Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Cell Cycle ; 12(3): 473-9, 2013 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-23324343

RESUMO

Circumstantial evidence suggests that colon carcinogenesis can ensue the transient tetraploidization of (pre-)malignant cells. In line with this notion, the tumor suppressors APC and TP53, both of which are frequently inactivated in colon cancer, inhibit tetraploidization in vitro and in vivo. Here, we show that-contrarily to their wild-type counterparts- Tp53 (-/-) colonocytes are susceptible to drug-induced or spontaneous tetraploidization in vitro. Colon organoids generated from tetraploid Tp53 (-/-) cells exhibit a close-to-normal morphology as compared to their diploid Tp53 (-/-) counterparts, yet the colonocytes constituting these organoids are characterized by an increased cell size and an elevated expression of the immunostimulatory protein calreticulin on the cell surface. The subcutaneous injection of tetraploid Tp53 (-/-) colon organoids led to the generation of proliferating tumors in immunodeficient, but not immunocompetent, mice. Thus, tetraploid Tp53 (-/-) colonocytes fail to survive in immunocompetent mice and develop neoplastic lesions in immunocompromised settings only. These results suggest that tetraploidy is particularly oncogenic in the context of deficient immunosurveillance.


Assuntos
Transformação Celular Neoplásica/metabolismo , Neoplasias do Colo/imunologia , Vigilância Imunológica/imunologia , Proteína Supressora de Tumor p53/genética , Animais , Antineoplásicos/farmacologia , Calreticulina/biossíntese , Transformação Celular Neoplásica/genética , Colo/metabolismo , Colo/patologia , Neoplasias do Colo/genética , Proteínas de Membrana/biossíntese , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Nocodazol/farmacologia , Tetraploidia , Proteína Supressora de Tumor p53/metabolismo
2.
Methods Mol Biol ; 965: 93-120, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23296653

RESUMO

One of the most prominent features of cellular senescence, a stress response that prevents the propagation of cells that have accumulated potentially oncogenic alterations, is a permanent loss of proliferative potential. Thus, at odds with quiescent cells, which resume proliferation when stimulated to do so, senescent cells cannot proceed through the cell cycle even in the presence of mitogenic factors. Here, we describe a set of cytofluorometric techniques for studying how chemical and/or physical stimuli alter the cell cycle in vitro, in both qualitative and quantitative terms. Taken together, these methods allow for the identification of bona fide cytostatic effects as well as for a refined characterization of cell cycle distributions, providing information on proliferation, DNA content as well as on the presence of cell cycle phase-specific markers. At the end of the chapter, a set of guidelines is offered to assist researchers that approach the study of the cell cycle with the interpretation of results.


Assuntos
Ciclo Celular , Citometria de Fluxo/métodos , Contagem de Células , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Ciclina B1/metabolismo , Células HCT116 , Histonas/metabolismo , Humanos , Microesferas , Compostos de Fenilureia/metabolismo , Fosforilação/efeitos dos fármacos , Fase S/efeitos dos fármacos
3.
J Exp Med ; 208(9): 1823-34, 2011 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-21859844

RESUMO

Extracellular adenosine triphosphate (ATP) can activate purinergic receptors of the plasma membrane and modulate multiple cellular functions. We report that ATP is released from HIV-1 target cells through pannexin-1 channels upon interaction between the HIV-1 envelope protein and specific target cell receptors. Extracellular ATP then acts on purinergic receptors, including P2Y2, to activate proline-rich tyrosine kinase 2 (Pyk2) kinase and transient plasma membrane depolarization, which in turn stimulate fusion between Env-expressing membranes and membranes containing CD4 plus appropriate chemokine co-receptors. Inhibition of any of the constituents of this cascade (pannexin-1, ATP, P2Y2, and Pyk2) impairs the replication of HIV-1 mutant viruses that are resistant to conventional antiretroviral agents. Altogether, our results reveal a novel signaling pathway involved in the early steps of HIV-1 infection that may be targeted with new therapeutic approaches.


Assuntos
Trifosfato de Adenosina/metabolismo , Membrana Celular/metabolismo , Infecções por HIV/metabolismo , HIV-1/fisiologia , Mutação , Receptores Purinérgicos P2Y2/metabolismo , Trifosfato de Adenosina/genética , Adulto , Terapia Antirretroviral de Alta Atividade/métodos , Membrana Celular/genética , Conexinas/genética , Conexinas/metabolismo , Farmacorresistência Viral/efeitos dos fármacos , Farmacorresistência Viral/genética , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/genética , Quinase 2 de Adesão Focal/genética , Quinase 2 de Adesão Focal/metabolismo , Infecções por HIV/tratamento farmacológico , Infecções por HIV/genética , Humanos , Masculino , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Receptores Purinérgicos P2Y2/genética , Transdução de Sinais , Replicação Viral/efeitos dos fármacos , Replicação Viral/genética
4.
Methods Mol Biol ; 761: 47-63, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21755440

RESUMO

During malignant transformation, cells can increase their ploidy and hence become polyploid (mostly tetraploid). Frequently, however, tetraploid cells undergo asymmetric divisions, in turn entailing a reduction in ploidy and the acquisition of a pseudo-diploid, aneuploid state. To investigate such a stepwise aneuploidization process, we developed a cytofluorometric method (based on the heterogeneity in cell size and/or chromatin content) that allows for the cloning and subsequent functional analysis of cells with distinct ploidies. Here, we detail this methodology, which has been instrumental for investigating the functional link between ploidy status and oncogenesis.


Assuntos
Separação Celular , Diploide , Tetraploidia , Linhagem Celular Tumoral , Separação Celular/instrumentação , Separação Celular/métodos , Humanos , Neoplasias/genética , Neoplasias/patologia , Plasmídeos/genética , Coloração e Rotulagem , Transfecção
5.
Autophagy ; 7(8): 883-91, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21460612

RESUMO

Autophagic flux can be measured by determining the declining abundance of autophagic substrates such as sequestosome 1 (SQSTM1, better known as p62), which is sequestered in autophagosomes upon its direct interaction with LC3. However, the total amount of p62 results from two opposed processes, namely its synthesis (which can be modulated by some cellular stressors including autophagy inducers) and its degradation. To avoid this problem, we generated a stable cell line expressing a chimeric protein composed by p62 and the HaloTag (®) protein, which serves as a receptor for fluorescent HaloTag (®) ligands. Upon labeling with HaloTag (®) ligands (which form covalent, near-to-undissociable bonds with the Halotag (®) receptor) and washing, the resulting fluorescent labeling is not influenced by de novo protein synthesis, therefore allowing for the specific monitoring of the fusion protein decline without any interference by protein synthesis. We demonstrate that a HaloTag (®) -p62 fusion protein stably expressed in suitable cell lines can be used to monitor autophagy by flow cytometry and automated fluorescence microscopy. We surmise that this system could be adapted to high-throughput applications.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Autofagia , Citometria de Fluxo/métodos , Microscopia de Fluorescência/métodos , Linhagem Celular Tumoral , Humanos , Organelas/metabolismo , Frações Subcelulares/metabolismo , Especificidade por Substrato
6.
Cell Cycle ; 9(14): 2823-9, 2010 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-20686359

RESUMO

The tumor suppressor protein p53 plays a major role in preserving genomic stability. p53 suppresses a pathway leading from normal diploidy to neoplastic aneuploidy (via an intermediate metastable stage of tetraploidy) at two levels: first by preventing the generation/survival of tetraploid cells, and second by repressing their aberrant multipolar division. Here, we report the characterization of p53(-/-) tetraploid cells, which-at difference with both their p53(-/-) diploid and their p53(+/+) tetraploid counterparts-manifest a marked hyperphosporylation of the mitogen-activated protein kinase MAPK14 (best known as p38alpha) that is particularly strong during mitosis. In p53(-/-) tetraploid cells, phosphorylated p38alpha accumulated at centrosomes during the metaphase and at midbodies during the telophase. Selective knockdown or pharmacological inhibition of p38alpha had a dramatic effect on p53(-/-) (but not p53(+/+)) tetraploids, causing the activation of the spindle assembly checkpoint, an arrest during the metaphase, a major increase in abnormal bipolar and monopolar mitoses, as well as an increment in the generation of multinuclear cells. We conclude that the mitotic progression of p53(-/-) (but not p53(+/+)) tetraploids heavily relies on p38alpha, revealing a novel function for this protein in the context of aneuploidizing cell divisions.


Assuntos
Proteína Quinase 14 Ativada por Mitógeno/metabolismo , Poliploidia , Proteína Supressora de Tumor p53/metabolismo , Linhagem Celular Tumoral , Centrossomo/metabolismo , Humanos , Metáfase , Proteína Quinase 14 Ativada por Mitógeno/genética , Mitose , Fosforilação , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Telófase , Tubulina (Proteína)/metabolismo , Proteína Supressora de Tumor p53/genética
7.
Cancer Res ; 70(10): 4123-32, 2010 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-20424115

RESUMO

During childhood, the thyroid gland is one of the most sensitive organs to the carcinogenetic effects of ionizing radiation that may lead to papillary thyroid carcinoma (PTC) associated with RET/PTC oncogene rearrangement. Exposure to ionizing radiation induces a transient "oxidative burst" through radiolysis of water, which can cause DNA damage and mediates part of the radiation effects. H(2)O(2) is a potent DNA-damaging agent that induces DNA double-strand breaks, and consequently, chromosomal aberrations. Irradiation by 5 Gy X-ray increased extracellular H(2)O(2). Therefore, we investigated the implication of H(2)O(2) in the generation of RET/PTC1 rearrangement after X-ray exposure. We developed a highly specific and sensitive nested reverse transcription-PCR method. By using the human thyroid cell line HTori-3, previously found to produce RET/PTC1 after gamma-irradiation, we showed that H(2)O(2), generated during a 5 Gy X-ray irradiation, causes DNA double-strand breaks and contributes to RET/PTC1 formation. Pretreatment of cells with catalase, a scavenger of H(2)O(2), significantly decreased RET/PTC1 rearrangement formation. Finally, RET/PTC chromosomal rearrangement was detected in HTori-3.1 cells after exposure of cells to H(2)O(2) (25 micromol/L), at a dose that did not affect the cell viability. This study shows for the first time that H(2)O(2) is able to cause RET/PTC1 rearrangement in thyroid cells and consequently highlights that oxidative stress could be responsible for the occurrence of RET/PTC1 rearrangement found in thyroid lesions even in the absence of radiation exposure.


Assuntos
Carcinoma Papilar/patologia , Rearranjo Gênico/efeitos da radiação , Peróxido de Hidrogênio/farmacologia , Proteínas de Fusão Oncogênica/genética , Proteínas Tirosina Quinases/genética , Glândula Tireoide/efeitos da radiação , Neoplasias da Glândula Tireoide/genética , Western Blotting , Carcinoma Papilar/genética , Carcinoma Papilar/metabolismo , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/efeitos da radiação , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/efeitos da radiação , Células Cultivadas , Embrião de Mamíferos/citologia , Embrião de Mamíferos/efeitos dos fármacos , Embrião de Mamíferos/efeitos da radiação , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/efeitos da radiação , Rearranjo Gênico/efeitos dos fármacos , Humanos , Pulmão/citologia , Pulmão/efeitos dos fármacos , Pulmão/efeitos da radiação , Proteínas de Fusão Oncogênica/metabolismo , Oxidantes/farmacologia , Proteínas Tirosina Quinases/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Glândula Tireoide/efeitos dos fármacos , Glândula Tireoide/metabolismo , Neoplasias da Glândula Tireoide/metabolismo , Neoplasias da Glândula Tireoide/patologia , Raios X
8.
EMBO J ; 29(7): 1272-84, 2010 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-20186124

RESUMO

Tetraploidy can constitute a metastable intermediate between normal diploidy and oncogenic aneuploidy. Here, we show that the absence of p53 is not only permissive for the survival but also for multipolar asymmetric divisions of tetraploid cells, which lead to the generation of aneuploid cells with a near-to-diploid chromosome content. Multipolar mitoses (which reduce the tetraploid genome to a sub-tetraploid state) are more frequent when p53 is downregulated and the product of the Mos oncogene is upregulated. Mos inhibits the coalescence of supernumerary centrosomes that allow for normal bipolar mitoses of tetraploid cells. In the absence of p53, Mos knockdown prevents multipolar mitoses and exerts genome-stabilizing effects. These results elucidate the mechanisms through which asymmetric cell division drives chromosomal instability in tetraploid cells.


Assuntos
Carcinoma/metabolismo , Neoplasias do Colo/metabolismo , Genes mos , Mitose , Poliploidia , Proteína Supressora de Tumor p53/metabolismo , Aneuploidia , Animais , Carcinoma/genética , Linhagem Celular Tumoral , Centrossomo/metabolismo , Instabilidade Cromossômica , Neoplasias do Colo/genética , Feminino , Deleção de Genes , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos , Camundongos Nus , Proteína Supressora de Tumor p53/genética
9.
Cell Cycle ; 8(22): 3723-8, 2009 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-19855167

RESUMO

Chemotherapy can induce anticancer immune responses. In contrast to a widely extended prejudice, apoptotic cell death is often more efficient in eliciting a protective anticancer immune response than necrotic cell death. Recently, we have found that purinergic receptors of the P2X7 type are required for the anticancer immune response induced by chemotherapy. ATP is the endogenous ligand that has the highest affinity for P2X7. Therefore, we investigated the capacity of a panel of chemotherapeutic agents to induce ATP release from cancer cells. Here, we describe that multiple distinct anticancer drugs reduce the intracellular concentration of ATP before and during the manifestation of apoptotic characteristics such as the dissipation of the mitochondrial transmembrane potential and the exposure of phosphatidylserine residues on the plasma membrane. Indeed, as apoptosis progresses, intracellular ATP concentrations decrease, although even advanced-stage apoptotic cells still contain sizeable ATP levels. Only when cells enter secondary necrosis, the ATP concentration falls to undetectable levels. Concomitantly, a wide range of chemotherapeutic agents causes the release of ATP into the extracellular space as they induce tumor cell death. Hence, ATP release is a general correlate of apoptotic cell death induced by conventional anticancer therapies.


Assuntos
Trifosfato de Adenosina/metabolismo , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias/tratamento farmacológico , Animais , Cádmio/farmacologia , Linhagem Celular Tumoral , Cisplatino/farmacologia , Etoposídeo/farmacologia , Citometria de Fluxo , Imunofluorescência , Camundongos , Mitomicina/farmacologia , Mitoxantrona/farmacologia , Compostos Organoplatínicos/farmacologia , Oxaliplatina , Esfingosina/análogos & derivados , Esfingosina/farmacologia , Tapsigargina/farmacologia
10.
Cell Cycle ; 8(7): 1030-5, 2009 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-19270519

RESUMO

Tetraploid cells may constitute a metastable intermediate between normal euploidy and cancer-associated aneuploidy. Tetraploid cells are relatively more resistant against DNA damaging agents and are genetically unstable, due to their tendency towards multipolar, asymmetric division. Therefore, it is important to develop strategies for the selective removal of tetraploid cells. Here, we show that targeting the mitotic kinesin Eg5 (also known as kinesin spindle protein, KSP) by a small interfering RNA (siRNA) or by the pharmacological inhibitor dimethylenastron (DIMEN) kills tetraploid tumor cells more efficiently than their diploid precursors. Cell death occurs after an attempt of monoastral mitosis that, in diploid cells, is followed by a prolonged mitotic arrest and morphological reversion to the interphase, with a 4n DNA content. In contrast, DIMEN-treated tetraploid cells exhibit a shorter mitotic arrest, bipolar or multipolar karyokinesis, followed by apoptosis of the daughter cells, as assessed by fluorescence videomicroscopy of cells that express a histone 2B-GFP fusion construct to monitor their chromosomes. Cell death occurred with hallmarks of apoptosis, namely loss of the mitochondrial transmembrane potential and terminal chromatin compaction. In conclusion, tetraploid cells are particular vulnerable to undergo mitotic catastrophe after genetic or pharmacological inhibition of Eg5.


Assuntos
Antineoplásicos/farmacologia , Cinesinas/antagonistas & inibidores , Neoplasias/patologia , Poliploidia , Quinazolinas/farmacologia , Tionas/farmacologia , Apoptose , Linhagem Celular Tumoral , Técnicas de Silenciamento de Genes , Humanos , Cinesinas/genética , Mitose/efeitos dos fármacos , Neoplasias/genética , Neoplasias/metabolismo , RNA Interferente Pequeno/genética
11.
J Clin Invest ; 117(7): 1844-55, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17557121

RESUMO

Tumor radioresponsiveness depends on endothelial cell death, which leads in turn to tumor hypoxia. Radiation-induced hypoxia was recently shown to trigger tumor radioresistance by activating angiogenesis through hypoxia-inducible factor 1-regulated (HIF-1-regulated) cytokines. We show here that combining targeted radioiodide therapy with angiogenic inhibitors, such as canstatin, enhances direct tumor cell apoptosis, thereby overcoming radio-induced HIF-1-dependent tumor survival pathways in vitro and in vivo. We found that following dual therapy, HIF-1alpha increases the activity of the canstatin-induced alpha(v)beta(5) signaling tumor apoptotic pathway and concomitantly abrogates mitotic checkpoint and tetraploidy triggered by radiation. Apoptosis in conjunction with mitotic catastrophe leads to lethal tumor damage. We discovered that HIF-1 displays a radiosensitizing activity that is highly dependent on treatment modalities by regulating key apoptotic molecular pathways. Our findings therefore support a crucial role for angiogenesis inhibitors in shifting the fate of radiation-induced HIF-1alpha activity from hypoxia-induced tumor radioresistance to hypoxia-induced tumor apoptosis. This study provides a basis for developing new biology-based clinically relevant strategies to improve the efficacy of radiation oncology, using HIF-1 as an ally for cancer therapy.


Assuntos
Apoptose/efeitos da radiação , Colágeno Tipo IV/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Neoplasias/metabolismo , Neoplasias/patologia , Fragmentos de Peptídeos/metabolismo , Adenoviridae/genética , Animais , Linhagem Celular , Colágeno Tipo IV/genética , Modelos Animais de Doenças , Regulação Neoplásica da Expressão Gênica , Terapia Genética , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Integrinas/metabolismo , Camundongos , Camundongos Transgênicos , Mitocôndrias/metabolismo , Neoplasias/irrigação sanguínea , Neoplasias/terapia , Fragmentos de Peptídeos/genética , Transdução de Sinais , Simportadores/genética , Simportadores/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Nat Med ; 13(1): 54-61, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17187072

RESUMO

Anthracyclin-treated tumor cells are particularly effective in eliciting an anticancer immune response, whereas other DNA-damaging agents such as etoposide and mitomycin C do not induce immunogenic cell death. Here we show that anthracyclins induce the rapid, preapoptotic translocation of calreticulin (CRT) to the cell surface. Blockade or knockdown of CRT suppressed the phagocytosis of anthracyclin-treated tumor cells by dendritic cells and abolished their immunogenicity in mice. The anthracyclin-induced CRT translocation was mimicked by inhibition of the protein phosphatase 1/GADD34 complex. Administration of recombinant CRT or inhibitors of protein phosphatase 1/GADD34 restored the immunogenicity of cell death elicited by etoposide and mitomycin C, and enhanced their antitumor effects in vivo. These data identify CRT as a key feature determining anticancer immune responses and delineate a possible strategy for immunogenic chemotherapy.


Assuntos
Apoptose/imunologia , Calreticulina/imunologia , Neoplasias do Colo/metabolismo , Animais , Antraciclinas/farmacologia , Antraciclinas/uso terapêutico , Antígenos de Diferenciação/metabolismo , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Calreticulina/genética , Calreticulina/metabolismo , Proteínas de Ciclo Celular/antagonistas & inibidores , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Membrana Celular/efeitos dos fármacos , Membrana Celular/imunologia , Membrana Celular/metabolismo , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/patologia , Células Dendríticas/imunologia , Eletroforese em Gel Bidimensional , Etoposídeo/farmacologia , Etoposídeo/uso terapêutico , Immunoblotting , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Mitomicina/farmacologia , Mitomicina/uso terapêutico , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/metabolismo , Fagocitose/imunologia , Proteína Fosfatase 1 , Transporte Proteico/efeitos dos fármacos , Interferência de RNA , Proteínas Recombinantes/farmacologia
13.
Biochem Pharmacol ; 72(11): 1396-404, 2006 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-16765323

RESUMO

The susceptibility of cells to apoptosis induction is deeply influenced by their position in the cell cycle. Unfortunately, however, current methods for the enrichment of cells in defined phases of the cell cycle are mostly based on the synchronization of cells by agents or conditions that are intrinsically toxic and induce apoptosis on their own. We developed a novel procedure for the purification of cells in distinct phases of the cell cycle. This method is based on the stable transfection of cells with a chimeric protein made up by histone H2B and green fluorescent protein (GFP). Cytofluorometric purification of cells defined by their size and their H2B-GFP-dependent fluorescence (which reflects chromatin and hence DNA content) allowed for the efficient separation of diploid and tetraploid cells in the fluorescence-activated cell sorter (FACS). Moreover, when applied to diploid cells, this method allowed for the enrichment of live, functional cells in the G1, S and G2 phases of the cell cycle. FACS-purified cells were viable and readily resumed the cell cycle upon reculture. While staurosporine was equally toxic for cells in any phase of the cell cycle, camptothecin was particularly toxic for cells in the S phase. Moreover, BAY11-7082, a specific inhibitor of the IKK complex required for NF-kappaB activation, exhibited a particular cell cycle-specific profile of toxicity (G2>S>G1). These results delineate a novel procedure for studying the intersection between cell cycle regulation and cell death mechanisms.


Assuntos
Apoptose , Neoplasias do Colo/patologia , Citometria de Fluxo/métodos , Interfase , Apoptose/efeitos dos fármacos , Bromodesoxiuridina/metabolismo , Camptotecina/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Neoplasias do Colo/genética , Neoplasias do Colo/metabolismo , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Imunofluorescência , Humanos , Interfase/efeitos dos fármacos , Nitrilas/farmacologia , Ploidias , Estaurosporina/farmacologia , Sulfonas/farmacologia
14.
Nat Med ; 12(2): 214-9, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16444265

RESUMO

The interferon (IFN)-gamma-induced TRAIL effector mechanism is a vital component of cancer immunosurveillance by natural killer (NK) cells in mice. Here we show that the main source of IFN-gamma is not the conventional NK cell but a subset of B220(+)Ly6C(-) dendritic cells, which are atypical insofar as they express NK cell-surface molecules. Upon contact with a variety of tumor cells that are poorly recognized by NK cells, B220(+)NK1.1(+) dendritic cells secrete high levels of IFN-gamma and mediate TRAIL-dependent lysis of tumor cells. Adoptive transfer of these IFN-producing killer dendritic cells (IKDCs) into tumor-bearing Rag2(-/-)Il2rg(-/-) mice prevented tumor outgrowth, whereas transfer of conventional NK cells did not. In conclusion, we identified IKDCs as pivotal sensors and effectors of the innate antitumor immune response.


Assuntos
Células Dendríticas/classificação , Células Dendríticas/imunologia , Neoplasias Experimentais/imunologia , Transferência Adotiva , Animais , Apresentação de Antígeno , Antígenos Ly , Antígenos de Superfície/metabolismo , Proteínas Reguladoras de Apoptose/imunologia , Antígeno CD11c/metabolismo , Linhagem Celular Tumoral , Citotoxicidade Imunológica , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Células Dendríticas/ultraestrutura , Feminino , Interferon gama/biossíntese , Subunidade gama Comum de Receptores de Interleucina , Células Matadoras Naturais/imunologia , Lectinas Tipo C/metabolismo , Antígenos Comuns de Leucócito/metabolismo , Glicoproteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Nus , Microscopia Eletrônica , Subfamília B de Receptores Semelhantes a Lectina de Células NK , Receptores de Interleucina/deficiência , Receptores de Interleucina/genética , Ligante Indutor de Apoptose Relacionado a TNF , Fator de Necrose Tumoral alfa/imunologia
15.
J Exp Med ; 202(12): 1691-701, 2005 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-16365148

RESUMO

Systemic anticancer chemotherapy is immunosuppressive and mostly induces nonimmunogenic tumor cell death. Here, we show that even in the absence of any adjuvant, tumor cells dying in response to anthracyclins can elicit an effective antitumor immune response that suppresses the growth of inoculated tumors or leads to the regression of established neoplasia. Although both antracyclins and mitomycin C induced apoptosis with caspase activation, only anthracyclin-induced immunogenic cell death was immunogenic. Caspase inhibition by Z-VAD-fmk or transfection with the baculovirus inhibitor p35 did not inhibit doxorubicin (DX)-induced cell death, yet suppressed the immunogenicity of dying tumor cells in several rodent models of neoplasia. Depletion of dendritic cells (DCs) or CD8+T cells abolished the immune response against DX-treated apoptotic tumor cells in vivo. Caspase inhibition suppressed the capacity of DX-killed cells to be phagocytosed by DCs, yet had no effect on their capacity to elicit DC maturation. Freshly excised tumors became immunogenic upon DX treatment in vitro, and intratumoral inoculation of DX could trigger the regression of established tumors in immunocompetent mice. These results delineate a procedure for the generation of cancer vaccines and the stimulation of anti-neoplastic immune responses in vivo.


Assuntos
Antibióticos Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Caspases/metabolismo , Doxorrubicina/farmacologia , Mitomicina/farmacologia , Neoplasias/tratamento farmacológico , Neoplasias/imunologia , Clorometilcetonas de Aminoácidos/farmacologia , Animais , Antibióticos Antineoplásicos/uso terapêutico , Linfócitos T CD8-Positivos/imunologia , Inibidores de Caspase , Linhagem Celular Tumoral , Células Dendríticas/imunologia , Doxorrubicina/uso terapêutico , Immunoblotting , Marcação In Situ das Extremidades Cortadas , Camundongos , Mitomicina/uso terapêutico , Neoplasias/prevenção & controle , Ratos , Vacinação/métodos , Proteínas Virais/genética , Proteínas Virais/farmacologia
16.
Mol Cell Biol ; 25(3): 1025-40, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15657430

RESUMO

Mammalian cells were observed to die under conditions in which nutrients were depleted and, simultaneously, macroautophagy was inhibited either genetically (by a small interfering RNA targeting Atg5, Atg6/Beclin 1-1, Atg10, or Atg12) or pharmacologically (by 3-methyladenine, hydroxychloroquine, bafilomycin A1, or monensin). Cell death occurred through apoptosis (type 1 cell death), since it was reduced by stabilization of mitochondrial membranes (with Bcl-2 or vMIA, a cytomegalovirus-derived gene) or by caspase inhibition. Under conditions in which the fusion between lysosomes and autophagosomes was inhibited, the formation of autophagic vacuoles was enhanced at a preapoptotic stage, as indicated by accumulation of LC3-II protein, ultrastructural studies, and an increase in the acidic vacuolar compartment. Cells exhibiting a morphology reminiscent of (autophagic) type 2 cell death, however, recovered, and only cells with a disrupted mitochondrial transmembrane potential were beyond the point of no return and inexorably died even under optimal culture conditions. All together, these data indicate that autophagy may be cytoprotective, at least under conditions of nutrient depletion, and point to an important cross talk between type 1 and type 2 cell death pathways.


Assuntos
Adenina/análogos & derivados , Apoptose/fisiologia , Autofagia/fisiologia , Lisossomos/metabolismo , Mitocôndrias/metabolismo , Fagossomos/metabolismo , Adenina/toxicidade , Animais , Apoptose/efeitos dos fármacos , Proteínas Reguladoras de Apoptose , Autofagia/efeitos dos fármacos , Proteína 5 Relacionada à Autofagia , Proteína Beclina-1 , Inibidores de Caspase , Caspases/metabolismo , Células Cultivadas , Inibidores Enzimáticos/toxicidade , Células HeLa , Humanos , Proteínas Imediatamente Precoces/metabolismo , Camundongos , Microscopia Eletrônica de Transmissão , Proteínas Associadas aos Microtúbulos/metabolismo , Monensin/toxicidade , Proteínas/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , RNA Interferente Pequeno/metabolismo , Proteínas Virais/metabolismo
17.
Oncogene ; 24(5): 737-45, 2005 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-15580299

RESUMO

Two cell lines that exemplify erythropoietin (EPO) receptor-positive tumors, human renal carcinoma cell lines RCC and the myelomonocytic leukemia cell line U937, were investigated for the apoptosis-modulatory potential of EPO. Cells cultured in the presence of EPO exhibited an elevated apoptotic response to cancer chemotherapeutic agents such as daunorubicin (Dauno) and vinblastine (VBL). Chemosensitization by EPO did not involve an increase in p53 activation, yet correlated with enhanced Bax/Bak-dependent mitochondrial membrane perturbation and caspase maturation. In vitro monotherapy with Dauno or VBL induced the degradation of IkappaBalpha, provoked the translocation of NF-kappaB p65/50 to the nucleus and stimulated the expression of an NF-kappaB-activatable reporter gene. All these signs of NF-kappaB activation were perturbed in the presence of EPO. Inhibition of JAK2, one of the receptor-proximal elements of EPO-mediated signal transduction, greatly diminished the EPO-mediated chemosensitization and NF-kappaB inhibition. EPO lost its death-facilitating effects in the presence of an NF-kappaB inhibitor, underscoring the cause-effect relationship between EPO-mediated chemosensitization and NF-kappaB inhibition. Altogether, these results suggest that, at least in a specific subset of tumors, EPO receptor agonists can prevent activation of the NF-kappaB pathway, thereby enhancing the propensity of EPO receptor-positive tumor cells to undergo apoptosis.


Assuntos
Eritropoetina/farmacologia , NF-kappa B/antagonistas & inibidores , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Carcinoma de Células Renais , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Daunorrubicina/farmacologia , Humanos , Janus Quinase 2 , Neoplasias Renais , Transporte Proteico/efeitos dos fármacos , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Receptores da Eritropoetina/efeitos dos fármacos , Receptores da Eritropoetina/fisiologia , Transdução de Sinais/efeitos dos fármacos , Células U937 , Vimblastina/farmacologia
18.
J Cell Sci ; 117(Pt 23): 5643-53, 2004 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-15494371

RESUMO

Cells expressing the human immunodeficiency virus (HIV-1) envelope glycoprotein complex (Env) can fuse with CD4+ cells. When the apoptotic pathway is initiated in Env+ cells ('donor cells'), co-culture with a healthy CD4+ fusion partner ('acceptor cells') results in apoptosis of the syncytium and thus is 'contagious'. The cell-to-cell transmission of the lethal signal was only observed when the nuclei from donor cells exhibited pre-apoptotic chromatin condensation (PACC), correlating with comet assay-detectable DNA strand breaks, which precede caspase activation, as well as the loss of the mitochondrial transmembrane potential. Transmission of the lethal signal resulted into mitochondrial alterations, and caspase-dependent nuclear pyknosis with chromatinolysis affecting both the donor and the acceptor nuclei. In the presence of caspase inhibitors, all nuclei of the syncytium formed by fusion of the pre-apoptotic and the healthy cell manifested PACC, exhibited DNA lesions and lost transcriptional activity. Transmission of the lethal signal did not require donor cells to contain a nucleus or mitochondrial DNA, yet was inhibited when two mitochondrion-stabilizing proteins, Bcl-2 or vMIA, were overexpressed. Contagious apoptosis could be induced in primary human T cells, as well as in vivo, in T cells exposed to dying Env-expressing cells. Altogether, these data point to a novel mechanism through which HIV-1 can induce bystander killing.


Assuntos
Apoptose/imunologia , Antígenos CD4/metabolismo , Produtos do Gene env/fisiologia , HIV-1 , Transdução de Sinais/imunologia , Apoptose/efeitos dos fármacos , Efeito Espectador/efeitos dos fármacos , Efeito Espectador/fisiologia , Antígenos CD4/genética , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/fisiologia , Caspases/metabolismo , Núcleo Celular/metabolismo , Núcleo Celular/patologia , Técnicas de Cocultura , DNA Mitocondrial/fisiologia , Inibidores Enzimáticos/toxicidade , Produtos do Gene env/genética , Produtos do Gene env/metabolismo , Inativação Gênica , Genes Reporter , Células Gigantes/patologia , Proteína gp41 do Envelope de HIV/farmacologia , Células HeLa , Humanos , Células Jurkat , Fragmentos de Peptídeos/farmacologia , Fragmentos de Peptídeos/toxicidade , Proteínas Proto-Oncogênicas c-bcl-2/genética , Transfecção
19.
Oncogene ; 23(49): 8049-64, 2004 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-15377997

RESUMO

The mitochondrial permeability transition pore complex (PTPC) is involved in the control of the mitochondrial membrane permeabilization during apoptosis, necrosis and autophagy. Indeed, the adenine nucleotide translocator (ANT) and the voltage-dependent anion channel (VDAC), two major components of PTPC, are the targets of a variety of proapoptotic inducers. Using co-immunoprecipitation and proteomic analysis, we identified some of the interacting partners of ANT in several normal tissues and human cancer cell lines. During chemotherapy-induced apoptosis, some of these interactions were constant (e.g. ANT-VDAC), whereas others changed strongly concomitantly with the dissipation of the mitochondrial transmembrane potential and until nuclear degradation occurred (e.g. Bax, Bcl-2, subunits of the respiratory chain, a subunit of the phosphatase PP2A, phospholipase PLC beta 4 and IP3 receptor). In addition, a glutathione-S-transferase (GST) interacts with ANT in normal tissue, in colon carcinoma cells and in vitro. This interaction is lost during apoptosis induction, suggesting that GST behaves as an endogenous repressor of PTPC and ANT pore opening. Thus, ANT is connected to mitochondrial proteins as well as to proteins from other organelles such as the endoplasmic reticulum forming a dynamic polyprotein complex. Changes within this ANT interactome coordinate the lethal response of cells to apoptosis induction.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Canais Iônicos/fisiologia , Translocases Mitocondriais de ADP e ATP/fisiologia , Animais , Linhagem Celular Tumoral , Glutationa/fisiologia , Glutationa Transferase/fisiologia , Humanos , Cinética , Proteínas de Transporte da Membrana Mitocondrial , Poro de Transição de Permeabilidade Mitocondrial , Proteômica , Proteínas Proto-Oncogênicas c-bcl-2/fisiologia , Ratos , Proteína X Associada a bcl-2
20.
J Cell Sci ; 117(Pt 19): 4461-8, 2004 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-15316071

RESUMO

Stress granules (SG) are dynamic cytoplasmic foci in which stalled translation initiation complexes accumulate. In conditions of acute cellular redox, stress cells manipulated to lose the expression of apoptosis-inducing factor (AIF) nucleate SG signature proteins (e.g. TIA-1, PABP1) more efficiently than AIF-positive controls. AIF also inhibited SG formation induced by the RasGAP-associated endoribonuclease G3BP. Retransfection of mouse AIF into cells subjected to human AIF-specific siRNA revealed that only AIF imported into mitochondria could repress SGs and that redox-active domains of AIF, which are dispensable for its apoptogenic action, were required for SG inhibition. In response to oxidative stress, AIF-negative cells were found to deplete non-oxidized glutathione more rapidly than AIF-expressing cells. Exogenous supplementation of glutathione inhibited SG formation elicited by arsenate or G3BP. Together, these data suggest that the oxidoreductase function of AIF is required for the maintenance of glutathione levels in stress conditions and that glutathione is a major regulator of SG.


Assuntos
Apoptose/fisiologia , Proteínas de Transporte/metabolismo , Flavoproteínas/metabolismo , Glutationa/análogos & derivados , Proteínas de Membrana/metabolismo , Organelas/metabolismo , Transporte Proteico/fisiologia , Acetilcisteína/toxicidade , Apoptose/efeitos dos fármacos , Fator de Indução de Apoptose , Arseniatos/toxicidade , DNA Helicases , Glutationa/metabolismo , Glutationa/toxicidade , Células HeLa , Humanos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Organelas/efeitos dos fármacos , Oxirredução/efeitos dos fármacos , Proteínas de Ligação a Poli-ADP-Ribose , Transporte Proteico/efeitos dos fármacos , RNA Helicases , Proteínas com Motivo de Reconhecimento de RNA , RNA Interferente Pequeno/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA