Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
1.
Int J Cancer ; 133(1): 235-46, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23292912

RESUMO

In this work, we have analyzed the expression of different members of the ErbB family in human samples of testicular germ cell tumors (GCTs). We observed expression of ErbB1 or ErbB2 in different tumor subtypes, but we also found high expression of ErbB3 in all GCTs tested. This pattern of expression was maintained when primary tumors were orthotopically implanted in nude mice. We have chosen a choriocarcinoma model characterized by high levels of ErbB1, but also of ErbB2 and ErbB3, to assay the in vivo effect of ErbB inhibitors on tumoral growth. Our results showed a complete lack of effect (refractoriness) to the pure ErbB1 receptor inhibitors cetuximab and gefitinib. While these inhibitors blocked ErbB1 phosphorylation, ErbB2 phosphorylation was not affected, suggesting an ErbB1-independent activation of this receptor. To confirm the importance of ErbB2 activation, animals were treated with lapatinib, a dual ErbB1 and ErbB2 inhibitor. Lapatinib treatment caused a 50% inhibition in tumor growth, an effect correlated with a blockade of both ErbB1 and ErbB2 phosphorylation levels, and of downstream signaling pathways (Akt, ERKs and Stat3). ErbB2 activation could still occur due to the formation of ErbB2/ErbB3 heterodimers, and ErbB3 activation was completely inhibited by lapatinib. Finally, combined inhibition of ErbB1 (gefitinib) and ErbB3 activities (knockdown expression by shRNA) inhibited tumoral testicular cells proliferation in a similar way to lapatinib. Our results explain why lapatinib but not anti-ErbB1 agents might be effective for treatment of testicular GCT patients.


Assuntos
Antineoplásicos/farmacologia , Receptores ErbB/antagonistas & inibidores , Neoplasias Embrionárias de Células Germinativas/tratamento farmacológico , Neoplasias Embrionárias de Células Germinativas/metabolismo , Quinazolinas/farmacologia , Neoplasias Testiculares/tratamento farmacológico , Neoplasias Testiculares/metabolismo , Animais , Anticorpos Monoclonais Humanizados/farmacologia , Western Blotting , Carcinoma Embrionário/tratamento farmacológico , Carcinoma Embrionário/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Cetuximab , Coriocarcinoma/tratamento farmacológico , Coriocarcinoma/metabolismo , Tumor do Seio Endodérmico/tratamento farmacológico , Tumor do Seio Endodérmico/metabolismo , Receptores ErbB/metabolismo , Gefitinibe , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Imunoprecipitação , Lapatinib , Masculino , Camundongos , Camundongos Nus , Neoplasias Experimentais , Fosforilação/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Reação em Cadeia da Polimerase em Tempo Real , Receptor ErbB-2/antagonistas & inibidores , Receptor ErbB-3/antagonistas & inibidores , Teratocarcinoma/tratamento farmacológico , Teratocarcinoma/metabolismo , Transplante Heterólogo
2.
Int J Cancer ; 131(1): 244-52, 2012 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-21826647

RESUMO

Deregulation of ErbB/HER receptor tyrosine kinases has been linked to several types of cancer. The mechanism of activation of these receptors includes establishment of receptor dimers. Here, we have analyzed the action of different small molecule HER tyrosine kinase inhibitors (TKIs) on HER receptor dimerization. Breast cancer cell lines were treated with distinct TKIs and the formation of HER2-HER3 dimers was analyzed by coimmunoprecipitation and western blot or by Förster resonance energy transfer assays. Antibody-dependent cellular cytotoxicity was analyzed by measuring the release of lactate dehydrogenase and cell viability. Lapatinib and neratinib interfered with ligand-induced dimerization of HER receptors; while pelitinib, gefitinib, canertinib or erlotinib did not. Moreover, lapatinib and neratinib were able to disrupt previously formed receptor dimers. Structural analyses allowed the elucidation of the mechanism by which some TKIs prevent the formation of HER receptor dimers, while others do not. Experiments aimed at defining the functional importance of dimerization indicated that TKIs that impeded dimerization prevented down-regulation of HER2 receptors, and favored the action of trastuzumab. We postulate that TKIs that prevent dimerization and down-regulation of HER2 may augment the antitumoral action of trastuzumab, and this mechanism of action should be considered in the treatment of HER2 positive tumors which combine TKIs with antireceptor antibodies.


Assuntos
Receptores ErbB/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Multimerização Proteica/efeitos dos fármacos , Receptor ErbB-2/antagonistas & inibidores , Aminoquinolinas/farmacologia , Compostos de Anilina/farmacologia , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais Humanizados/farmacologia , Neoplasias da Mama , Linhagem Celular Tumoral , Proliferação de Células , Receptores ErbB/química , Receptores ErbB/metabolismo , Cloridrato de Erlotinib , Feminino , Gefitinibe , Humanos , Lapatinib , Morfolinas/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Quinazolinas/farmacologia , Quinolinas/farmacologia , Receptor ErbB-2/química , Receptor ErbB-2/imunologia , Receptor ErbB-2/metabolismo , Trastuzumab
3.
J Cell Biol ; 122(1): 95-101, 1993 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-8314849

