Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Cancer Res ; 56(20): 4791-8, 1996 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-8841000

RESUMO

Expression and function of the Fas apoptotic pathway was investigated in normal and malignant human breast epithelial cells. Nontransformed mammary epithelial cell lines all expressed high levels of Fas mRNA and protein, but only one of seven breast cancer cell lines (T47D) expressed high levels of Fas. Apoptosis was induced in the nontransformed lines when they were incubated with the anti-Fas antibody. However, all of the breast cancer cell lines tested, except T47D, were resistant to Fas-mediated apoptosis. Four of five Fas-resistant breast cancer cell lines became sensitive to Fas-mediated apoptosis upon treatment with IFN-gamma. Fas mRNA increased slightly in both cell lines that became sensitive and in the cell line that remained resistant to Fas-mediated apoptosis upon IFN-gamma treatment. However, the cell surface expression of Fas showed little or no increase in any of the cell lines tested upon IFN-gamma treatment. In contrast to Fas expression, interleukin-1beta-converting enzyme (ICE) expression increased only in the cell lines that became Fas sensitive after IFN-gamma treatment. The importance of ICE and/or ICE-like proteases in Fas-mediated apoptosis in these cells was confirmed by inhibition of Fas-mediated apoptosis by a specific ICE inhibitor, YVAD-cmk. Fas sensitivity was reconstituted in the IFN-gamma-resistant cell line by transfection of ICE into that cell line. Together, these data suggest that down-regulation of Fas and its pathway may be a step in tumor progression and that modulation of Fas expression may provide an approach to inducing apoptosis in breast cancer cells.


Assuntos
Apoptose , Neoplasias da Mama/metabolismo , Mama/metabolismo , Cisteína Endopeptidases/biossíntese , Interferon gama/farmacologia , Receptor fas/fisiologia , Apoptose/efeitos dos fármacos , Northern Blotting , Western Blotting , Mama/patologia , Neoplasias da Mama/enzimologia , Neoplasias da Mama/patologia , Caspase 1 , Cisteína Endopeptidases/genética , Indução Enzimática/efeitos dos fármacos , Feminino , Humanos , Transfecção , Células Tumorais Cultivadas , Receptor fas/metabolismo
2.
Cancer Res ; 56(18): 4236-43, 1996 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-8797598

RESUMO

Sodium butyrate-induced differentiation of breast cancer cell lines was used to identify protein tyrosine phosphatases (PTPs) involved in differentiation and growth inhibition of breast cancer cells. Of 42 PTPs analyzed, 31 were expressed in the ZR75-1 breast cancer cell line. Expression of four PTPs (DEP-1, SAP, PTP gamma, and PAC) was regulated in ZR75-1 cells undergoing differentiation. Expression of two of these PTPs (DEP-1 and SAP) was also regulated in the SKBr-3 cell line undergoing differentiation. In view of its marked induction with differentiation in an estrogen receptor (ER)-positive and an ER-negative breast cancer cell line, DEP-1 was investigated for a role in growth inhibition or induction of differentiation in breast cancer cells. A DEP-1 cDNA construct under control of a constitutively active cytomegalovirus promoter was transfected into the ZR75-1, SKBR-3, and MCF-7 breast cancer cell lines, and resistant colonies were selected with G418. DEP-1 expression inhibited the development of resistant colonies by 3-5-fold in all three lines compared to transfection with vector alone. Three stable MCF-7 cell lines expressing DEP-1 under control of an inducible metallothionein promoter were then established. In these lines, induction of DEP-1 expression inhibited breast cancer cell growth by 5-10-fold. These data describe PTPs expressed and regulated in breast cancer cell lines during differentiation and identify one PTP, DEP-1, that inhibits the growth of breast cancer cells in vitro.


Assuntos
Neoplasias da Mama/enzimologia , Neoplasias da Mama/patologia , Proteínas Tirosina Fosfatases/biossíntese , Northern Blotting , Butiratos/farmacologia , Ácido Butírico , Diferenciação Celular , Divisão Celular , Linhagem Celular , Membrana Celular/enzimologia , Citomegalovirus/genética , Indução Enzimática , Feminino , Regulação Enzimológica da Expressão Gênica , Humanos , Cinética , Metalotioneína/genética , Reação em Cadeia da Polimerase , Regiões Promotoras Genéticas , Proteínas Tirosina Fosfatases Classe 3 Semelhantes a Receptores , Proteínas Recombinantes/biossíntese , Transcrição Gênica , Células Tumorais Cultivadas
3.
Cancer Res ; 59(3): 734-41, 1999 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-9973225

RESUMO

Expression and function of the TRAIL apoptotic pathway was investigated in normal and malignant breast epithelial cells. Glutathione-S-transferase (GST)-TRAIL extracellular domain fusion proteins were produced to analyze TRAIL-induced apoptosis. Only GST-TRAIL constructs containing regions homologous to the Fas self-association and ligand binding domains could induce apoptosis. GST-TRAIL induced significant (>90%) apoptosis in just one of eight normal and one of eight malignant breast cell lines. All other lines were relatively resistant to TRAIL-induced apoptosis. Activating TRAIL receptors DR4 and DR5 were expressed in all normal and malignant breast cell lines. The inhibitory receptor TRID was highly expressed in one of four normal and two of seven malignant breast cell lines. DR4, DR5, or TRID expression did not correlate with sensitivity to TRAIL-induced apoptosis. Incubation of cell lines with doxorubicin or 5-fluorouracil significantly augmented TRAIL-induced apoptosis in most breast cell lines. By fractional inhibition analysis, the toxicity of the combination of TRAIL and doxorubicin or 5-fluorouracil was synergistic compared with either agent alone. In contrast, melphalan and paclitaxel augmented TRAIL-induced apoptosis in few cell lines, and methotrexate did not augment it in any cell line. Augmentation of TRAIL-induced apoptosis by doxorubicin or 5-fluorouracil was mediated through caspase activation. This was evidenced by the fact that chemotherapy agents that synergized with TRAIL (e.g., doxorubicin) themselves caused cleavage of caspase-3 and poly(ADP-ribose) polymerase (PARP), and their toxicity was blocked by the caspase inhibitor Z-Val-Ala-Asp(OMe)-CH2 (ZVAD-fmk). The combination of TRAIL and doxorubicin caused significantly greater caspase-3 and PARP cleavage, and the combined toxicity also was inhibited by ZVAD-fmk. In contrast, chemotherapy agents that did not augment TRAIL-induced apoptosis (e.g., methotrexate) caused minimal caspase-3 and PARP cleavage by themselves, and their toxicity was not inhibited by ZVAD-fmk. These drugs also did not increase caspase-3 or PARP cleavage when combined with TRAIL. In summary, few breast cell lines are sensitive to TRAIL-induced apoptosis, and no difference in sensitivity is found between normal and malignant cell lines. Treatment with chemotherapy provides an approach to sensitize breast cancer cells to TRAIL-induced apoptosis.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Glicoproteínas de Membrana/farmacologia , Fator de Necrose Tumoral alfa/farmacologia , Antineoplásicos/administração & dosagem , Proteínas Reguladoras de Apoptose , Caspase 3 , Caspases/metabolismo , Doxorrubicina/administração & dosagem , Doxorrubicina/farmacologia , Ativação Enzimática , Fluoruracila/administração & dosagem , Fluoruracila/farmacologia , Glutationa Transferase/farmacologia , Glutationa Transferase/fisiologia , Humanos , Melfalan/administração & dosagem , Melfalan/farmacologia , Glicoproteínas de Membrana/administração & dosagem , Glicoproteínas de Membrana/fisiologia , Paclitaxel/administração & dosagem , Paclitaxel/farmacologia , Proteínas Recombinantes de Fusão/farmacologia , Ligante Indutor de Apoptose Relacionado a TNF , Células Tumorais Cultivadas , Fator de Necrose Tumoral alfa/administração & dosagem , Fator de Necrose Tumoral alfa/fisiologia
4.
Cancer Res ; 61(12): 4892-900, 2001 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-11406568

RESUMO

We investigated whether combined treatment with tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and trastuzumab could enhance the specific killing of cells that overexpress the erbB-2 receptor. The combination resulted in an enhancement of TRAIL-mediated apoptosis in all cell lines overexpressing erbB-2 receptor compared with either reagent alone. In contrast, there was no effect in cell lines with low levels of the erb-B2 receptor. Trastuzumab treatment resulted in down-regulation of the erbB-2 receptor in all erbB-2-overexpressing cell lines. Similar enhancement of TRAIL toxicity was observed when the erbB-2 receptor was down-regulated using antisense oligodeoxynucleotides. Down-regulation of the erbB-2 receptor protein by trastuzumab or antisense oligodeoxynucleotides decreased Akt kinase activation but not mitogen-activated protein kinase activation. Down-regulation of Akt kinase activity by a phosphatidylinositol 3'-kinase inhibitor (LY294002) also resulted in enhancement of TRAIL-mediated apoptosis. Expression of a constitutively active form of Akt kinase in an erbB-2-overexpressing cell line completely abrogated the increase in TRAIL-mediated apoptosis by trastuzumab and significantly reduced the biological effect of either reagent alone. Therefore, down-regulation of the erbB-2 receptor by trastuzumab enhances TRAIL-mediated apoptosis by inhibiting Akt kinase activity. These data suggest that the combination of trastuzumab and TRAIL may allow enhanced therapeutic efficacy and specificity in the treatment of erbB-2-overexpressing tumors.


Assuntos
Anticorpos Monoclonais/farmacologia , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias da Mama/patologia , Glicoproteínas de Membrana/farmacologia , Neoplasias Ovarianas/patologia , Proteínas Serina-Treonina Quinases , Receptor ErbB-2/biossíntese , Fator de Necrose Tumoral alfa/farmacologia , Anticorpos Monoclonais Humanizados , Apoptose/fisiologia , Proteínas Reguladoras de Apoptose , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Regulação para Baixo/efeitos dos fármacos , Sinergismo Farmacológico , Feminino , Humanos , Glicoproteínas de Membrana/genética , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/metabolismo , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Receptor ErbB-2/genética , Receptor ErbB-2/fisiologia , Proteínas Recombinantes de Fusão/farmacologia , Ligante Indutor de Apoptose Relacionado a TNF , Trastuzumab , Células Tumorais Cultivadas , Fator de Necrose Tumoral alfa/genética
5.
Oncogene ; 35(47): 6077-6086, 2016 11 24.
Artigo em Inglês | MEDLINE | ID: mdl-27157619

RESUMO

Notch receptors have been implicated as oncogenic drivers in several cancers, the most notable example being NOTCH1 in T-cell acute lymphoblastic leukemia (T-ALL). To characterize the role of activated NOTCH3 in cancer, we generated an antibody that detects the neo-epitope created upon gamma-secretase cleavage of NOTCH3 to release its intracellular domain (ICD3), and sequenced the negative regulatory region (NRR) and PEST (proline, glutamate, serine, threonine) domain coding regions of NOTCH3 in a panel of cell lines. We also characterize NOTCH3 tumor-associated mutations that result in activation of signaling and report new inhibitory antibodies. We determined the structural basis for receptor inhibition by obtaining the first co-crystal structure of a NOTCH3 antibody with the NRR protein and defined two distinct epitopes for NRR antibodies. The antibodies exhibit potent anti-leukemic activity in cell lines and tumor xenografts harboring NOTCH3 activating mutations. Screening of primary T-ALL samples reveals that 2 of 40 tumors examined show active NOTCH3 signaling. We also identified evidence of NOTCH3 activation in 12 of 24 patient-derived orthotopic xenograft models, 2 of which exhibit activation of NOTCH3 without activation of NOTCH1. Our studies provide additional insights into NOTCH3 activation and offer a path forward for identification of cancers that are likely to respond to therapy with NOTCH3 selective inhibitory antibodies.


Assuntos
Anticorpos Monoclonais/farmacologia , Antineoplásicos/farmacologia , Mutação , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Receptor Notch3/antagonistas & inibidores , Receptor Notch3/genética , Substituição de Aminoácidos , Animais , Linhagem Celular Tumoral , Códon , Modelos Animais de Doenças , Epitopos/química , Epitopos/imunologia , Feminino , Humanos , Camundongos , Modelos Moleculares , Leucemia-Linfoma Linfoblástico de Células T Precursoras/tratamento farmacológico , Leucemia-Linfoma Linfoblástico de Células T Precursoras/metabolismo , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patologia , Conformação Proteica , Receptor Notch3/química , Receptor Notch3/metabolismo , Transdução de Sinais/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Leukemia ; 30(2): 379-89, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26293646

RESUMO

We have previously demonstrated that interleukin-17A (IL-17) producing T helper 17 cells are significantly elevated in blood and bone marrow (BM) in multiple myeloma (MM) and IL-17A promotes MM cell growth via the expression of IL-17 receptor. In this study, we evaluated anti-human IL-17A human monoclonal antibody (mAb), AIN457 in MM. We observe significant inhibition of MM cell growth by AIN457 both in the presence and the absence of BM stromal cells (BMSCs). Although IL-17A induces IL-6 production, AIN457 significantly downregulated IL-6 production and MM cell adhesion in MM-BMSC co-culture. AIN457 also significantly inhibited osteoclast cell differentiation. More importantly, in the SCIDhu model of human myeloma administration of AIN457 weekly for 4 weeks after the first detection of tumor in mice led to a significant inhibition of tumor growth and reduced bone damage compared with isotype control mice. To understand the mechanism of action of anti-IL-17A mAb, we report, here, that MM cells express IL-17A. We also observed that IL-17A knockdown inhibited MM cell growth and their ability to induce IL-6 production in co-cultures with BMSC. These pre-clinical observations suggest efficacy of AIN457 in myeloma and provide the rationale for its clinical evaluation for anti-myeloma effects and for improvement of bone disease.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Interleucina-17/antagonistas & inibidores , Mieloma Múltiplo/tratamento farmacológico , Animais , Anticorpos Monoclonais Humanizados , Modelos Animais de Doenças , Humanos , Interleucina-6/biossíntese , Masculino , Camundongos , Osteoclastos/efeitos dos fármacos , Sindecana-1/análise
7.
Oncogene ; 18(22): 3365-75, 1999 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-10362357

RESUMO

We have cloned a new human gene, cbl-3, which encodes a protein with marked homology to the cbl family of proteins. The predicted protein encoded by this gene retains the conserved phosphotyrosine binding domain (PTB) in the N-terminal and the zinc finger but is significantly shorter (MW 52.5 kDa) than the other mammalian cbl proteins. The protein lacks the extensive proline rich domain and leucine zipper seen in c-cbl and cbl-b and structurally most resembles the C. elegans and Drosophila cbl proteins. The gene is ubiquitously expressed with highest expression in the aerodigestive tract, prostate, adrenal gland, and salivary gland. The protein is phosphorylated and recruited to the EGFR upon EGF stimulation and inhibits EGF stimulated MAP kinase activation. In comparison to the other mammalian cbl proteins (e.g. cbl-b), cbl-3 interacts with a restricted range of proteins containing Src Homology 3 regions. An alternatively spliced form of the cbl-3 protein was also identified which deletes a critical region of the PTB domain and which does not interact with the EGFR nor inhibit EGF stimulated MAP kinase activation. These data demonstrate that cbl-3, a novel mammalian cbl protein, is a regulator of EGFR mediated signal transduction.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Cromossomos Humanos Par 19 , Proteínas de Drosophila , Proteínas/genética , Proteínas/metabolismo , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Ubiquitina-Proteína Ligases , Sequência de Aminoácidos , Sequência de Bases , Sítios de Ligação , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Proteínas de Transporte/genética , Mapeamento Cromossômico , Clonagem Molecular , Ativação Enzimática , Receptores ErbB/metabolismo , Regulação da Expressão Gênica , Humanos , Dados de Sequência Molecular , Fosfoproteínas/genética , Fosforilação , Proteínas Proto-Oncogênicas c-cbl , Homologia de Sequência de Aminoácidos , Transdução de Sinais , Domínios de Homologia de src
8.
Oncogene ; 18(10): 1855-66, 1999 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-10086340

RESUMO

The role of cbl-b in signaling by the epidermal growth factor receptor (EGFR) was studied and compared with c-cbl. We demonstrate in vivo, that cbl-b, like c-cbl, is phosphorylated and recruited to the EGFR upon EGF stimulation and both cbl proteins can bind to the Grb2 adaptor protein. To investigate the functional role of cbl proteins in EGFR signaling, we transfected cbl-b or c-cbl into 32D cells overexpressing the EGFR (32D/EGFR). This cell line is absolutely dependent on exogenous IL-3 or EGF for sustained growth. 32D/EGFR cells overexpressing cbl-b showed markedly inhibited growth in EGF compared to c-cbl transfectants and vector controls. This growth inhibition by cbl-b was the result of a dramatic increase in the number of cells undergoing apoptosis. Consistent with this finding, cbl-b overexpression markedly decreased the amplitude and duration of AKT activation upon EGF stimulation compared to either vector controls or c-cbl overexpressing cells. In addition, the duration of EGF mediated MAP kinase and Jun kinase activation in cells overexpressing cbl-b is shortened. These data demonstrate that cbl-b inhibits EGF-induced cell growth and that cbl-b and c-cbl have distinct roles in EGF mediated signaling.


