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










Base de dados
Intervalo de ano de publicação
1.
Int J Mol Sci ; 24(19)2023 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-37834037

RESUMO

Topoisomerases are interesting targets in cancer chemotherapy. Here, we describe the design and synthesis of a novel copper(II) indenoisoquinoline complex, WN198. The new organometallic compound exhibits a cytotoxic effect on five adenocarcinoma cell lines (MCF-7, MDA-MB-231, HeLa, HT-29, and DU-145) with the lowest IC50 (0.37 ± 0.04 µM) for the triple-negative MDA-MB-231 breast cancer cell line. Below 5 µM, WN198 was ineffective on non-tumorigenic epithelial breast MCF-10A cells and Xenopus oocyte G2/M transition or embryonic development. Moreover, cancer cell lines showed autophagy markers including Beclin-1 accumulation and LC3-II formation. The DNA interaction of this new compound was evaluated and the dose-dependent topoisomerase I activity starting at 1 µM was confirmed using in vitro tests and has intercalation properties into DNA shown by melting curves and fluorescence measurements. Molecular modeling showed that the main interaction occurs with the aromatic ring but copper stabilizes the molecule before binding and so can putatively increase the potency as well. In this way, copper-derived indenoisoquinoline topoisomerase I inhibitor WN198 is a promising antitumorigenic agent for the development of future DNA-damaging treatments.


Assuntos
Antineoplásicos , Inibidores da Topoisomerase I , Humanos , Inibidores da Topoisomerase I/farmacologia , Cobre/farmacologia , Proliferação de Células , Inibidores da Topoisomerase/farmacologia , Antineoplásicos/química , DNA/farmacologia , Linhagem Celular Tumoral , Ensaios de Seleção de Medicamentos Antitumorais , Estrutura Molecular , Relação Estrutura-Atividade , Apoptose
2.
Nat Commun ; 13(1): 956, 2022 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-35177596

RESUMO

Cellular senescence is implicated in a great number of diseases including cancer. Although alterations in mitochondrial metabolism were reported as senescence drivers, the underlying mechanisms remain elusive. We report the mechanism altering mitochondrial function and OXPHOS in stress-induced senescent fibroblasts. We demonstrate that TRPC3 protein, acting as a controller of mitochondrial Ca2+ load via negative regulation of IP3 receptor-mediated Ca2+ release, is down regulated in senescence regardless of the type of senescence inducer. This remodelling promotes cytosolic/mitochondrial Ca2+ oscillations and elevates mitochondrial Ca2+ load, mitochondrial oxygen consumption rate and oxidative phosphorylation. Re-expression of TRPC3 in senescent cells diminishes mitochondrial Ca2+ load and promotes escape from OIS-induced senescence. Cellular senescence evoked by TRPC3 downregulation in stromal cells displays a proinflammatory and tumour-promoting secretome that encourages cancer epithelial cell proliferation and tumour growth in vivo. Altogether, our results unravel the mechanism contributing to pro-tumour behaviour of senescent cells.


Assuntos
Carcinogênese/patologia , Neoplasias/patologia , Canais de Cátion TRPC/metabolismo , Cálcio/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Senescência Celular , Retículo Endoplasmático/metabolismo , Células HEK293 , Humanos , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Mitocôndrias/metabolismo , Fosforilação Oxidativa , Cultura Primária de Células
3.
Cancer Res ; 79(13): 3306-3319, 2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-31101761

RESUMO

Expression of the suppressor of cytokine signaling-1 (SOCS1) is inactivated in hematopoietic and solid cancers by promoter methylation, miRNA-mediated silencing, and mutations. Paradoxically, SOCS1 is also overexpressed in many human cancers. We report here that the ability of SOCS1 to interact with p53 and regulate cellular senescence depends on a structural motif that includes tyrosine (Y)80 in the SH2 domain of SOCS1. Mutations in this motif are found at low frequency in some human cancers, and substitution of Y80 by a phosphomimetic residue inhibits p53-SOCS1 interaction and its functional consequences, including stimulation of p53 transcriptional activity, growth arrest, and cellular senescence. Mass spectrometry confirmed SOCS1 Y80 phosphorylation in cells, and a new mAb was generated to detect its presence in tissues by IHC. A tyrosine kinase library screen identified the SRC family as Y80-SOCS1 kinases. SRC family kinase inhibitors potentiated the SOCS1-p53 pathway and reinforced SOCS1-induced senescence. Samples from human lymphomas that often overexpress SOCS1 also displayed SRC family kinase activation, constitutive phosphorylation of SOCS1 on Y80, and SOCS1 cytoplasmic localization. Collectively, these results reveal a mechanism that inactivates the SOCS1-p53 senescence pathway and suggest that inhibition of SRC family kinases as personalized treatment in patients with lymphomas may be successful. SIGNIFICANCE: These findings show that SOCS1 phosphorylation by the SRC family inhibits its tumor-suppressive activity, indicating that patients with increased SOCS1 phosphorylation may benefit from SRC family kinase inhibitors.


Assuntos
Senescência Celular , Linfoma/patologia , Domínios e Motivos de Interação entre Proteínas , Proteína 1 Supressora da Sinalização de Citocina/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Quinases da Família src/metabolismo , Humanos , Linfoma/genética , Linfoma/metabolismo , Fosforilação , Transdução de Sinais , Proteína 1 Supressora da Sinalização de Citocina/genética , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/genética , Tirosina/química , Tirosina/genética , Tirosina/metabolismo , Domínios de Homologia de src , Quinases da Família src/genética
4.
Cell Cycle ; 18(6-7): 759-770, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30874462

RESUMO

Senescence is a tumor suppressor program characterized by a stable growth arrest while maintaining cell viability. Senescence-associated ribogenesis defects (SARD) have been shown to regulate senescence through the ability of the ribosomal protein S14 (RPS14 or uS11) to bind and inhibit the cyclin-dependent kinase 4 (CDK4). Here we report another ribosomal protein that binds and inhibits CDK4 in senescent cells: L22 (RPL22 or eL22). Enforcing the expression of RPL22/eL22 is sufficient to induce an RB and p53-dependent cellular senescent phenotype in human fibroblasts. Mechanistically, RPL22/eL22 can interact with and inhibit CDK4-Cyclin D1 to decrease RB phosphorylation both in vitro and in cells. Briefly, we show that ribosome-free RPL22/eL22 causes a cell cycle arrest which could be relevant during situations of nucleolar stress such as cellular senescence or the response to cancer chemotherapy.


Assuntos
Ciclo Celular/fisiologia , Ciclina D1/metabolismo , Quinase 4 Dependente de Ciclina/metabolismo , Proteínas de Ligação a RNA/metabolismo , Proteínas Ribossômicas/metabolismo , Ribossomos/metabolismo , Pontos de Checagem do Ciclo Celular/fisiologia , Linhagem Celular , Senescência Celular/fisiologia , Células HEK293 , Humanos , Fosforilação/fisiologia , Proteína do Retinoblastoma/metabolismo , Transdução de Sinais/fisiologia , Proteína Supressora de Tumor p53/metabolismo
5.
Cytokine ; 117: 15-22, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30776684

RESUMO

The senescence-associated secretory phenotype (SASP) defines the ability of senescent cells to express and secrete a variety of extracellular modulators that includes cytokines, chemokines, proteases, growth factors and bioactive lipids. The role of the SASP depends on the context. The SASP reinforces the senescent cell cycle arrest, stimulates the immune-mediated clearance of potentially tumorigenic cells, limits fibrosis and promotes wound healing and tissue regeneration. On the other hand, the SASP can mediate chronic inflammation and stimulate the growth and survival of tumor cells. The regulation of the SASP occurs at multiple levels including chromatin remodelling, activation of specific transcription factors such as C/EBP and NF-κB, control of mRNA translation and intracellular trafficking. Several SASP modulators have already been identified setting the stage for future research on their clinical applications.


Assuntos
Senescência Celular , Reprogramação Celular , Senescência Celular/genética , Epigênese Genética , Humanos , Lipídeos/análise , NF-kappa B/metabolismo , Neoplasias/patologia
6.
Aging Cell ; 18(2): e12889, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30614183

RESUMO

Most cancers arise in old individuals, which also accumulate senescent cells. Cellular senescence can be experimentally induced by expression of oncogenes or telomere shortening during serial passage in culture. In vivo, precursor lesions of several cancer types accumulate senescent cells, which are thought to represent a barrier to malignant progression and a response to the aberrant activation of growth signaling pathways by oncogenes (oncogene toxicity). Here, we sought to define gene expression changes associated with cells that bypass senescence induced by oncogenic RAS. In the context of pancreatic ductal adenocarcinoma (PDAC), oncogenic KRAS induces benign pancreatic intraepithelial neoplasias (PanINs), which exhibit features of oncogene-induced senescence. We found that the bypass of senescence in PanINs leads to malignant PDAC cells characterized by gene signatures of epithelial-mesenchymal transition, stem cells, and mitochondria. Stem cell properties were similarly acquired in PanIN cells treated with LPS, and in primary fibroblasts and mammary epithelial cells that bypassed Ras-induced senescence after reduction of ERK signaling. Intriguingly, maintenance of cells that circumvented senescence and acquired stem cell properties was blocked by metformin, an inhibitor of complex I of the electron transport chain or depletion of STAT3, a protein required for mitochondrial functions and stemness. Thus, our studies link bypass of senescence in premalignant lesions to loss of differentiation, acquisition of stemness features, and increased reliance on mitochondrial functions.


Assuntos
Senescência Celular/efeitos dos fármacos , Metformina/farmacologia , Células-Tronco/efeitos dos fármacos , Animais , Diferenciação Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Humanos , Camundongos , Células-Tronco/citologia , Relação Estrutura-Atividade , Células Tumorais Cultivadas
7.
Oncoscience ; 6(11-12): 386-389, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31984217

RESUMO

Suppressor of cytokine signaling (SOCS) family members are upregulated following JAK-STAT pathway activation by cytokines. SOCS proteins are recognized inhibitors of cytokine signaling playing roles in cell growth and differentiation. Moreover, SOCS1 and SOCS3 have been shown to be involved in tumor suppression through their ability to interact with p53 leading to the activation of its transcriptional program and showing the implication of SOCS family members in the regulation of apoptosis, ferroptosis and senescence. More recently, we demonstrated that the SRC family of non-receptor tyrosine kinases (SFK) can phosphorylate SOCS1 leading to its homodimerization and inhibiting its interaction with p53. Then, we reactivated the SOCS1-p53 tumor suppressor axis with the SFK inhibitor dasatinib in combination with the p53 activating compound PRIMA. This work suggests new avenues for cancer treatment and leaves open several new questions that deserve to be addressed.

8.
Nat Cell Biol ; 20(7): 789-799, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29941930

RESUMO

Cellular senescence is a tumour suppressor programme characterized by a stable cell cycle arrest. Here we report that cellular senescence triggered by a variety of stimuli leads to diminished ribosome biogenesis and the accumulation of both rRNA precursors and ribosomal proteins. These defects were associated with reduced expression of several ribosome biogenesis factors, the knockdown of which was also sufficient to induce senescence. Genetic analysis revealed that Rb but not p53 was required for the senescence response to altered ribosome biogenesis. Mechanistically, the ribosomal protein S14 (RPS14 or uS11) accumulates in the soluble non-ribosomal fraction of senescent cells, where it binds and inhibits CDK4 (cyclin-dependent kinase 4). Overexpression of RPS14 is sufficient to inhibit Rb phosphorylation, inducing cell cycle arrest and senescence. Here we describe a mechanism for maintaining the senescent cell cycle arrest that may be relevant for cancer therapy, as well as biomarkers to identify senescent cells.


Assuntos
Pontos de Checagem do Ciclo Celular , Senescência Celular , Neoplasias/metabolismo , Proteína do Retinoblastoma/metabolismo , Ribossomos/metabolismo , Fatores de Coagulação Sanguínea/genética , Fatores de Coagulação Sanguínea/metabolismo , Quinase 4 Dependente de Ciclina/genética , Quinase 4 Dependente de Ciclina/metabolismo , Células HEK293 , Humanos , Neoplasias/genética , Neoplasias/patologia , Células PC-3 , Fosforilação , Ligação Proteica , Precursores de RNA/biossíntese , Precursores de RNA/genética , RNA Ribossômico/biossíntese , RNA Ribossômico/genética , Proteínas de Ligação a RNA , Proteína do Retinoblastoma/genética , Proteínas Ribossômicas/genética , Proteínas Ribossômicas/metabolismo , Ribossomos/genética , Transdução de Sinais , Fatores de Tempo
9.
Cell Calcium ; 69: 4-18, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28760561

RESUMO

Transient Receptor Potential (TRP) family mediate the influx of monovalent and/or divalent cations into cells in response to environmental stimuli. Pharmacological or genetic manipulations of TRP channels demonstrate that TRP channels influence cell death rates, prolonging or shortening of cell survival. Due to their diverse cellular localization, TRP channels mediated Ca2+ influx generates distinct intracellular Ca2+ signals that regulate downstream pathways converging to apoptosis or survival. In this review, we summarize the accumulated knowledge focused on how TRP channel regulate cell fate and may affect different pathologies including cardiovascular, neurological, metabolic or neoplastic disorders.


Assuntos
Cálcio/metabolismo , Canais de Potencial de Receptor Transitório/metabolismo , Animais , Morte Celular , Sobrevivência Celular , Humanos
10.
Aging (Albany NY) ; 9(10): 2137-2162, 2017 10 28.
Artigo em Inglês | MEDLINE | ID: mdl-29081404

RESUMO

The mechanism by which p53 suppresses tumorigenesis remains poorly understood. In the context of aberrant activation of the JAK/STAT5 pathway, SOCS1 is required for p53 activation and the regulation of cellular senescence. In order to identify p53 target genes acting during the senescence response to oncogenic STAT5A, we characterized the transcriptome of STAT5A-expressing cells after SOCS1 inhibition. We identified a set of SOCS1-dependent p53 target genes that include several secreted proteins and genes regulating oxidative metabolism and ferroptosis. Exogenous SOCS1 was sufficient to regulate the expression of p53 target genes and sensitized cells to ferroptosis. This effect correlated with the ability of SOCS1 to reduce the expression of the cystine transporter SLC7A11 and the levels of glutathione. SOCS1 and SOCS1-dependent p53 target genes were induced during the senescence response to oncogenic STAT5A, RasV12 or the tumor suppressor PML. However, while SOCS1 sensitized cells to ferroptosis neither RasV12 nor STAT5A mimicked the effect. Intriguingly, PML turned cells highly resistant to ferroptosis. The results indicate different susceptibilities to ferroptosis in senescent cells depending on the trigger and suggest the possibility of killing senescent cells by inhibiting pathways that mediate ferroptosis resistance.


Assuntos
Senescência Celular/fisiologia , Regulação da Expressão Gênica/fisiologia , Proteína 1 Supressora da Sinalização de Citocina/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Humanos
11.
Cytokine ; 82: 80-6, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26841929

RESUMO

Normal cell proliferation is controlled by a balance between signals that promote or halt cell proliferation. Micro RNAs are emerging as key elements in providing fine signal balance in different physiological situations. Here we report that STAT5 signaling induces the miRNAs miR-19 and miR-155, which potentially antagonize the tumor suppressor axis composed by the STAT5 target gene SOCS1 (suppressor of cytokine signaling-1) and its downstream effector p53. MiRNA sponges against miR-19 or miR-155 inhibit the functions of these miRNAs and potentiate the induction of SOCS1 and p53 in mouse leukemia cells and in human myeloma cells. Adding a catalytic RNA motif of the hammerhead type within miRNA sponges against miR-155 leads to decreased miR-155 levels and increased their ability of inhibiting cell growth and cell migration in myeloma cells. The results indicate that antagonizing miRNA activity can reactivate tumor suppressor pathways downstream cytokine stimulation in tumor cells.


Assuntos
Leucemia/metabolismo , MicroRNAs/metabolismo , Mieloma Múltiplo/metabolismo , RNA Catalítico/biossíntese , RNA Neoplásico/metabolismo , Proteína 1 Supressora da Sinalização de Citocina/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Humanos , Leucemia/genética , Camundongos , MicroRNAs/antagonistas & inibidores , MicroRNAs/genética , Mieloma Múltiplo/genética , Células RAW 264.7 , RNA Catalítico/genética , RNA Neoplásico/antagonistas & inibidores , RNA Neoplásico/genética , Proteína 1 Supressora da Sinalização de Citocina/genética , Proteína Supressora de Tumor p53/genética
12.
Biochim Biophys Acta ; 1853(9): 1974-9, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25764980

RESUMO

Senescence is one of the primary responses to the activation of oncoproteins or down-regulation of tumor suppressors in normal cells and is therefore considered as being anti-tumorigenic but the mechanisms controlling this process are still much unknown. Calcium (Ca²âº) plays a major role in many cellular processes and calcium channels control many of the "hallmarks of cancer" but their involvement in tumor initiation is poorly understood and remains unclear. Therefore, in this article we review some striking senescence-associated characteristics and their potential regulation by Ca²âº. The main aim is to produce plausible hypothesis on how calcium homeostasis may participate in cancer-related senescence. This article is part of a Special Issue entitled: 13th European Symposium on Calcium.


Assuntos
Sinalização do Cálcio , Transformação Celular Neoplásica/metabolismo , Senescência Celular , Homeostase , Neoplasias/metabolismo , Animais , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Humanos , Neoplasias/genética , Neoplasias/patologia
13.
Proc Natl Acad Sci U S A ; 111(37): E3870-9, 2014 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-25172921

RESUMO

Transient receptor potential vanilloid subfamily member 6 (TRPV6) is a highly selective calcium channel that has been considered as a part of store-operated calcium entry (SOCE). Despite its first discovery in the early 2000s, the role of this channel in prostate cancer (PCa) remained, until now, obscure. Here we show that TRPV6 mediates calcium entry, which is highly increased in PCa due to the remodeling mechanism involving the translocation of the TRPV6 channel to the plasma membrane via the Orai1/TRPC1-mediated Ca(2+)/Annexin I/S100A11 pathway, partially contributing to SOCE. The TRPV6 calcium channel is expressed de novo by the PCa cell to increase its survival by enhancing proliferation and conferring apoptosis resistance. Xenografts in nude mice and bone metastasis models confirmed the remarkable aggressiveness of TRPV6-overexpressing tumors. Immunohistochemical analysis of these demonstrated the increased expression of clinical markers such as Ki-67, prostate specific antigen, synaptophysin, CD31, and CD56, which are strongly associated with a poor prognosis. Thus, the TRPV6 channel acquires its oncogenic potential in PCa due to the remodeling mechanism via the Orai1-mediated Ca(2+)/Annexin I/S100A11 pathway.


Assuntos
Canais de Cálcio/metabolismo , Membrana Celular/metabolismo , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Canais de Cátion TRPV/metabolismo , Animais , Anexina A1/metabolismo , Apoptose , Neoplasias Ósseas/diagnóstico por imagem , Neoplasias Ósseas/secundário , Cálcio/metabolismo , Carcinogênese/patologia , Linhagem Celular Tumoral , Sobrevivência Celular , Progressão da Doença , Retículo Endoplasmático/metabolismo , Células HEK293 , Humanos , Imuno-Histoquímica , Masculino , Camundongos Nus , Invasividade Neoplásica , Proteínas de Neoplasias/metabolismo , Proteína ORAI1 , Fenótipo , Transporte Proteico , Radiografia , Proteínas S100/metabolismo , Transdução de Sinais , Ensaios Antitumorais Modelo de Xenoenxerto
14.
Mol Biol Cell ; 25(5): 554-65, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24403608

RESUMO

The expression of the forkhead transcription factor checkpoint suppressor 1 (CHES1), also known as FOXN3, is reduced in many types of cancers. We show here that CHES1 decreases protein synthesis and cell proliferation in tumor cell lines but not in normal fibroblasts. Conversely, short hairpin RNA-mediated depletion of CHES1 increases tumor cell proliferation. Growth suppression depends on the CHES1 forkhead DNA-binding domain and correlates with the nuclear localization of CHES1. CHES1 represses the expression of multiple genes, including the kinases PIM2 and DYRK3, which regulate protein biosynthesis, and a number of genes in cilium biogenesis. CHES1 binds directly to the promoter of PIM2, and in cells expressing CHES1 the levels of PIM2 are reduced, as well as the phosphorylation of the PIM2 target 4EBP1. Overexpression of PIM2 or eIF4E partially reverses the antiproliferative effect of CHES1, indicating that PIM2 and protein biosynthesis are important targets of the antiproliferative effect of CHES1. In several human hematopoietic cancers, CHES1 and PIM2 expressions are inversely correlated, suggesting that repression of PIM2 by CHES1 is clinically relevant.


Assuntos
Proteínas de Ciclo Celular/fisiologia , Proliferação de Células , Biossíntese de Proteínas/genética , Proteínas Serina-Treonina Quinases/genética , Proteínas Proto-Oncogênicas/genética , Proteínas Repressoras/fisiologia , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Fibroblastos/metabolismo , Fatores de Transcrição Forkhead , Regulação Neoplásica da Expressão Gênica , Humanos , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo
15.
Aging Cell ; 12(3): 489-98, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23521863

RESUMO

We show that the antidiabetic drug metformin inhibits the expression of genes coding for multiple inflammatory cytokines seen during cellular senescence. Conditioned medium (CM) from senescent cells stimulates the growth of prostate cancer cells but treatment of senescent cells with metformin inhibited this effect. Bioinformatic analysis of genes downregulated by metformin suggests that the drug blocks the activity of the transcription factor NF-κB. In agreement, metformin prevented the translocation of NF-κB to the nucleus and inhibited the phosphorylation of IκB and IKKα/ß, events required for activation of the NF-κB pathway. These effects were not dependent on AMPK activation or on the context of cellular senescence, as metformin inhibited the NF-κB pathway stimulated by lipopolysaccharide (LPS) in ampk null fibroblasts and in macrophages. Taken together, our results provide a novel mechanism for the antiaging and antineoplastic effects of metformin reported in animal models and in diabetic patients taking this drug.


Assuntos
Senescência Celular/efeitos dos fármacos , Quinase I-kappa B/metabolismo , Metformina/farmacologia , NF-kappa B/metabolismo , Fator de Transcrição RelA/metabolismo , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Meios de Cultivo Condicionados , Citocinas/metabolismo , Ativação Enzimática/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica , Humanos , Hipoglicemiantes/farmacologia , Inflamação/genética , Lipopolissacarídeos/farmacologia , Macrófagos/efeitos dos fármacos , Masculino , Camundongos , Fosforilação/efeitos dos fármacos , Neoplasias da Próstata , Interferência de RNA , RNA Interferente Pequeno , Transdução de Sinais/efeitos dos fármacos , Fator de Transcrição RelA/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
16.
Clin Cancer Res ; 17(7): 1741-52, 2011 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-21350004

RESUMO

PURPOSE: Given that nerve growth factor has previously been shown to be involved in breast cancer progression, we have tested here the hypothesis that the other neurotrophins (NT) are expressed and have an influence in breast tumor growth. EXPERIMENTAL DESIGN: The expression of brain-derived neurotrophic factor (BDNF), NT-3 and NT-4/5, as well as the neurotrophin receptor p75(NTR), TrkB, and TrkC, was studied by RT-PCR, Western blotting, and immunohistochemistry in cell lines and tumor biopsies. The biological impacts of neurotrophins, and associated mechanisms, were analyzed in cell cultures and xenografted mice. RESULTS: BDNF and NT-4/5 were expressed and secreted by breast cancer cells, and the use of blocking antibodies suggested an autocrine loop mediating cell resistance to apoptosis. The corresponding tyrosine kinase receptor TrkB was only rarely observed at full length, whereas the expression of TrkB-T1, lacking the kinase domain, as well as p75(NTR), were detected in all tested breast cancer cell lines and tumor biopsies. In contrast, NT-3 and TrkC were not detected. SiRNA against p75(NTR) and TrkB-T1 abolished the antiapoptotic effect of BDNF and NT-4/5, whereas the pharmacological inhibitors K252a and PD98059 had no effect, suggesting the involvement of p75(NTR) and TrkB-T1, but not kinase activities from Trks and MAPK. In xenografted mice, anti-BDNF, anti-NT-4/5, anti-p75(NTR), or anti-TrkB-T1 treatments resulted in tumor growth inhibition, characterized by an increase in cell apoptosis, but with no change in proliferation. CONCLUSION: BDNF and NT-4/5 contribute to breast cancer cell survival and can serve as prospective targets in attempts to inhibit tumor growth.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/metabolismo , Neoplasias da Mama/patologia , Sobrevivência Celular , Fatores de Crescimento Neural/metabolismo , Animais , Anticorpos/farmacologia , Antineoplásicos/farmacologia , Apoptose , Fator Neurotrófico Derivado do Encéfalo/imunologia , Fator Neurotrófico Derivado do Encéfalo/farmacologia , Neoplasias da Mama/metabolismo , Carcinoma Ductal de Mama/metabolismo , Carcinoma Ductal de Mama/patologia , Carcinoma Intraductal não Infiltrante/metabolismo , Carcinoma Intraductal não Infiltrante/patologia , Carcinoma Lobular/metabolismo , Carcinoma Lobular/patologia , Carcinoma Medular/metabolismo , Carcinoma Medular/patologia , Linhagem Celular Tumoral , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Fatores Imunológicos/farmacologia , Metástase Linfática , Camundongos , Camundongos SCID , Transplante de Neoplasias , Fatores de Crescimento Neural/imunologia , Fatores de Crescimento Neural/farmacologia , Neurotrofina 3/genética , Neurotrofina 3/metabolismo , RNA Interferente Pequeno/genética , Receptor de Fator de Crescimento Neural/genética , Receptor de Fator de Crescimento Neural/imunologia , Receptor de Fator de Crescimento Neural/metabolismo , Receptor trkB/genética , Receptor trkB/metabolismo , Receptor trkC/genética , Receptor trkC/metabolismo , Transcrição Gênica , Transplante Heterólogo
17.
Cell Signal ; 22(12): 1864-73, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20667470

RESUMO

The p75 neurotrophin receptor (p75(NTR)) plays a critical role in various neuronal and non-neuronal cell types by regulating cell survival, differentiation and proliferation. To evaluate the influence of p75(NTR) in breast cancer development, we have established and characterized breast cancer cells which stably overexpress p75(NTR). We showed that p75(NTR) overexpression per se promoted cell survival to apoptogens with a concomitant slowdown of cell growth. The pro-survival effect is associated with an increased expression of the inhibitor of apoptosis protein-1 (c-IAP1), a decrease of TRAIL-induced cleavage of PARP, procaspase 9 and procaspase 3, and a decrease of cytochrome C release from the mitochondria. The anti-proliferative effect is due to a cell accumulation in G0/G1, associated with a decrease of Rb phosphorylation and an increase of p21(waf1). Interestingly, inhibition of p21(waf1) with siRNA not only restores proliferation but also abolishes the pro-survival effect of p75(NTR), indicating the key role of p21(waf1) in the biological functions of p75(NTR). Finally, using a SCID mice xenograft model, we showed that p75(NTR) overexpression favors tumor growth and strongly increases tumor resistance to anti-tumoral treatment. Together, our findings suggest that p75(NTR) overexpression in breast tumor cells could favor tumor survival and contribute to tumor resistance to drugs. This provides a rationale to consider p75(NTR) as a potential target for the future design of innovative therapeutic strategies.


Assuntos
Neoplasias da Mama/patologia , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Receptor de Fator de Crescimento Neural/metabolismo , Animais , Apoptose/efeitos dos fármacos , Caspase 3/metabolismo , Caspase 9/metabolismo , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Inibidor de Quinase Dependente de Ciclina p21/farmacologia , Grupo dos Citocromos c/metabolismo , Feminino , Humanos , Proteínas Inibidoras de Apoptose/metabolismo , Camundongos , Camundongos SCID , Mitocôndrias/genética , Mitocôndrias/metabolismo , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
18.
Mol Cancer ; 9: 157, 2010 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-20569463

RESUMO

BACKGROUND: Although several anti-angiogenic therapies have been approved in the treatment of cancer, the survival benefits of such therapies are relatively modest. Discovering new molecules and/or better understating signaling pathways of angiogenesis is therefore essential for therapeutic improvements. The objective of the present study was to determine the involvement of nerve growth factor (NGF) in breast cancer angiogenesis and the underlying molecular mechanisms. RESULTS: We showed that both recombinant NGF and NGF produced by breast cancer cells stimulated angiogenesis in Matrigel plugs in immunodeficient mice. NGF strongly increased invasion, cord formation and the monolayer permeability of endothelial cells. Moreover, NGF-stimulated invasion was under the control of its tyrosine kinase receptor (TrkA) and downstream signaling pathways such as PI3K and ERK, leading to the activation of matrix metalloprotease 2 and nitric oxide synthase. Interestingly, NGF increased the secretion of VEGF in both endothelial and breast cancer cells. Inhibition of VEGF, with a neutralizing antibody, reduced about half of NGF-induced endothelial cell invasion and angiogenesis in vivo. CONCLUSIONS: Our findings provided direct evidence that NGF could be an important stimulator for breast cancer angiogenesis. Thus, NGF, as well as the activated signaling pathways, should be regarded as potential new targets for anti-angiogenic therapy against breast cancer.


Assuntos
Neoplasias da Mama/irrigação sanguínea , Neovascularização Patológica , Fator de Crescimento Neural/farmacologia , Animais , Western Blotting , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Sistema de Sinalização das MAP Quinases , Camundongos , Camundongos SCID , Óxido Nítrico/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor trkA/metabolismo , Proteínas Recombinantes/farmacologia , Fator A de Crescimento do Endotélio Vascular/fisiologia
19.
Nutr Cancer ; 51(2): 178-83, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15860440

RESUMO

Polyunsaturated fatty acids (PUFAs) have been reported to enhance the efficacy of chemotherapeutic agents that produce reactive oxygen species such as anthracyclines. We previously reported in a human breast cancer cell line that the increased cytotoxic activity of anthracyclines by several PUFAs was abolished by antioxidants and enhanced by pro-oxidants, suggesting that lipid peroxidation was involved in this effect. To determine the relevance of this observation in vivo, we examined the effect of the oxidative status of the diet on the activity of epirubicin against N-methylnitrosourea-induced mammary tumors in Sprague-Dawley rats. Three groups of rats were fed a basal diet enriched with dietary n-3 PUFA (sardine oil, 15%) alone (control group), with addition of an antioxidant (alpha-tocopherol, 100 UI/kg diet), or with addition of an oxidant system (dehydroascorbate/naphthoquinone). When the first mammary tumor reached 1 cm2, epirubicin was administrated weekly for 3 wk, and subsequent change in tumor size was documented over time. Two weeks after the end of epirubicin injections, tumor size was increased by 34% in the control group. In the pro-oxidant group, tumor size was decreased by 50%. In contrast, tumor size was increased by 188% in the antioxidant group. Thus, addition of pro-oxidants in a fish oil-enriched diet increased the sensitization of mammary tumors to chemotherapy, whereas addition of alpha-tocopherol suppressed tumor response in vivo, indicating that interaction between components of the diet has to be carefully controlled during chemotherapy.


Assuntos
Antraciclinas/antagonistas & inibidores , Antraciclinas/farmacologia , Antioxidantes/farmacologia , Dieta/efeitos adversos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Óleos de Peixe/administração & dosagem , Neoplasias Mamárias Experimentais/tratamento farmacológico , alfa-Tocoferol/farmacologia , Tecido Adiposo/efeitos dos fármacos , Tecido Adiposo/metabolismo , Animais , Antibióticos Antineoplásicos/antagonistas & inibidores , Antibióticos Antineoplásicos/farmacologia , Carcinógenos , Modelos Animais de Doenças , Antagonismo de Drogas , Epirubicina/antagonistas & inibidores , Epirubicina/farmacologia , Ácidos Graxos Insaturados/farmacologia , Feminino , Malondialdeído/metabolismo , Neoplasias Mamárias Experimentais/induzido quimicamente , Metilnitrosoureia , Oxidantes/administração & dosagem , Ratos , Ratos Sprague-Dawley , Fatores de Tempo
20.
Nat Med ; 11(1): 77-84, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15619633

RESUMO

Chromatin is a dynamic macromolecular structure epigenetically modified to regulate specific gene expression. Altered chromatin function can lead to aberrant expression of growth regulators and may, ultimately, cause cancer. That many human diseases have epigenetic etiology has stimulated the development of 'epigenetic' therapies. Inhibitors of histone deacetylases (HDACIs) induce proliferation arrest, maturation and apoptosis of cancer cells, but not normal cells, in vitro and in vivo, and are currently being tested in clinical trials. We investigated the mechanism(s) underlying this tumor selectivity. We report that HDACIs induce, in addition to p21, expression of TRAIL (Apo2L, TNFSF10) by directly activating the TNFSF10 promoter, thereby triggering tumor-selective death signaling in acute myeloid leukemia (AML) cells and the blasts of individuals with AML. RNA interference revealed that the induction of p21, TRAIL and differentiation are separable activities of HDACIs. HDACIs induced proliferation arrest, TRAIL-mediated apoptosis and suppression of AML blast clonogenicity irrespective of French-American-British (FAB) classification status, karyotype and immunophenotype. No apoptosis was seen in normal CD34(+) progenitor cells. Our results identify TRAIL as a mediator of the anticancer action of HDACIs.


Assuntos
Apoptose/efeitos dos fármacos , Inibidores de Histona Desacetilases , Leucemia Mieloide/tratamento farmacológico , Glicoproteínas de Membrana/metabolismo , Receptores de Superfície Celular/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Doença Aguda , Proteínas Reguladoras de Apoptose , Humanos , Ligante Indutor de Apoptose Relacionado a TNF , Proteína Supressora de Tumor p53/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...