RESUMO

The ectodomain of proTGF-alpha, a membrane-anchored growth factor, is converted into soluble TGF-alpha by a regulated cellular proteolytic system that recognizes proTGF-alpha via the C-terminal valine of its cytoplasmic tail. In order to define the biochemical components involved in proTGF-alpha cleavage, we have used cells permeabilized with streptolysin O (SLO) that have been extensively washed to remove cytosol. PMA, acting through a Ca(2+)-independent protein kinase C, activates cleavage as efficiently in permeabilized cells as it does in intact cells. ProTGF-alpha cleavage is also stimulated by GTP gamma S through a mechanism whose pharmacological properties suggest the involvement of a heterotrimeric G protein acting upstream of the PMA-sensitive Ca(2+)-independent protein kinase C. Activated proTGF-alpha cleavage is dependent on ATP hydrolysis, appears not to require vesicular traffic, and acts specifically on proTGF-alpha that has reached the cell surface. These results indicate that proTGF-alpha is cleaved from the cell surface by a regulated system whose signaling, recognition, and proteolytic components are retained in cells devoid of cytosol.


Assuntos
Compostos de Alumínio , Fluoretos , Proteínas de Ligação ao GTP/metabolismo , Precursores de Proteínas/metabolismo , Processamento de Proteína Pós-Traducional , Fator de Crescimento Transformador alfa/biossíntese , Fator de Crescimento Transformador alfa/metabolismo , Alumínio/farmacologia , Animais , Células CHO , Membrana Celular/metabolismo , Permeabilidade da Membrana Celular , Cricetinae , Flúor/farmacologia , Guanosina 5'-O-(3-Tiotrifosfato)/farmacologia , Guanosina Difosfato/análogos & derivados , Guanosina Difosfato/farmacologia , Cinética , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/metabolismo , Ratos , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/metabolismo , Acetato de Tetradecanoilforbol/farmacologia , Tionucleotídeos/farmacologia , Transfecção
4.
J Cell Biol ; 132(3): 427-36, 1996 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-8636219

RESUMO

The extracellular domain of several membrane-anchored proteins can be released as a soluble fragment by the action of a cell surface endoproteolytic system. This cleavage results in the generation of a soluble and a cell-bound fragment. In the case of proteins with signaling capability, such as tyrosine kinase receptors, the cleavage process may have an effect on the kinase activity of the cell-bound receptor fragment. By using several cell lines that express the TrkA neurotrophin receptor, we show that this receptor tyrosine kinase is cleaved by a proteolytic system that mimics the one that acts at the cell surface. TrkA cleavage is regulated by protein kinase C and several receptor agonists (including the TrkA ligand NGF), occurs at the ectodomain in a membrane-proximal region, and is independent of lysosomal function. TrkA cleavage results in the generation of a cell-associated fragment that is phosphorylated on tyrosine residues. Tyrosine phosphorylation of this fragment is not detected in TrkA mutants devoid of kinase activity, suggesting that phosphorylation requires an intact TrkA kinase domain, and is not due to activation of an intermediate intracellular tyrosine kinase. The increased phosphotyrosine content of the cell-bound fragment may thus reflect higher catalytic activity of the truncated fragment. We postulate that cleavage of receptor tyrosine kinases by this naturally occurring cellular mechanism may represent an additional mean for the regulation of receptor activity.


Assuntos
Fatores de Crescimento Neural/farmacologia , Proteínas Proto-Oncogênicas/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Receptores de Fator de Crescimento Neural/metabolismo , Animais , Western Blotting , Células CHO , Linhagem Celular , Membrana Celular/metabolismo , Cricetinae , Humanos , Cinética , Lisossomos/metabolismo , Mutagênese Sítio-Dirigida , Fosforilação , Fosfotirosina/análise , Mutação Puntual , Proteínas Proto-Oncogênicas/biossíntese , Proteínas Proto-Oncogênicas/isolamento & purificação , Proto-Oncogenes , Receptores Proteína Tirosina Quinases/biossíntese , Receptores Proteína Tirosina Quinases/isolamento & purificação , Receptor trkA , Receptores de Fator de Crescimento Neural/biossíntese , Receptores de Fator de Crescimento Neural/isolamento & purificação , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Acetato de Tetradecanoilforbol/farmacologia , Transfecção
5.
Ann Oncol ; 19(11): 1860-9, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18641009

RESUMO

BACKGROUND: Receptor tyrosine kinases play an important role in breast cancer. One of them, the type I insulin-like growth factor, has been linked to resistance to trastuzumab (Herceptin), an agent that targets human epidermal growth factor receptor 2. Here, we show that the insulin-like growth factor-I receptor (IGF-IR) antagonist NVP-AEW541 inhibits proliferation of breast cancer cells and synergizes with trastuzumab. PATIENTS AND METHODS: Patient samples and breast cancer cell lines were evaluated for IGF-IR expression or activation by western blotting. 1-(4,5-Dimethylthiazol-2-yl)-3,5-diphenylformazan (MTT) uptake assays and Annexin V staining were used for the analyses of cell proliferation/apoptosis. Biochemical and genomic studies were carried out to gain insights into the mechanism of action of NVP-AEW541. RESULTS: The IGF-IR was expressed above normal levels in a number of breast cancer samples. Activation of this receptor was inhibited by NVP-AEW541 that also decreased cell proliferation and increased apoptosis. NVP-AEW541 decreased the amount of pAkt and increased the level of p27. Combination studies with several drugs used in the breast cancer clinic showed that NVP-AEW541 synergistically increased the action of trastuzumab. CONCLUSIONS: Our results show the anti-breast cancer action of NVP-AEW541 and support the clinical development of anti-IGF-IR agents, especially in combination with trastuzumab.


Assuntos
Anticorpos Monoclonais/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/enzimologia , Pirimidinas/farmacologia , Pirróis/farmacologia , Receptor ErbB-2/biossíntese , Receptor IGF Tipo 1/antagonistas & inibidores , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais Humanizados , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Sinergismo Farmacológico , Humanos , Pirimidinas/administração & dosagem , Pirróis/administração & dosagem , Receptor IGF Tipo 1/biossíntese , Trastuzumab
6.
Leukemia ; 21(4): 797-804, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17315026

RESUMO

Multiple myeloma (MM) is a malignancy of terminally differentiated B lymphocytes, characterized by accumulation of a monotypic plasma cell population in the bone marrow, monoclonal immunoglobulin in serum and/or urine and osteolytic lesions. Despite recent advances in the treatment, MM remains an incurable disease. This calls for an effort to develop novel therapeutics in order to eradicate the disease. Here we have evaluated the potential antimyeloma action of Pemetrexed, an antifolate drug that has shown promising results in other neoplastic diseases. Pemetrexed had a potent antimyeloma effect on cell lines sensitive and resistant to conventional therapeutic agents, and was also efficient on fresh cells from patients and in a murine MM model. Furthermore, Pemetrexed abrogated the protective action on MM cell death of several growth factors produced by the bone marrow microenvironment. Mechanistic studies indicated that Pemetrexed provoked this action by a combined effect that included cell cycle blockade, probably by p21 upregulation, and induction of apoptosis through caspase-dependent and -independent mechanisms. These data, together with the fact that Pemetrexed is already licensed for the therapy of other neoplastic diseases, opens the possibility for the inclusion of Pemetrexed in the therapeutic armamentarium to battle MM.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Glutamatos/farmacologia , Guanina/análogos & derivados , Mieloma Múltiplo/prevenção & controle , Animais , Antineoplásicos/uso terapêutico , Divisão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Glutamatos/uso terapêutico , Guanina/farmacologia , Guanina/uso terapêutico , Humanos , Camundongos , Camundongos SCID , Mieloma Múltiplo/tratamento farmacológico , Pemetrexede , Transplante Heterólogo
7.
Oncogene ; 36(12): 1733-1744, 2017 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-27641332

RESUMO

Long-term survival remains low for most patients with glioblastoma (GBM), which reveals the need for markers of disease outcome and novel therapeutic targets. We describe that ODZ1 (also known as TENM1), a type II transmembrane protein involved in fetal brain development, plays a crucial role in the invasion of GBM cells. Differentiation of glioblastoma stem-like cells drives the nuclear translocation of an intracellular fragment of ODZ1 through proteolytic cleavage by signal peptide peptidase-like 2a. The intracellular fragment of ODZ1 promotes cytoskeletal remodelling of GBM cells and invasion of the surrounding environment both in vitro and in vivo. Absence of ODZ1 by gene deletion or downregulation of ODZ1 by small interfering RNAs drastically reduces the invasive capacity of GBM cells. This activity is mediated by an ODZ1-triggered transcriptional pathway, through the E-box binding Myc protein, that promotes the expression and activation of Ras homolog family member A (RhoA) and subsequent activation of Rho-associated, coiled-coil containing protein kinase (ROCK). Overexpression of ODZ1 in GBM cells reduced survival of xenografted mice. Consistently, analysis of 122 GBM tumour samples revealed that the number of ODZ1-positive cells inversely correlated with overall and progression-free survival. Our findings establish a novel marker of invading GBM cells and consequently a potential marker of disease progression and a therapeutic target in GBM.


Assuntos
Regulação Neoplásica da Expressão Gênica , Glioblastoma/genética , Glioblastoma/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , Tenascina/genética , Transcrição Gênica , Proteína rhoA de Ligação ao GTP/genética , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Técnicas de Inativação de Genes , Glioblastoma/mortalidade , Glioblastoma/patologia , Xenoenxertos , Humanos , Camundongos , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Proteínas do Tecido Nervoso/deficiência , Proteínas do Tecido Nervoso/metabolismo , Prognóstico , Transporte Proteico , Proteólise , Transdução de Sinais , Tenascina/deficiência , Tenascina/metabolismo , Regulação para Cima , Quinases Associadas a rho/metabolismo
8.
J Natl Cancer Inst ; 85(16): 1327-33, 1993 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-8340945