Assuntos
Receptores ErbB/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Transdução de Sinais , Ubiquitina-Proteína Ligases , Animais , Linhagem Celular Transformada , Fator de Crescimento Epidérmico/metabolismo , Fator de Crescimento Epidérmico/farmacologia , Humanos , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-cbl , Coelhos
9.
Cell Death Differ ; 11(5): 527-41, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-14765134

RESUMO

The majority of ovarian cancer cells are resistant to apoptosis induced by tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). Subtoxic concentrations of the semisynthetic retinoid N-(4-hydroxyphenyl)retinamide (4HPR) enhanced TRAIL-mediated apoptosis in ovarian cancer cell lines but not in immortalized nontumorigenic ovarian epithelial cells. The enhancement of TRAIL-mediated apoptosis by 4HPR was not due to changes in the levels of proteins known to modulate TRAIL sensitivity. The combination of 4HPR and TRAIL enhanced cleavage of multiple caspases in the death receptor pathway (including the two initiator caspases, caspase-8 and caspase-9). The 4HPR and TRAIL combination leads to mitochondrial permeability transition, significant increase in cytochrome c release, and increased caspase-9 activation. Caspase-9 may further activate caspase-8, generating an amplification loop. Stable overexpression of Bcl-xL abrogates the interaction between 4HPR and TRAIL at the mitochondrial level by blocking cytochrome c release. As a consequence, a decrease in activation of caspase-9, caspase-8, and TRAIL-mediated apoptosis occurs. These results indicate that the enhancement in TRAIL-mediated apoptosis induced by 4HPR is due to the increase in activation of multiple caspases involving an amplification loop via the mitochondrial-death pathway. These findings offer a promising and novel strategy for the treatment of ovarian cancer.


Assuntos
Apoptose/efeitos dos fármacos , Fenretinida/toxicidade , Glicoproteínas de Membrana/metabolismo , Mitocôndrias/metabolismo , Receptores X de Retinoides/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Apoptose/fisiologia , Proteínas Reguladoras de Apoptose , Caspases/metabolismo , Citocromos c/metabolismo , Fragmentação do DNA/efeitos dos fármacos , Feminino , Humanos , Ovário/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF , Células Tumorais Cultivadas
10.
Gynecol Oncol ; 81(3): 380-90, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11371126

RESUMO

OBJECTIVES: The aim of this study was to investigate whether TNF-related apoptosis-inducing ligand (TRAIL) alone or in combination with chemotherapy could induce apoptosis in ovarian cancer cells resistant to chemotherapy. METHODS: Twelve chemoresistant epithelial cancer cell lines were treated with each chemotherapeutic drug alone (cisplatin, doxorubicin, or paclitaxel), TRAIL alone, or the combination. Toxicity was assessed using the MTS assay. To assess whether growth inhibition was due to apoptosis, TUNEL assay, caspase activation (measured by caspase-3 and PARP cleavage), and the sub G0/G1 fraction of cells were measured. Synergism was confirmed by fractional inhibition and dose-effect analysis. Expression of death and decoy receptors was studied by immunoblotting and an RNase protection assay. Statistical comparison of means was performed using Student's t test. RESULTS: The majority of the chemoresistant cells were also resistant to TRAIL alone. In contrast, the combination of TRAIL and chemotherapy resulted in a significant growth inhibition over a wide range of concentrations. This interaction was synergistic by dose-effect analysis. Flow cytometry demonstrated a significant increase in the fraction of apoptotic cells by the combination compared to each reagent alone. A significant enhancement in caspase and PARP cleavage was observed upon treatment with the combination. Finally, no correlation between induction of apoptosis and level of death receptors was found. CONCLUSIONS: The data suggest that almost all the ovarian cancer cells, which are resistant to chemotherapy, are also resistant to TRAIL. The combination of TRAIL and chemotherapy overcomes this resistance in a synergistic fashion by triggering caspase-mediated apoptosis. The combination of TRAIL and chemotherapy could be useful as a therapy for chemoresistant ovarian cancers.


Assuntos
Antineoplásicos/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Apoptose/efeitos dos fármacos , Glicoproteínas de Membrana/farmacologia , Neoplasias Ovarianas/tratamento farmacológico , Fator de Necrose Tumoral alfa/farmacologia , Antineoplásicos/administração & dosagem , Apoptose/fisiologia , Proteínas Reguladoras de Apoptose , Caspase 3 , Caspases/metabolismo , Divisão Celular/efeitos dos fármacos , Cisplatino/administração & dosagem , Cisplatino/farmacologia , Doxorrubicina/administração & dosagem , Doxorrubicina/farmacologia , Resistência a Múltiplos Medicamentos , Resistencia a Medicamentos Antineoplásicos , Sinergismo Farmacológico , Feminino , Humanos , Glicoproteínas de Membrana/administração & dosagem , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Paclitaxel/administração & dosagem , Paclitaxel/farmacologia , Poli(ADP-Ribose) Polimerases/metabolismo , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Receptores do Ligante Indutor de Apoptose Relacionado a TNF , Receptores do Fator de Necrose Tumoral/biossíntese , Receptores do Fator de Necrose Tumoral/genética , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/farmacologia , Ligante Indutor de Apoptose Relacionado a TNF , Células Tumorais Cultivadas , Fator de Necrose Tumoral alfa/administração & dosagem
11.
Breast Cancer Res Treat ; 64(2): 211-9, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11194457

RESUMO

Most breast cancer cell lines are resistant to TNF-related apoptosis inducing ligand (TRAIL) induced apoptosis. In sensitive breast cancer cell lines TRAIL rapidly induces the cleavage and activation of caspases leading to the subsequent cleavage of downstream caspase substrates. In contrast, there is no caspase activation in the resistant cell lines. The transcription factor NF-KB can inhibit apoptosis induced by a variety of stimuli including activation of death receptors. We investigated whether NF-kappaB contributes to the resistance of breast cancer cells to TRAIL induced apoptosis. All of the resistant breast cancer cell lines expressed NF-kappaB and had detectable NF-kappaB activity in nuclear extracts prior to treatment with TRAIL. Upon TRAIL treatment, a significant increase in NF-kappaB activity was seen in most of the cell lines. To directly test if NF-kappaB activity contributes to the resistance of these cell lines to TRAIL, we transiently transfected the resistant cell lines with an inhibitor of NF-kappaB (IkappaBdeltaN) and measured TRAIL induced apoptosis in control and transfected cells. All of the resistant cell lines tested showed an increase in TRAIL induced apoptosis when transfected with the IKBdeltaN. These results demonstrate that TRAIL resistant breast cancer cells fail to rapidly activate the apoptotic machinery but they do activate NF-kappaB. Inhibition of NF-kappaB activity increases the sensitivity to TRAIL mediated apoptosis in resistant cells. These results suggest that agents which inhibit NF-kappaB should increase the clinical efficacy of TRAIL in breast cancer cells.


Assuntos
Neoplasias da Mama/patologia , Glicoproteínas de Membrana/farmacologia , NF-kappa B/farmacologia , Fator de Necrose Tumoral alfa/farmacologia , Apoptose , Proteínas Reguladoras de Apoptose , Interações Medicamentosas , Feminino , Humanos , Ligante Indutor de Apoptose Relacionado a TNF , Transfecção , Células Tumorais Cultivadas/efeitos dos fármacos
12.
Mol Cell Biol Res Commun ; 2(2): 111-8, 1999 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10542134

RESUMO

Studies in C. elegans and Drosophila melanogaster suggest that cbl proteins are inhibitors of epidermal growth factor receptor (EGFR) function. Here we describe that overexpression of cbl-b, a homologue of the c-cbl protooncogene, inhibits EGFR-induced apoptosis in MDA-MB-468 breast cancer cells. Overexpression of cbl-b results in a shortened duration of EGFR activation upon EGF stimulation. This is demonstrated by decreased amounts of phosphorylated EGFR as well as by inhibition of multiple downstream signaling pathways. The inhibition of signaling by cbl-b results from increased ubiquitination and degradation of the activated EGFR. The inhibitory effects of cbl-b overexpression on apoptosis and on EGFR signaling are reversed by blocking proteosomal degradation of the EGFR. These data demonstrate that the mechanism by which cbl-b inhibits EGFR-induced apoptosis is by activation-dependent degradation of the EGFR. They imply that this mechanism may be a general one whereby cbl proteins regulate intracellular signaling.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Apoptose/fisiologia , Proteínas de Transporte/metabolismo , Receptores ErbB/fisiologia , Fosfoproteínas/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Ubiquitina-Proteína Ligases , Ubiquitinas/metabolismo , Acetilcisteína/análogos & derivados , Acetilcisteína/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias da Mama , Proteínas de Transporte/genética , Cisteína Endopeptidases/efeitos dos fármacos , Cisteína Endopeptidases/metabolismo , Inibidores de Cisteína Proteinase/farmacologia , Fator de Crescimento Epidérmico/farmacologia , Receptores ErbB/efeitos dos fármacos , Humanos , Quinases de Proteína Quinase Ativadas por Mitógeno/efeitos dos fármacos , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Complexos Multienzimáticos/efeitos dos fármacos , Complexos Multienzimáticos/metabolismo , Fosfoproteínas/genética , Complexo de Endopeptidases do Proteassoma , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-cbl , Transdução de Sinais , Células Tumorais Cultivadas
13.
J Biol Chem ; 276(29): 27677-84, 2001 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-11375397

RESUMO

Cbl proteins function as ubiquitin protein ligases for the activated epidermal growth factor receptor and, thus, negatively regulate its activity. Here we show that Cbl-b is ubiquitinated and degraded upon activation of the receptor. Epidermal growth factor (EGF)-induced Cbl-b degradation requires intact RING finger and tyrosine kinase binding domains and requires binding of the Cbl-b protein to the activated EGF receptor (EGFR). Degradation of both the EGFR and the Cbl-b protein is blocked by lysosomal and proteasomal inhibitors. Other components of the EGFR-signaling complex (i.e. Grb2 and Shc) are also degraded in an EGF-induced Cbl-b-dependent fashion. Our results suggest that the ubiquitin protein ligase function of Cbl-b is regulated by coordinated degradation of the Cbl-b protein along with its substrate. Furthermore, the data demonstrate that Cbl-b mediates degradation of multiple proteins in the EGFR-signaling complex.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Proteínas de Transporte/fisiologia , Receptores ErbB/metabolismo , Fosfoproteínas/fisiologia , Transdução de Sinais/fisiologia , Ubiquitina-Proteína Ligases , Proteínas de Transporte/química , Humanos , Hidrólise , Fosfoproteínas/química , Conformação Proteica , Proteínas Proto-Oncogênicas c-cbl , Células Tumorais Cultivadas , Ubiquitinas/metabolismo
14.
Mol Cell ; 4(6): 1029-40, 1999 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-10635327

RESUMO

Receptor desensitization is accomplished by accelerated endocytosis and degradation of ligand-receptor complexes. An in vitro reconstituted system indicates that Cbl adaptor proteins directly control downregulation of the receptor for the epidermal growth factor (EGFR) by recruiting ubiquitin-activating and -conjugating enzymes. We infer a sequential process initiated by autophosphorylation of EGFR at a previously identified lysosome-targeting motif that subsequently recruits Cbl. This is followed by tyrosine phosphorylation of c-Cbl at a site flanking its RING finger, which enables receptor ubiquitination and degradation. Whereas all three members of the Cbl family can enhance ubiquitination, two oncogenic Cbl variants, whose RING fingers are defective and phosphorylation sites are missing, are unable to desensitize EGFR. Our study identifies Cbl proteins as components of the ubiquitin ligation machinery and implies that they similarly suppress many other signaling pathways.


Assuntos
Proteínas de Caenorhabditis elegans , Fator de Crescimento Epidérmico/fisiologia , Receptores ErbB/fisiologia , Proteínas de Helminto/fisiologia , Ligases/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Transdução de Sinais , Sistema Livre de Células , Ativação Enzimática , Fosforilação , Proteínas Proto-Oncogênicas c-cbl , Tirosina , Enzimas Ativadoras de Ubiquitina , Ubiquitina-Proteína Ligases
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA