cells expressing mutant ts p53 with a p53 antisense oligonucleotide. Thus, even if the negative effect of wild-type p53 predominates under a competitive condition, these data indicate that mutant p53 may be important for up-regulation of IGF-IR. To further elucidate this issue, three melanoma cell lines (BE, SK-MEL-5, and SK-MEL-28) that overexpressed p53 were investigated. The BE cell line has a "hot spot" mutation (codon 248) and expresses only codon 248-mutant p53. SK-MEL-28 has a point mutation at codon 145. SK-MEL-5 cells did not exhibit any p53 mutations, but the absence of p21Waf1 expression suggested functionally aberrant p53. Our data suggest that interaction with Mdm-2 may underlie p53 inactivation in these cells. Using p53 antisense oligonucleotides, we demonstrated a substantial down-regulation of cell surface expression of IGF-IR proteins in all melanoma cell lines after 24 h. This was paralleled by decreased tyrosine phosphorylation of IGF-IR and growth arrest, and, subsequently, massive cell death was observed (this was also seen in BL-41tsp53-2 cells with mutant conformation of ts p53). Taken together, our results suggest that up-regulation of IGF-IR as a result of expression of aberrant p53 may be important for the growth and survival of malignant cells.


Assuntos
Acetilcisteína/análogos & derivados , Receptor IGF Tipo 1/biossíntese , Proteína Supressora de Tumor p53/fisiologia , Acetilcisteína/farmacologia , Divisão Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Inibidores de Cisteína Proteinase/farmacologia , Humanos , Melanoma/metabolismo , Oligonucleotídeos Antissenso/genética , Oligonucleotídeos Antissenso/farmacologia , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/biossíntese , Proteína Supressora de Tumor p53/genética
13.
Oncogene ; 19(37): 4298-301, 2000 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-10980604

RESUMO

The EWS/FLI-1 fusion gene is characteristic of most cases of Ewing's sarcoma and has been shown to be crucial for tumor transformation and cell growth. In this study we demonstrate a drastic down-regulation of the EWS/FLI-1 protein, and a growth arrest, following serum depletion of Ewing's sarcoma cells. This indicates that growth factor circuits may be involved in regulation of the fusion gene product. Of four different growth factors tested, basic fibroblast growth factor (bFGF) was found to be of particular significance. In fact, upon treatment of serum-depleted cells with bFGF, expression of the EWS/FLI-1 protein and growth of the Ewing's sarcoma cells were restored. In addition, a bFGF-neutralizing antibody, which was confirmed to inhibit FGF receptor (FGFR) phosphorylation, caused down-regulation of EWS/FLI-1. Experiments using specific cell cycle blockers (thymidine and colcemide) suggest that EWS/FLI-1 is directly linked to bFGF stimulation, and not indirectly to cell proliferation. We also demonstrated expression of FGFRs in several tumor samples of Ewing's sarcoma. Taken together, our data suggest that expression of FGFR is a common feature of Ewing's sarcoma and, in particular, that the bFGF pathway may be important for the maintenance of a malignant phenotype of Ewing's sarcoma cells through up-regulating the EWS/FLI-1 protein. Oncogene (2000) 19, 4298 - 4301


Assuntos
Neoplasias Ósseas/patologia , Fator 2 de Crescimento de Fibroblastos/fisiologia , Regulação Neoplásica da Expressão Gênica/fisiologia , Proteínas de Neoplasias/fisiologia , Proteínas de Fusão Oncogênica/biossíntese , Sarcoma de Ewing/patologia , Fatores de Transcrição/biossíntese , Adenocarcinoma/patologia , Anticorpos Monoclonais/farmacologia , Neoplasias Ósseas/metabolismo , Ciclo Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Cromossomos Humanos Par 11/genética , Cromossomos Humanos Par 11/ultraestrutura , Cromossomos Humanos Par 22/genética , Cromossomos Humanos Par 22/ultraestrutura , Meios de Cultura Livres de Soro , Demecolcina/farmacologia , Sinergismo Farmacológico , Fator de Crescimento Epidérmico/farmacologia , Fator 2 de Crescimento de Fibroblastos/antagonistas & inibidores , Fator 2 de Crescimento de Fibroblastos/imunologia , Fibroblastos/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Fator de Crescimento Insulin-Like I/farmacologia , Masculino , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Proteínas de Fusão Oncogênica/genética , Fator de Crescimento Derivado de Plaquetas/farmacologia , Neoplasias da Próstata/patologia , Proteína Proto-Oncogênica c-fli-1 , Proteína EWS de Ligação a RNA , Receptores de Fatores de Crescimento de Fibroblastos/efeitos dos fármacos , Receptores de Fatores de Crescimento de Fibroblastos/fisiologia , Sarcoma de Ewing/metabolismo , Timidina/farmacologia , Fatores de Transcrição/genética , Translocação Genética , Células Tumorais Cultivadas/efeitos dos fármacos , Células Tumorais Cultivadas/metabolismo
14.
Mol Cell Endocrinol ; 165(1-2): 131-7, 2000 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-10940491

RESUMO

Recent data indicate that the estrogen receptor (ER) blocker tamoxifen (TAM) can induce cell death in malignant melanoma cells. However, as shown in the present study and several other studies melanoma cells usually do not express classical ERs. In the present study we investigated whether the cytotoxic effect of TAM on melanoma cells could depend on interference with the expression or function of the insulin-like growth factor-1 receptor (IGF-1R), a plasma membrane receptor important for cell survival in this tumor cell type. Several melanoma cell lines were included in the analysis. Administration of TAM at a concentration of 15 microm or more resulted in cell death of the melanoma cells within 48 h. TAM treatment was correlated to a slight to moderate inhibition of IGF-1 binding to IGF-1R. Since it has been reported that TAM can increase the release of IGF binding proteins (IGFBPs) we then investigated whether this mechanism could underly the decreased IGF-1 binding. However, we could demonstrate that the amount of released IGFBPs were unchanged or decreased in TAM-treated cells. Whereas TAM did not have any strong effect on IGF-1 binding and the expression of IGF-1R at the cell surface, it was was found to efficently block tyrosine phosphorylation of IGF-1R beta-subunit. Taken together, our data suggest that TAM-induced cytotoxicity of malignant melanoma cells can be due to inactivation of IGF-1R.


Assuntos
Moduladores de Receptor Estrogênico/farmacologia , Fator de Crescimento Insulin-Like I/metabolismo , Melanoma/tratamento farmacológico , Melanoma/patologia , Tamoxifeno/farmacologia , Antineoplásicos Hormonais/farmacologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Morte Celular/efeitos dos fármacos , Morte Celular/fisiologia , Feminino , Humanos , Proteínas de Ligação a Fator de Crescimento Semelhante a Insulina/metabolismo , Melanoma/metabolismo , Receptor IGF Tipo 1/metabolismo , Células Tumorais Cultivadas
15.
Anticancer Drug Des ; 15(1): 67-72, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10888037

RESUMO

The insulin-like growth factor-1 receptor (IGF-1R) has been shown to be of critical importance for tumor development and tumor cell survival of various types of malignancies. We have previously demonstrated that an adequate N-linked glycosylation of IGF-1R is required for its translocation to the cell surface in melanoma cells. This raises the possibility of using glycosylation inhibitors as therapeutic agents against IGF-1R-dependent malignancies. In this study we show that inhibition of N-linked glycosylation using tunicamycin or the 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitor lovastatin resulted in down-regulation of IGF-1R at the cell surface in Ewing's sarcoma cell lines (RD-ES and ES-1 cells). The down-regulation of plasma membrane-bound IGF-1R was correlated with a drastic decrease in IGF-1R autophosphorylation, suggesting biochemical inactivation of the receptor. Whereas RD-ES and ES-1 cells responded differently with regard to DNA synthesis, the decrease in IGF-1R expression was accompanied by a rapid and substantial decrease in survival of both cell lines. Our data suggest that relatively untoxic HMG-CoA reductase inhibitors (e.g. lovastatin) could have therapeutic significance in IGF-1R-dependent neoplasms like Ewing's sarcoma.


Assuntos
Antineoplásicos/farmacologia , Regulação para Baixo , Receptor IGF Tipo 1/antagonistas & inibidores , Sarcoma de Ewing/patologia , Antineoplásicos/uso terapêutico , Divisão Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Glicosilação , Humanos , Lovastatina/farmacologia , Receptor IGF Tipo 1/metabolismo , Sarcoma de Ewing/tratamento farmacológico , Sarcoma de Ewing/metabolismo , Células Tumorais Cultivadas , Tunicamicina/farmacologia
16.
Growth Factors ; 17(3): 193-202, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-10705577

RESUMO

Insulin-like growth factor-1 receptor (IGF-1R) has been shown to be important for melanoma cell growth and survival. In this study we first show, using immunohistochemistry, that progression from benign nevi to malignant melanoma is paralleled by an increased expression of IGF-1R and a down-regulation of the cyclin-dependent kinase inhibitor p27Kip1. Even though the expression of p27Kip1 was drastically reduced compared to benign tumors, detectable amounts of it could be assayed by Western blotting in cultured melanoma cells. To analyze whether there is a causative relationship between the IGF-1 pathway and p27Kip1 expression, melanoma cells were treated with alpha IR-3, an antibody blocking the IGF-1 binding to IGF-1R, or Tunicamycin, which inhibits the translocation of IGF-1R to the cell surface. From these studies we could conclude that the overall expression of p27Kip1 is independent of the IGF-1 pathway. In contrast, the association of p27Kip1 with the different cyclins was drastically affected. Both TM and alpha IR-3 decreased the binding of p27Kip1 to cyclin D1, whose expression was drastically reduced. On the other hand there was an increased binding of p27Kip1 to cyclin E and cyclin A. This redistribution of p27Kip1 may be a mechanism for growth arrest and induction of apoptosis following interruption of the IGF-1 pathway in melanoma cells.


Assuntos
Proteínas de Ciclo Celular , Ciclinas/metabolismo , Inibidores Enzimáticos/metabolismo , Regulação Neoplásica da Expressão Gênica , Melanoma/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Receptor IGF Tipo 1/metabolismo , Proteínas Supressoras de Tumor , Western Blotting , Inibidor de Quinase Dependente de Ciclina p27 , Humanos , Imuno-Histoquímica , Melanoma/patologia , Nevo/metabolismo , Nevo/patologia , Células Tumorais Cultivadas
17.
Exp Cell Res ; 246(1): 38-46, 1999 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-9882513

RESUMO

The Ewing's sarcoma cell line RD-ES, which carries the EWS/FLI-1 fusion gene, responded to the 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitor lovastatin with growth arrest. Replenishment of mevalonate (MVA) to the arrested cells restored cell growth. However, if tunicamycin (TM), which is an inhibitor of N-linked glycosylation, was present together with MVA the cells remained arrested, indicating that N-linked glycosylation is of importance for growth of Ewing's sarcoma cells. Inhibition of the biosynthesis of EWS/FLI-1 fusion protein by treatment with antisense oligonucleotides also led to growth arrest, suggesting that this protein is of importance for cell growth. We investigated whether MVA synthesis and N-linked glycosylation could be involved in regulation of the expression of the EWS/FLI-1 fusion protein, which in fact contains four potential sites for N-linked glycosylation. We found that inhibition of both HMG-CoA reductase and N-linked glycosylation drastically decreased the expression of the fusion protein, which mainly appears in the cell nuclei. There was no significant difference in the inhibitory effect on the fusion protein between the cytoplasm and the cell nuclei, indicating that the transport of the fusion protein to the cell nucleus is not affected. The fusion protein did not exhibit any gel electrophoretic mobility shift after treatment of the cells with lovastatin or TM, and it did not incorporate [3H]glucosamine. Therefore we can conclude that the fusion protein is not a glycoprotein. The decreased expression of the fusion protein following lovastatin or TM treatment was found to be due to a lowered stability of de novo-synthesized fusion protein. The down-regulation of the fusion protein was correlated to growth arrest. Furthermore, the kinetics between the expression of EWS/FLI-1 fusion protein and the initiation of DNA synthesis in MVA-stimulated cells were similar. Taken together, our data suggest that the regulatory role of N-linked glycosylation in the expression of the EWS/FLI-1 fusion protein is important for growth of Ewing's sarcoma cells. Possible mechanisms underlying TM-induced decrease in EWS/FLI-1 expression may involve the breaking of growth factor receptor pathways.


Assuntos
Regulação da Expressão Gênica/efeitos dos fármacos , Ácido Mevalônico/metabolismo , Proteínas de Fusão Oncogênica/genética , Sarcoma de Ewing/metabolismo , Fatores de Transcrição/genética , Sequência de Aminoácidos , Western Blotting , Divisão Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Citoplasma/metabolismo , DNA de Neoplasias/biossíntese , Regulação para Baixo/efeitos dos fármacos , Glucosamina/metabolismo , Glicosilação/efeitos dos fármacos , Humanos , Hidroximetilglutaril-CoA Redutases/metabolismo , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Lovastatina/farmacologia , Ácido Mevalônico/farmacologia , Dados de Sequência Molecular , Peso Molecular , Oligonucleotídeos Antissenso/genética , Proteínas de Fusão Oncogênica/biossíntese , Proteínas de Fusão Oncogênica/química , Proteínas de Fusão Oncogênica/metabolismo , Proteína Proto-Oncogênica c-fli-1 , Proteína EWS de Ligação a RNA , Sarcoma de Ewing/enzimologia , Sarcoma de Ewing/patologia , Fatores de Tempo , Fatores de Transcrição/biossíntese , Fatores de Transcrição/química , Fatores de Transcrição/metabolismo , Células Tumorais Cultivadas , Tunicamicina/farmacologia
18.
Rom J Morphol Embryol ; 44(1-4): 65-71, 1998.
Artigo em Inglês | MEDLINE | ID: mdl-15678845

RESUMO

We present a study made during 4 years (1992-1996), which pursued the underlining of the atypical intraductal hiperplasias (A.I.D.H.) lesions, met isolated or in the association with mamar carcinoma. Our study included a 188 number of the breast tumors, among: in the 23 cases we established the existence of the modification by the A.I.D.H, type at the fibrocystics disease associated or not with the other benign diseases of the breast (fibroadenosis, intraductal papiloma) and in the 63 cases there were the modification by the AIDH associated with in situ or invasiv carcinoma. Epithelial hyperplasia is frequently associated with the fibrocystic changes, being included in the category of fibrocystic or proliferating modifications. The synonymous terms used for the epithelial hyperplasia are the hiperplazia ductala, or the epitelioza, or the papilomatosis The last two are suggested by the proliferation possibility (papillary or linear) of the epithelial or the mio-epithelial cells. Regardless of the microscopic aspect of the lesion, that should be acknowledged and treated as it is, due to the increased risk of the development of a carcinoma later on, and also due to the ratio of association between the modification and the mammary carcinoma. The risk of occurrence of subsequent carcinoma is augmented in the presence of the epithelial atipii and also increases in the presence of a mammary carcinoma at the relatives of the first rank (1.3). In this context, the importance of the differential diagnosis between the simple intraductal hyperplasia and the atypical one, the difficulty of differentiation from intraductal carcinoma in some cases, and finally the association with an increased risk of subsequent occurrence of carcinoma, constitute into sufficient arguments to consider this topic separately.


Assuntos
Neoplasias da Mama/patologia , Adenocarcinoma/complicações , Adenocarcinoma/patologia , Neoplasias da Mama/complicações , Carcinoma Ductal/complicações , Carcinoma Ductal/patologia , Carcinoma Intraductal não Infiltrante/complicações , Carcinoma Intraductal não Infiltrante/patologia , Carcinoma Medular/complicações , Carcinoma Medular/patologia , Carcinoma Papilar/complicações , Carcinoma Papilar/patologia , Feminino , Fibroadenoma/complicações , Fibroadenoma/patologia , Humanos , Hiperplasia/complicações , Hiperplasia/patologia , Papiloma/complicações , Papiloma/patologia , Estudos Retrospectivos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...