RESUMO

BACKGROUND: A variety of human tumors frequently express high levels of epidermal growth factor (EGF) receptor and its ligand, transforming growth factor alpha (TGF-alpha), which in some tumors is associated with poor prognosis. Monoclonal antibodies (MAbs) that block the binding of TGF-alpha or EGF to the receptor can inhibit proliferation of tumor cells that express the receptor. Studies suggest that these MAbs may enhance the antitumor effects of chemotherapy. PURPOSE: Our purpose was to study, in vitro and in vivo, the antitumor effects of doxorubicin in combination with anti-EGF receptor MAbs against tumor cells expressing high levels of EGF receptor. Our goal was to achieve maximum initial cytoreduction with high-dose doxorubicin in association with prolonged blockade of EGF receptor with MAbs. METHODS: Anti-EGF receptor MAbs 528 (isotype IgG2a) and 225 (isotype IgG1) were used in combination with doxorubicin against cells from human A431 squamous cell carcinoma and human MDA-468 breast adenocarcinoma. Both A431 and MDA-468 cells express high levels of EGF receptors and TGF-alpha. Cultured cells were treated with doxorubicin (range, 0-10 nM) in the presence or absence of MAb 528 or 225 (range, 0-30 nM). At 48 hours, doxorubicin-containing medium was removed, and treatment with antibody was continued for 5 days, when cell proliferation assays were performed. The activity of the agents and the combinations against well-established xenografts in BALB/c nude mice was also studied. In nude mice, doxorubicin was given at doses of 50-100 micrograms/20 g body weight on 2 successive days, and MAbs 528 and 225 were given at a dose range of 0-2 mg intraperitoneally twice a week. RESULTS: MAbs 528 and 225 both enhanced the antitumor effects of doxorubicin against A431 and MDA-468 tumor cells, producing additive growth suppression in cell cultures. MAb 528 increased the antitumor effects of doxorubicin by 32%-42%, and similar results were obtained with MAb 225. In BALB/c athymic mice, the treatment of well-established xenografts with either doxorubicin or anti-EGF receptor MAb alone temporarily inhibited growth, but the combination of both agents substantially enhanced antitumor activity over that of doxorubicin alone in A431 and MDA-468 cell xenografts. The combination treatment of mice bearing A431 xenografts resulted in tumor eradication of 40%-100% in the surviving mice in several independent experiments. The enhanced antitumor activity was dose dependent. CONCLUSIONS: Our results suggest that anti-EGF receptor MAbs substantially enhance the effects of doxorubicin against well-established xenografts of tumor cells expressing high levels of EGF receptors. IMPLICATIONS: Clinical trials with anti-EGF receptor MAbs are being conducted, and trials with anti-EGF receptor MAbs combined with doxorubicin are planned.


Assuntos
Adenocarcinoma/terapia , Anticorpos Monoclonais/uso terapêutico , Carcinoma de Células Escamosas/terapia , Doxorrubicina/uso terapêutico , Receptores ErbB/imunologia , Adenocarcinoma/tratamento farmacológico , Animais , Carcinoma de Células Escamosas/tratamento farmacológico , Terapia Combinada , Relação Dose-Resposta a Droga , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Transplante de Neoplasias , Células Tumorais Cultivadas
9.
Cancer Res ; 50(13): 3892-6, 1990 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-2162247

RESUMO

Different subtypes of voltage-operated calcium channels (VOCCs) are expressed in different tissues and can be distinguished by functional and pharmacological criteria. One type of high voltage-activated calcium channel, specifically recognized by the peptide neurotoxin omega-conotoxin (omega CTx), is expressed only in neurons. Seven different human small cell lung carcinoma (SCC) cell lines were also found to bind 125I-omega CTx. The binding was specific, saturable, and of high affinity. 125I-omega CTx binding was not antagonized by the calcium channel ligands verapamil, nitrendipine, and diltiazem. There was a correlation between the amount of toxin binding and the detection of depolarization-induced calcium fluxes studied with the fluorimetric probe Fura2. Fura2 experiments also demonstrated that, in addition to omega CTx-sensitive calcium channels, SCC cell lines also expressed omega CTx-insensitive calcium channels, which were antagonized by nitrendipine and verapamil. 125I-omega CTx-labeled VOCCs from SCC cells were, furthermore, precipitated by anti-VOCC autoantibodies obtained from patients affected by the Lambert-Eaton myasthenic syndrome, a neuromuscular disease often associated with SCC. The present findings further indicate the presence of neuronal molecules with important biological function on SCC plasma membrane and add new insights into the pathogenetic mechanism of autoimmune neurological paraneoplastic diseases, like Lambert-Eaton myasthenic syndrome.


Assuntos
Canais de Cálcio/metabolismo , Carcinoma de Células Pequenas/metabolismo , Conotoxinas , Neoplasias Pulmonares/metabolismo , Peptídeos Cíclicos/metabolismo , Sítios de Ligação , Ligação Competitiva , Canais de Cálcio/análise , Canais de Cálcio/efeitos dos fármacos , Carcinoma de Células Pequenas/análise , Humanos , Neoplasias Pulmonares/análise , Nitrendipino/farmacologia , Células Tumorais Cultivadas , Verapamil/farmacologia
10.
Oncogene ; 35(21): 2756-65, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26364598

RESUMO

Advances in the treatment of breast cancer have resulted in increased survival. However, in the metastatic setting, the disease remains incurable. Therefore, understanding of the mechanisms that promote dissemination of breast cancer cells may favor the development of novel therapeutic strategies to fight those tumors. Here, we show that the ErbB ligands, Neuregulins (NRGs), promote metastatic dissemination of breast cancer cells by switching on a kinase-metalloproteinase network. Clinicopathological analyses demonstrated that NRG expression in breast tumors associated to lymph node invasion and poor patient outcome. Preclinical in vivo analyses showed that NRG expression favored in situ tumor growth, local spreading and metastatic dissemination. Genomic, biochemical and functional studies identified matrix metalloproteinases, particularly stromelysin 2 and collagenase 3, as key mediators of the NRG-induced dissemination properties of breast cancer cells. Mechanistic analyses demonstrated that NRG augmented metalloproteinase expression through a route controlled by ERK1/2 kinases. ERK1/2 increased collagenase 3 expression by controlling the activity of an SBF1-related transcription factor. In conclusion, we describe a pathway linked to breast cancer dissemination. The clinical availability of agents that target some of the components of this signalling pathway suggests that patients with tumors fed by NRGs or other factors able to activate the ERK-Collagenase 3 route may benefit from agents that act on that signalling axis.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Metaloproteinase 13 da Matriz/metabolismo , Neurregulinas/metabolismo , Animais , Neoplasias da Mama/genética , Linhagem Celular Tumoral , Intervalo Livre de Doença , Feminino , Xenoenxertos , Humanos , Células MCF-7 , Camundongos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Transdução de Sinais
11.
Oncogene ; 4(11): 1299-305, 1989 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-2573029

RESUMO

Two clones of NIH3T3 fibroblasts, NEN37 and NEN7, overexpressing chimeric EGF/neu receptors (3 x 10(5) and 1 x 10(6) receptors/cell, respectively), were treated with EGF in order to identify the array of intracellular signals generated after activation of the neu proto-oncogene product. The results thus obtained were correlated with the effects of EGF on cell growth, investigated by both [3H]thymidine incorporation and long term (5 days) proliferation studies. In addition to the stimulation of the neu tyrosine kinase, previously reported by Lehvaslaiho et al. (EMBO J., 8, 159-166, 1989), EGF (10(-9)-10(-8) M) was found to induce marked increases of both [Ca2+]i and plasma membrane potential (investigated by the fura-2 and bis-oxonol techniques) which, in their initial phase, were only marginally dependent on the presence of Ca2+ in the incubation medium. These responses were inhibited, but only in part (40-50%) by phorbol ester activators of protein kinase C. Moreover, inositolphosphate analysis (by anion exchange chromatography) revealed hydrolysis of membrane polyphosphoinositides. All these effects of EGF were more prompt and much larger in NEN7 than NEN37 cells. The EGF concentration-dependence curves (measured by both [3H]thymidine incorporation and long-term proliferation assay) were quite different in the two cell clones. In the cells expressing the lower number of receptors measurable growth stimulation was observed at 10(-10), and maximal effect at 10(-9) M EGF. In NEN7 cells the curve was much more shallow, with measurable stimulation already at 10(-12) and maximal effect at 10(-8) M EGF. The maximal growth effect was approximately the same for the two cell clones. It is concluded that the intracellular signals identified here may play a limited role in the neu-induced cell proliferation, but are possibly involved in the acquisition of the tumoral phenotype typically expressed by the EGF-treated NEN7 cells.


Assuntos
Receptores ErbB/genética , Proteínas Proto-Oncogênicas/genética , Proto-Oncogenes , Animais , Cálcio/metabolismo , Divisão Celular/efeitos dos fármacos , Membrana Celular/efeitos dos fármacos , Membrana Celular/fisiologia , Células Cultivadas , Quimera , Células Clonais , Replicação do DNA/efeitos dos fármacos , Fator de Crescimento Epidérmico/farmacologia , Receptores ErbB/fisiologia , Genes , Fosfatos de Inositol/metabolismo , Cinética , Potenciais da Membrana/efeitos dos fármacos , Camundongos , Proteínas Tirosina Quinases/genética , Receptor ErbB-2 , Transdução de Sinais
12.
Trends Pharmacol Sci ; 10(10): 411-4, 1989 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-2694537

RESUMO

The EGF receptor, which is homologous to the v-erb-B oncogene product, has intrinsic tyrosine kinase activity, and mediates an increase in polyphosphoinositide turnover and [Ca2+]i. Recently, great progress has been made in understanding the mechanism of signal transduction at this receptor. Jacopo Meldolesi and colleagues discuss how this knowledge may lead to a better understanding of the control of cell proliferation.


Assuntos
Receptores ErbB/fisiologia , Transdução de Sinais , Animais , Membrana Celular/metabolismo , Membrana Celular/fisiologia , Receptores ErbB/efeitos dos fármacos , Receptores ErbB/metabolismo , Humanos
13.
Leukemia ; 29(3): 705-14, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25102946

RESUMO

The development of resistance to therapy is unavoidable in the history of multiple myeloma patients. Therefore, the study of its characteristics and mechanisms is critical in the search for novel therapeutic approaches to overcome it. This effort is hampered by the absence of appropriate preclinical models, especially those mimicking acquired resistance. Here we present an in vivo model of acquired resistance based on the continuous treatment of mice bearing subcutaneous MM1S plasmacytomas. Xenografts acquired resistance to two generations of immunomodulatory drugs (IMiDs; lenalidomide and pomalidomide) in combination with dexamethasone, that was reversible after a wash-out period. Furthermore, lenalidomide-dexamethasone (LD) or pomalidomide-dexamethasone (PD) did not display cross-resistance, which could be due to the differential requirements of the key target Cereblon and its substrates Aiolos and Ikaros observed in cells resistant to each combination. Differential gene expression profiles of LD and PD could also explain the absence of cross-resistance. Onset of resistance to both combinations was accompanied by upregulation of the mitogen-activated protein kinase/extracellular signal-regulated kinase (ERK) kinase (MEK)/ERK pathway and addition of selumetinib, a small-molecule MEK inhibitor, could resensitize resistant cells. Our results provide insights into the mechanisms of acquired resistance to LD and PD combinations and offer possible therapeutic approaches to addressing IMiD resistance in the clinic.


Assuntos
Antineoplásicos/farmacologia , Dexametasona/farmacologia , Regulação Neoplásica da Expressão Gênica , Plasmocitoma/tratamento farmacológico , Talidomida/análogos & derivados , Proteínas Adaptadoras de Transdução de Sinal , Animais , Apoptose/efeitos dos fármacos , Benzimidazóis/farmacologia , Linhagem Celular Tumoral , Modelos Animais de Doenças , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Quimioterapia Combinada , MAP Quinases Reguladas por Sinal Extracelular/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Fator de Transcrição Ikaros/genética , Fator de Transcrição Ikaros/metabolismo , Lenalidomida , Camundongos , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/genética , Mieloma Múltiplo/metabolismo , Mieloma Múltiplo/patologia , Transplante de Neoplasias , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Plasmocitoma/genética , Plasmocitoma/metabolismo , Plasmocitoma/patologia , Transdução de Sinais , Talidomida/farmacologia , Transativadores/genética , Transativadores/metabolismo
14.
Endocrinology ; 122(4): 1419-25, 1988 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-2894297

RESUMO

The effects of alpha 1-adrenergic agents on GH release and intracellular free Ca2+ concentration ([Ca2+]i) were investigated in purified rat somatotroph preparations. Phenylephrine (PHE) stimulated in vitro GH release; the maximal effect (2.5-fold stimulation) occurred at 1 microM PHE. The effect was completely blocked by the alpha-adrenergic antagonist phentolamine and partially counteracted by the beta-antagonist propranolol. Experiments with the fluorescent Ca2+ probe fura 2 show that PHE causes [Ca2+]i to rise from 178 +/- 31 nM (mean +/- SE; n = 25) to 370 +/- 55 nM (n = 9). This effect was complete within 20 sec and was maintained for at least 5-10 min. The rise was rapidly interrupted by administration of 1 microM phentolamine. The beta-receptor agonist isoproterenol caused a small [Ca2+]i rise due to action on alpha 1-adrenoreceptors. The PHE-induced [Ca2+]i rise showed two components: an initial peak due to Ca2+ mobilization from intracellular stores and a subsequent rise due to Ca2+ influx from the extracellular space. Somatostatin (SRIF) lowered both resting [Ca2+]i and Ca2+ influx stimulated by PHE. Pertussis toxin pretreatment did not modify PHE-induced [Ca2+]i changes, while it completely prevented the effect of SRIF on both resting and triggered [Ca2+]i, thus suggesting that a GTP-binding protein sensitive to the toxin is involved in the transduction of SRIF action. The increase in cAMP induced by cholera toxin pretreatment modified neither PHE nor SRIF action on [Ca2+]i. In conclusion, in rat somatotrophs Ca2+ mobilization and influx are stimulated by alpha 1-adrenergic agents, and this triggered [Ca2+]i rise results in a stimulation of GH release. In these cells SRIF is able to reduce both resting [Ca2+]i levels and [Ca2+]i increases induced by alpha 1-adrenergic activation.


Assuntos
Cálcio/metabolismo , Hormônio do Crescimento/metabolismo , Adeno-Hipófise/metabolismo , Receptores Adrenérgicos alfa/metabolismo , Adenosina Difosfato Ribose/metabolismo , Animais , Proteínas de Ligação ao GTP/metabolismo , Isoproterenol/farmacologia , Masculino , Toxina Pertussis , Fentolamina/farmacologia , Fenilefrina/farmacologia , Adeno-Hipófise/efeitos dos fármacos , Propranolol/farmacologia , Ratos , Ratos Endogâmicos , Somatostatina/farmacologia , Fatores de Virulência de Bordetella/farmacologia
15.
FEBS Lett ; 235(1-2): 178-82, 1988 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-2456948

RESUMO

Binding of omega-conotoxin, a peptide toxin specific for some subtypes of voltage-operated calcium channels (VOCCs), was investigated in IMR32 neuroblastoma and PC12 pheochromocytoma cell lines. In both cell types, binding was specific, saturable and of high affinity. Association was rapid and dissociation almost non-existent. Dihydropyridines and verapamil failed to affect toxin binding, while high concentrations of CaCl2 completely antagonized it. Depolarization with high K+ induced a [Ca2+]i rise (revealed by the fura-2 fluorimetric technique) that consisted of an initial (0.5-1 min) peak followed by a prolonged (several minutes) plateau phase. omega-Conotoxin blocked mainly the first phase, while the dihydropyridine Ca2+ channel blocker, nitrendipine, primarily affected the plateau. This result suggests that in the two cell lines investigated, omega-conotoxin acts mainly on a subgroup of VOCCs that is resistant to dihydropyridines.


Assuntos
Neoplasias das Glândulas Suprarrenais/metabolismo , Cálcio/metabolismo , Canais Iônicos/metabolismo , Venenos de Moluscos/metabolismo , Neuroblastoma/metabolismo , Feocromocitoma/metabolismo , ômega-Conotoxinas , Animais , Bloqueadores dos Canais de Cálcio , Cloreto de Cálcio/farmacologia , Di-Hidropiridinas/farmacologia , Humanos , Canais Iônicos/efeitos dos fármacos , Venenos de Moluscos/farmacologia , Nitrendipino/farmacologia , Potássio/farmacologia , Ratos , Células Tumorais Cultivadas
16.
FEBS Lett ; 513(2-3): 282-8, 2002 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-11904165

RESUMO

Transforming growth factor-beta (TGF-beta) plays a pivotal role in the extracellular matrix accumulation observed in chronic progressive tissue fibrosis, but the intracellular signaling mechanism by which TGF-beta stimulates this process remains poorly understood. We examined whether mitogen-activated protein kinase (MAPK) routes were involved in TGF-beta1-induced collagen expression in L(6)E(9) myoblasts. TGF-beta1 induced p38 and extracellular signal-regulated kinase (ERK) 1/2 phosphorylation whereas no effect on Jun N-terminal kinase phosphorylation was observed. Biochemical blockade of p38 but not of the ERK MAPK pathway abolished TGF-beta1-induced alpha(2)(I) collagen mRNA expression and accumulation. These data indicate that TGF-beta1-induced p38 activation is involved in TGF-beta1-stimulated collagen synthesis.


Assuntos
Colágeno Tipo I/biossíntese , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fator de Crescimento Transformador beta/farmacologia , Animais , Células Cultivadas , Colágeno Tipo I/genética , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , RNA Mensageiro/biossíntese , Ratos , Fator de Crescimento Transformador beta1 , Proteínas Quinases p38 Ativadas por Mitógeno
17.
J Neuroendocrinol ; 3(1): 51-6, 1991 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-19215446

RESUMO

Abstract The effect of thyrotrophin-releasing hormone (TRH) on intracellular free Ca(2+) concentration, [Ca(2+))i, was investigated with the fluorescent dye fura-2 in cell suspensions obtained from 13 human growth hormone-secreting adenomas and 6 adrenocorticotrophin-secreting adenomas. Preoperatively, 9 out of 13 acromegalic patients showed a positive growth hormone response to TRH administration while none of the 6 patients with Cushing's disease had a plasma adrenocorticotrophin increase after TRH injection. In all the growth hormone-secreting adenomas the addition of TRH (100 nM) caused a significant rise in [Ca(2+)]i (from a resting level of 133+/-40 (+/-SD) to a value of 284+/-119 nM at 100 nM TRH, n = 42; P<0.001). The transient induced by TRH was found to have a dual origin, one due to Ca(2+) mobilization from intracellular stores which was maintained in presence of EGTA (3mM) and verapamil (10 muM) and a plateau phase due to Ca(2+) influx from the extracellular media. Somatostatin (0.1 muM) lowered both resting [Ca(2+)]i and TRH-induced transients. The effect of gonadotrophin-releasing hormone on [Ca(2+)]i was evaluated on cell suspensions obtained from 6 growth hormone-secreting adenomas. Gonadotrophin-releasing hormone (100 nM) caused a marked rise in [Ca(2+)]i (from 179+/-25 to 283+/-15nM) on the cell suspension obtained from the only in vivo responsive adenoma while it was ineffective in the remaining 5. Although TRH was ineffective in modifying plasma adrenocorticotrophin levels in all patients with Cushing's disease, in 5 out of 6 tumors the addition of 100 nM TRH caused a significant rise in [Ca(2+)]i (from 102.5 +/- 36 to 163+/-66 nM, n = 22; P < 0.005). However, the effect of TRH on [Ca(2+)]i was significantly lower than that caused by arginine vasopressin, a physiological stimulator of adrenocorticotrophin release ([Ca(2+)]i values; 145+/-78 nM at 100 nM TRH versus 300+/-140 at 10 nM arginine vasopressin, n = 15; P<0.05). Moreover, the effect of arginine vasopressin on [Ca(2+)]i was detectable at concentrations as low as 0.1 nM while TRH was effective at concentrations higher than 1 nM. By contrast, gonadotrophin-releasing hormone was ineffective in increasing [Ca(2)]i in all the adrenocorticotrophin-secreting adenomas studied. Collectively, these data indicate that sensitivity to TRH is present in almost all the growth hormone- and adrenocorticotrophin-secreting adenomas independently of the responsiveness of the individual patients to the peptide.

18.
Oncogene ; 33(2): 148-56, 2014 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-23246963

RESUMO

Triple-negative breast cancer (TNBC) is an aggressive form of breast cancer. Despite response to chemotherapy, relapses are frequent and resistance to available treatments is often seen in the metastatic setting. Therefore, identification of new therapeutic targets is required. With this aim, we have profiled the activation status of 44 receptor tyrosine kinases (RTKs) and their major signaling pathways in patient-derived TNBC tumors. Frequent co-activation of several RTKs as well as the extracellular signal-regulated protein kinases 1 and 2 (Erk1/2) and mammalian target of rapamycin (mTOR) routes was found. Pharmacologic targeting of the activated kinases indicated that agents that attack the mTOR route are more potent and efficient antitumoral treatments than agents targeting RTKs. mTOR signals through two multiprotein complexes, mTORC1 and mTORC2. We used a genetic approach to explore the contribution of each of the two mTOR branches to the regulation of cell number of TNBC cells. RNA interference experiments indicated that mTORC1 predominated over mTORC2 in the control of TNBC cell proliferation. Moreover, RNA interference of mTOR had a superior antiproliferative action than separately acting on mTORC1 or mTORC2. To analyze the relevance of mTOR targeting in vivo, we used mice with TNBC. Treatment of these mice with BEZ235, a drug that targets mTOR, slowed tumor growth. Mechanistically, BEZ235 delayed cell cycle progression without affecting cell viability. Our results show that TNBCs are particularly sensitive to inhibition of the mTOR pathway, and indicate that mTOR targeting may be a more efficient anti-TNBC therapy than exclusively acting on the mTORC1 branch of the pathway. This is relevant as most mTOR inhibitors used in the clinic act on mTORC1. Collectively with the fact that BEZ235 synergized with drugs commonly used in the treatment of TNBC, our data support the clinical development of agents that act on mTOR as a therapy for this disease.


Assuntos
Inibidores de Proteínas Quinases/uso terapêutico , Receptores Proteína Tirosina Quinases/metabolismo , Serina-Treonina Quinases TOR/antagonistas & inibidores , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Animais , Pontos de Checagem do Ciclo Celular , Ativação Enzimática , Feminino , Humanos , Imidazóis/uso terapêutico , Alvo Mecanístico do Complexo 1 de Rapamicina , Alvo Mecanístico do Complexo 2 de Rapamicina , Camundongos , Camundongos Endogâmicos BALB C , Complexos Multiproteicos/fisiologia , Quinolinas/uso terapêutico , Serina-Treonina Quinases TOR/fisiologia , Neoplasias de Mama Triplo Negativas/enzimologia , Neoplasias de Mama Triplo Negativas/genética
19.
Oncogene ; 32(11): 1452-9, 2013 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-22641219

RESUMO

The transmembrane tyrosine kinase HER2 (ErbB2, neu) is a prototypical biomarker for breast cancers and a therapeutic target. Although anti-HER2 therapies are remarkably effective, HER2-positive tumors are heterogeneous and some subtypes do not respond or develop resistance to these therapies. Here we show that H2NTF, a novel N-terminal fragment of HER2, is expressed at variable levels in 60% of the breast cancer samples analyzed. Characterization of H2NTF shows that it is devoid of the tyrosine kinase domain but it readily interacts with full-length HER2 and other HER receptors. As a consequence, H2NTF acts as a dominant-negative, attenuating the signaling triggered by full-length HER receptors. Expression of H2NTF results in resistance to the treatment with low concentrations of trastuzumab in vitro. However, cells expressing H2NTF and non-expressing cells have similar sensitivity to trastuzumab in vivo, indicating that H2NTF/trastuzumab complexes trigger antibody-dependent cell-mediated cytotoxicity.


Assuntos
Neoplasias da Mama/genética , Carcinoma/genética , Receptor ErbB-2/química , Receptor ErbB-2/genética , Sequência de Aminoácidos , Animais , Neoplasias da Mama/epidemiologia , Carcinoma/epidemiologia , Feminino , Regulação Neoplásica da Expressão Gênica , Frequência do Gene , Genes Dominantes , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos NOD , Camundongos Nus , Camundongos SCID , Modelos Biológicos , Dados de Sequência Molecular , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Estrutura Terciária de Proteína/genética , Estrutura Terciária de Proteína/fisiologia , Receptor ErbB-2/metabolismo , Células Tumorais Cultivadas
20.
Leukemia ; 27(2): 430-40, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22763387

RESUMO

Proteasome inhibitors (PIs), namely bortezomib, have become a cornerstone therapy for multiple myeloma (MM), potently reducing tumor burden and inhibiting pathologic bone destruction. In clinical trials, carfilzomib, a next generation epoxyketone-based irreversible PI, has exhibited potent anti-myeloma efficacy and decreased side effects compared with bortezomib. Carfilzomib and its orally bioavailable analog oprozomib, effectively decreased MM cell viability following continual or transient treatment mimicking in vivo pharmacokinetics. Interactions between myeloma cells and the bone marrow (BM) microenvironment augment the number and activity of bone-resorbing osteoclasts (OCs) while inhibiting bone-forming osteoblasts (OBs), resulting in increased tumor growth and osteolytic lesions. At clinically relevant concentrations, carfilzomib and oprozomib directly inhibited OC formation and bone resorption in vitro, while enhancing osteogenic differentiation and matrix mineralization. Accordingly, carfilzomib and oprozomib increased trabecular bone volume, decreased bone resorption and enhanced bone formation in non-tumor bearing mice. Finally, in mouse models of disseminated MM, the epoxyketone-based PIs decreased murine 5TGM1 and human RPMI-8226 tumor burden and prevented bone loss. These data demonstrate that, in addition to anti-myeloma properties, carfilzomib and oprozomib effectively shift the bone microenvironment from a catabolic to an anabolic state and, similar to bortezomib, may decrease skeletal complications of MM.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Reabsorção Óssea/tratamento farmacológico , Mieloma Múltiplo/tratamento farmacológico , Osteogênese/efeitos dos fármacos , Inibidores de Proteassoma/uso terapêutico , Administração Oral , Animais , Western Blotting , Medula Óssea/efeitos dos fármacos , Medula Óssea/metabolismo , Medula Óssea/patologia , Reabsorção Óssea/etiologia , Ácidos Borônicos/administração & dosagem , Bortezomib , Calcificação Fisiológica/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Compostos de Epóxi/farmacologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos SCID , Mieloma Múltiplo/complicações , Oligopeptídeos/administração & dosagem , Osteoblastos/efeitos dos fármacos , Osteoblastos/patologia , Osteoclastos/efeitos dos fármacos , Osteoclastos/patologia , Pirazinas/administração & dosagem , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Estromais/efeitos dos fármacos , Células Estromais/metabolismo , Células Estromais/patologia , Células Tumorais Cultivadas , Microambiente Tumoral/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA