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










Base de dados
Intervalo de ano de publicação
1.
J Immunother ; 34(3): 229-35, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21389875

RESUMO

LL-37 is a human cationic host defense peptide (antimicrobial peptide) belonging to the cathelicidin family of peptides. In this study, LL-37 was shown to kill stimulated and nonstimulated CD4(+)CD25(+)FoxP3(+) T cells (regulatory T cells; Tregs) through apoptosis, while having no cytotoxic effect on CD4(+)CD25(-) T cells at the same LL-37 concentrations. Of interest, Tregs were much more sensitive to LL-37 than many other cells, dying at 10-fold lower concentrations than other cell types tested. LL-37 exposure resulted in DNA fragmentation, chromatin condensation, and apoptotic body formation, all indicative of an apoptotic form of cell death. The importance of granzyme family members in the apoptosis of Tregs after LL-37 treatment was analyzed by using C57Bl/6 lymphocytes obtained from mice that were homozygous for null mutations in the granzyme B gene, and both the granzyme A and B genes. Granzyme A and granzyme B were both shown to play a role in LL-37-induced apoptosis of Tregs. Further analysis showed that apoptosis occurred primarily through caspase-dependent apoptosis at high LL-37 concentrations. However, grA-dependent/caspase-independent cell death was also observed. This suggests that LL-37 induces apoptosis in Tregs through multiple different mechanisms, initiated by the LL-37-induced leakage of granzymes from cytolytic granules. Our results imply that LL-37 administered at the site of a tumor could influence the adaptive antitumor immune response by killing Tregs and thus inhibiting their suppressor activity.


Assuntos
Peptídeos Catiônicos Antimicrobianos , Apoptose/efeitos dos fármacos , Caspases/metabolismo , Fragmentação do DNA/efeitos dos fármacos , Granzimas/deficiência , Linfócitos T Reguladores/efeitos dos fármacos , Imunidade Adaptativa/efeitos dos fármacos , Sequência de Aminoácidos , Animais , Peptídeos Catiônicos Antimicrobianos/síntese química , Peptídeos Catiônicos Antimicrobianos/farmacologia , Peptídeos Catiônicos Antimicrobianos/uso terapêutico , Apoptose/genética , Apoptose/imunologia , Caspases/genética , Cromatina , Granzimas/genética , Homozigoto , Humanos , Ativação Linfocitária , Linfócitos Nulos/citologia , Linfócitos Nulos/efeitos dos fármacos , Linfócitos Nulos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Dados de Sequência Molecular , Neoplasias/tratamento farmacológico , Neoplasias/imunologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/imunologia , Catelicidinas
2.
Exp Cell Res ; 317(4): 531-8, 2011 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-21134367

RESUMO

LL-37 is a human cationic host defense peptide (antimicrobial peptide) belonging to the cathelicidin family of peptides. In this study, LL-37 was shown to kill stimulated CD8(+) T cells (Cytotoxic T lymphocytes; CTLs) via apoptosis, while having no cytotoxic effect on non-stimulated CD8(+) or CD4(+) T cells or stimulated CD4(+) T cells. Of interest, the CD8(+) cells were much more sensitive to LL-37 than many other cell types. LL-37 exposure resulted in DNA fragmentation, chromatin condensation, and the release of both granzyme A and granzyme B from intracellular granules. The importance of granzyme family members in the apoptosis of CTLs following LL-37 treatment was analyzed by using C57BL/6 lymphocytes obtained from mice that were homozygous for null mutations in the granzyme B gene, the granzyme A gene, or both granzymes A and B. Granzymes A and B were both shown to play an important role in LL-37-induced apoptosis of CTLs. Further analysis revealed that apoptosis occurred primarily through granzyme A-mediated caspase-independent apoptosis. However, caspase-dependent cell death was also observed. This suggests that LL-37 induces apoptosis in CTLs via multiple different mechanisms, initiated by the LL-37-induced leakage of granzymes from cytolytic granules. Our results imply the existence of a novel mechanism of crosstalk between the inflammatory and adaptive immune systems. Cells such as neutrophils, at the site of a tumor for example, could influence the effector, activity of CTL through the secretion of LL-37.


Assuntos
Peptídeos Catiônicos Antimicrobianos/farmacologia , Apoptose/efeitos dos fármacos , Granzimas/fisiologia , Linfócitos T Citotóxicos/efeitos dos fármacos , Imunidade Adaptativa , Animais , Inflamação , Camundongos , Camundongos Endogâmicos C57BL , Receptor Cross-Talk , Catelicidinas
3.
Exp Mol Pathol ; 88(3): 371-5, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20171209

RESUMO

Although current treatments based on the use of B-cell-specific anti-CD20 monoclonal antibodies and aggressive combinatorial chemotherapy have improved the survival of patients suffering from B-cell non-Hodgkin's lymphoma (NHL), some individuals fail to respond to treatment and relapses remain common. New and more effective treatments for B-cell NHL are therefore required. Bovine lactoferricin (LfcinB) is a cationic antimicrobial peptide that is cytotoxic for several human tumor cell lines but does not harm healthy cells. Here we show that in vitro treatment with LfcinB caused Raji and Ramos human B-lymphoma cells to die by apoptosis, as indicated by DNA fragmentation, chromatin condensation, and nuclear disintegration. LfcinB killed B-lymphoma cells more efficiently at low serum concentrations and was inhibited in the presence of exogenous bovine serum albumin, suggesting partial neutralization of cationic LfcinB by anionic serum components. LfcinB-induced apoptosis in B-lymphoma cells was caspase-independent since caspase-3 activation was not detected by Western blotting and the general caspase inhibitor z-VAD-fmk did not prevent LfcinB-induced DNA fragmentation. Importantly, immune-deficient SCID/beige mice that were inoculated intravenously with Ramos B-lymphoma cells in order to model B-cell NHL exhibited extended survival following systemic administration of LfcinB, indicating that LfcinB warrants further investigation as a novel therapeutic agent for the possible treatment of B-cell NHL.


Assuntos
Lactoferrina/farmacologia , Linfoma de Células B/tratamento farmacológico , Sequência de Aminoácidos , Animais , Peptídeos Catiônicos Antimicrobianos/química , Peptídeos Catiônicos Antimicrobianos/farmacologia , Antineoplásicos/química , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Caspase 3/metabolismo , Bovinos , Linhagem Celular Tumoral , Meios de Cultura , Humanos , Lactoferrina/química , Linfoma de Células B/enzimologia , Linfoma de Células B/patologia , Camundongos , Camundongos SCID , Dados de Sequência Molecular , Transplante de Neoplasias , Soroalbumina Bovina , Transplante Heterólogo
4.
Mol Cancer Res ; 7(5): 689-702, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19435812

RESUMO

LL-37 is a human cationic host defense peptide (antimicrobial peptide) belonging to the cathelicidin family of peptides. In this study, LL-37 was shown to kill Jurkat T leukemia cells via apoptosis. A loss of mitochondrial membrane potential, DNA fragmentation, and phosphatidylserine externalization were detected following LL-37 exposure, whereas apoptosis was independent of caspase family members. The specific apoptotic pathway induced by LL-37 was defined through the utilization of Jurkat cells modified to express antiapoptotic proteins, as well as cells deficient in various proteins associated with apoptosis. Of interest, both Bcl-2-overexpressing cells and cells deficient in Bax and Bak proteins displayed a significant reduction in LL-37-induced apoptosis. In addition, Jurkat cells modified in the Fas receptor-associated pathway showed no reduction in apoptosis when exposed to LL-37. Analysis of the involvement of apoptosis-inducing factor (AIF) in LL-37-mediated apoptosis revealed that AIF transferred from the mitochondria to the nucleus of cells exposed to LL-37, where it may lead to large-scale DNA fragmentation and chromatin condensation. AIF knockdown analysis resulted in LL-37-resistant cells. This suggests that AIF is mandatory in LL-37-mediated killing. Lastly, chelation or inhibition of Ca(2+) or calpains inhibited LL-37-mediated killing. Further analysis revealed that calpains were required for LL-37-mediated Bax translocation to mitochondria. Together, these data show that LL-37-induced apoptosis is mediated via the mitochondria-associated pathway in a caspase-independent and calpain- and AIF-dependent manner that involves Bax activation and translocation to mitochondria.


Assuntos
Peptídeos Catiônicos Antimicrobianos/farmacologia , Fator de Indução de Apoptose/metabolismo , Apoptose/efeitos dos fármacos , Calpaína/metabolismo , Proteína X Associada a bcl-2/metabolismo , Sequência de Aminoácidos , Peptídeos Catiônicos Antimicrobianos/síntese química , Fator de Indução de Apoptose/genética , Cálcio/metabolismo , Calpaína/antagonistas & inibidores , Caspase 9/metabolismo , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Inibidores de Cisteína Proteinase/farmacologia , Relação Dose-Resposta a Droga , Células HeLa , Humanos , Immunoblotting , Marcação In Situ das Extremidades Cortadas , Células Jurkat , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Modelos Biológicos , Dados de Sequência Molecular , Oligopeptídeos/farmacologia , Transporte Proteico/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , RNA Interferente Pequeno/genética , Transfecção , Catelicidinas
5.
Int J Oncol ; 32(6): 1325-33, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18497995

RESUMO

2-Chloro-2'-deoxyadenosine (CdA; cladribine) is a chemotherapeutic agent used in the treatment of certain leukemias. However, the signalling events that govern CdA-mediated cytotoxicity in leukemia cells remain unclear. We show here that CdA treatment caused Jurkat human T leukemia cells to die via apoptosis in a dose- and time-dependent fashion. Bcl-2 overexpression protected Jurkat T leukemia cells from CdA-induced apoptosis and loss of mitochondrial transmembrane potential (Delta Psi m). Furthermore, mitochondria that were isolated from Jurkat T leukemia cells and then exposed to CdA showed a loss of Delta Psi m, indicating that CdA directly compromised outer mitochondrial membrane integrity. CdA treatment of Jurkat T leukemia cells resulted in the activation of caspase-3, -8, and -9, while inhibition of these caspases prevented the CdA-induced loss of Delta Psi m, as well as DNA fragmentation. In addition, caspase-3 inhibition prevented caspase-8 activation while caspase-8 inhibition prevented caspase-9 activation. Death receptor signalling was not involved in CdA-induced apoptosis since cytotoxicity was not affected by FADD-deficiency or antibody neutralization of either Fas ligand or tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). Taken together, these data suggested that CdA-induced apoptosis in Jurkat T leukemia cells was mediated via a caspase-3-dependent mitochondrial feedback amplification loop. CdA treatment also increased p38 mitogen-activated protein (MAPK) and extracellular signal-regulated kinase 1 and 2 (ERK1/2) phosphorylation in Jurkat T leukemia cells. Although ERK1/2 inhibition did not affect CdA-mediated cytotoxicity, inhibition of p38 MAPK had an enhancing effect, which suggested a cytoprotective function for p38 MAPK. Agents that inhibit p38 MAPK might therefore increase the effectiveness of CdA-based chemotherapy.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Caspase 3/metabolismo , Cladribina/farmacologia , Retroalimentação Fisiológica , Mitocôndrias/efeitos dos fármacos , Proteína de Domínio de Morte Associada a Fas/metabolismo , Humanos , Células Jurkat/metabolismo , Células Jurkat/patologia , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mitocôndrias/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Transdução de Sinais
6.
Int J Oncol ; 32(3): 527-35, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18292929

RESUMO

The resistance of many human cancers to immune-based therapies, including adoptive immunotherapy and the administration of therapeutic cancer vaccines, has been attributed to tumor-associated immune suppression, due in part to immunosuppressive molecules located within the tumor microenvironment. Adenosine is a purine nucleoside found within the interstitial fluid of solid tumors at concentrations that are able to inhibit cell-mediated immune responses to tumor cells. It is well established that extracellular adenosine inhibits T lymphocyte activation and effector function, including T cell adhesion to tumor cells and cytotoxic activity, by signaling primarily through A2a and A3 adenosine receptors on the surface of T cells. Importantly, A2a adenosine receptor-deficient mice exhibit enhanced anti-tumor immune responses by CD8+ T cells, as well as a dramatic reduction in the growth of experimental tumors in comparison to wild-type controls. A2a adenosine receptor signaling has also been implicated in adenosine-mediated inhibition of cytokine production and cytotoxic activity by activated natural killer (NK) cells, although the process of NK cell granule exocytosis is apparently suppressed via a distinct and as yet uncharacterized adenosine receptor. In this report, we review the evidence that adenosine is a potent inhibitor of cellular immune responses and may therefore be a major barrier to the successful immunotherapy of human carcinomas. The signaling pathways through which adenosine exerts its inhibitory effects on cell-mediated immune responses are also discussed. The accumulated evidence suggests that the effectiveness of immune-based therapies for solid tumors may be enhanced by selective antagonism of the adenosine receptor subtypes through which adenosine inhibits the anti-tumor activity of T lymphocytes and NK cells.


Assuntos
Adenosina/farmacologia , Células Matadoras Naturais/efeitos dos fármacos , Linfócitos T/efeitos dos fármacos , Evasão Tumoral/efeitos dos fármacos , Adenosina/metabolismo , Adenosina/fisiologia , Adenosina Desaminase/metabolismo , Adenosina Desaminase/fisiologia , Animais , Humanos , Imunoterapia , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Ativação Linfocitária/efeitos dos fármacos , Modelos Biológicos , Neoplasias/imunologia , Neoplasias/metabolismo , Neoplasias/terapia , Receptores Purinérgicos P1/metabolismo , Receptores Purinérgicos P1/fisiologia , Linfócitos T/imunologia , Linfócitos T/metabolismo
7.
Exp Cell Res ; 313(12): 2634-50, 2007 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-17570361

RESUMO

Bovine lactoferricin (LfcinB) is a cationic antimicrobial peptide that kills Jurkat T-leukemia cells by the mitochondrial pathway of apoptosis. However, the process by which LfcinB triggers mitochondria-dependent apoptosis is not well understood. Here, we show that LfcinB-induced apoptosis in Jurkat T-leukemia cells was preceded by LfcinB binding to, and progressive permeabilization of the cell membrane. Colloidal gold electron microscopy revealed that LfcinB entered the cytoplasm of Jurkat T-leukemia cells prior to the onset of mitochondrial depolarization. LfcinB was not internalized by endocytosis because endocytosis inhibitors did not prevent LfcinB-induced cytotoxicity. Furthermore, intracellular delivery of LfcinB via fusogenic liposomes caused the death of Jurkat T-leukemia cells, as well as normal human fibroblasts. Collectively, these findings suggest that LfcinB caused damage to the cell membrane that allowed LfcinB to enter the cytoplasm of Jurkat T-leukemia cells and mediate cytotoxicity. In addition, confocal microscopy showed that intracellular LfcinB co-localized with mitochondria in Jurkat T-leukemia cells, while flow cytometry and colloidal gold electron microscopy showed that LfcinB rapidly associated with purified mitochondria. Furthermore, purified mitochondria treated with LfcinB rapidly lost transmembrane potential and released cytochrome c. We conclude that LfcinB-induced apoptosis in Jurkat T-leukemia cells resulted from cell membrane damage and the subsequent disruption of mitochondrial membranes by internalized LfcinB.


Assuntos
Apoptose/efeitos dos fármacos , Permeabilidade da Membrana Celular/efeitos dos fármacos , Lactoferrina/farmacologia , Mitocôndrias/efeitos dos fármacos , Linfócitos T/citologia , Linfócitos T/efeitos dos fármacos , Animais , Biotinilação , Bovinos , Linhagem Celular Transformada , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Citocromos c/metabolismo , Fragmentação do DNA/efeitos dos fármacos , Endocitose/efeitos dos fármacos , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Humanos , Células Jurkat , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mitocôndrias/metabolismo , Mitocôndrias/ultraestrutura , Receptores de Superfície Celular/metabolismo , Linfócitos T/ultraestrutura
8.
Am J Pathol ; 169(5): 1753-66, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17071598

RESUMO

Angiogenesis is a complex process whereby new blood vessels form from pre-existing vasculature in response to proangiogenic factors such as basic fibroblast growth factor (bFGF) and the 165-kd isoform of vascular endothelial growth factor (VEGF165). Angiogenesis inhibitors show considerable potential in the treatment of cancer because angiogenesis is necessary for tumor growth beyond a few millimeters in diameter because of the tumor's need for oxygen and nutrient supply, as well as waste removal. Bovine lactoferricin (LfcinB) is a peptide fragment of iron- and heparin-binding lactoferrin obtained from cow's milk. Here we provide in vivo and in vitro evidence that LfcinB has potent antiangiogenic activity. LfcinB strongly inhibited both bFGF- and VEGF165-induced angiogenesis in Matrigel plugs implanted in C57BL/6 mice. In addition, LfcinB inhibited the in vitro proliferation and migration of human umbilical vein endothelial cells (HUVECs) in response to bFGF or VEGF165 but was not cytotoxic to HUVECs. Rather, LfcinB complexed with heparin-like structures on the HUVEC surface that are involved in the binding of bFGF and VEGF165 to their respective receptors, thereby preventing receptor-stimulated angiogenesis. These findings suggest that LfcinB may have utility as an antiangiogenic agent for the treatment of human cancers.


Assuntos
Ligação Competitiva , Células Endoteliais/efeitos dos fármacos , Fator 2 de Crescimento de Fibroblastos/farmacologia , Heparina/metabolismo , Lactoferrina/farmacologia , Neovascularização Fisiológica/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/farmacologia , Animais , Ligação Competitiva/efeitos dos fármacos , Bovinos , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Colágeno/efeitos dos fármacos , Colágeno/metabolismo , Combinação de Medicamentos , Células Endoteliais/citologia , Humanos , Laminina/efeitos dos fármacos , Laminina/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Ligação Proteica/efeitos dos fármacos , Proteoglicanas/efeitos dos fármacos , Proteoglicanas/metabolismo
9.
Expert Opin Investig Drugs ; 15(8): 933-46, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16859395

RESUMO

Cancer treatment by conventional chemotherapy is hindered by toxic side effects and the frequent development of multi-drug resistance by cancer cells. Cationic antimicrobial peptides (CAPs) are a promising new class of natural-source drugs that may avoid the shortcomings of conventional chemotherapy because certain CAPs exhibit selective cytotoxicity against a broad spectrum of human cancer cells, including neoplastic cells that have acquired a multi-drug-resistant phenotype. Tumour cell killing by CAPs is usually by a cell membrane-lytic effect, although some CAPs can trigger apoptosis in cancer cells via mitochondrial membrane disruption. Furthermore, certain CAPs are potent inhibitors of blood vessel development (angiogenesis) that is associated with tumour progression. This article reviews the mechanisms by which CAPs exert anticancer activity and discusses the potential application of selected CAPs as therapeutic agents for the treatment of human cancers.


Assuntos
Peptídeos Catiônicos Antimicrobianos/farmacologia , Antineoplásicos/farmacologia , Membrana Celular/efeitos dos fármacos , Membranas Mitocondriais/efeitos dos fármacos , Neoplasias/tratamento farmacológico , Proteínas/farmacologia , Sequência de Aminoácidos , Animais , Peptídeos Catiônicos Antimicrobianos/química , Peptídeos Catiônicos Antimicrobianos/uso terapêutico , Antineoplásicos/síntese química , Antineoplásicos/uso terapêutico , Apoptose , Sobrevivência Celular , Defensinas/química , Defensinas/farmacologia , Defensinas/uso terapêutico , Portadores de Fármacos , Desenho de Fármacos , Humanos , Lactoferrina/química , Lactoferrina/farmacologia , Lactoferrina/uso terapêutico , Magaininas , Dados de Sequência Molecular , Neoplasias/patologia , Estrutura Secundária de Proteína , Proteínas/química , Proteínas/uso terapêutico , Proteínas de Xenopus/química , Proteínas de Xenopus/farmacologia , Proteínas de Xenopus/uso terapêutico
10.
Oncol Rep ; 15(5): 1385-90, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16596215

RESUMO

Bovine lactoferricin (LfcinB) is a cationic peptide that selectively induces caspase-dependent apoptosis in human leukemia and carcinoma cell lines. Ceramide is a second messenger in apoptosis signaling that has been shown to increase the cytotoxicity of various anti-cancer drugs. In this study, we determined whether manipulation of intracellular ceramide levels enhanced LfcinB-induced apoptosis of estrogen-nonresponsive MDA-MB-435 breast carcinoma cells. LfcinB caused DNA fragmentation and morphological changes consistent with apoptosis in MDA-MB-435 breast cancer cell cultures, but did not affect the viability of untransformed mammary epithelial cells. MDA-MB-435 breast cancer cells also exhibited DNA fragmentation and morphological changes consistent with apoptosis following exposure to the cell-permeable ceramide analog C6. An additive increase in DNA fragmentation was observed when both LfcinB and C6 ceramide were added to MDA-MB-435 breast cancer cell cultures. A greater than additive increase in DNA fragmentation was seen when LfcinB was used in combination with tamoxifen, which prevents the metabolism of endogenous ceramide to glucosylceramide by glucosylceramide synthase, as well as blocking estrogen receptor signaling. However, a selective inhibitor of glucosylceramide synthase,1-phenyl-2-palmitoylamino-3-morpholino-1-propanol, failed to further increase DNA fragmentation by LfcinB, suggesting that tamoxifen enhanced LfcinB-induced apoptosis in breast cancer cells via a mechanism that did not involve glucosylceramide synthase inhibition. We conclude that combination therapy with LfcinB and tamoxifen warrants further investigation for possible use in the treatment of breast cancer.


Assuntos
Antibacterianos/farmacologia , Antineoplásicos Hormonais/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias da Mama/tratamento farmacológico , Ceramidas/farmacologia , Lactoferrina/farmacologia , Tamoxifeno/farmacologia , Animais , Mama/efeitos dos fármacos , Mama/metabolismo , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Bovinos , Células Cultivadas , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Humanos , Receptores de Estrogênio/metabolismo
11.
Int J Oncol ; 27(6): 1717-26, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16273228

RESUMO

Paclitaxel is a microtubule-stabilizing and apoptosis-inducing drug that is commonly used to treat metastatic breast cancer, although the mechanism of paclitaxel-induced apoptosis remains incompletely understood. Furthermore, adhesion molecule expression is attenuated on mouse mastocytoma and human leukemia cells that survive short-term culture in the presence of paclitaxel. In the present study we show that MDA-MB-435 human breast carcinoma cells that survived culture for 72 h in the presence of submaximal cytotoxic concentrations of paclitaxel (0.02 and 0.01 microg/ml) showed decreased expression of the adhesion molecule ICAM-1. Paclitaxel treatment of MDA-MB-435 cells was associated with the generation of reactive oxygen species (ROS), dissipation of mitochondrial transmembrane potential, and the activation of caspase-3. The antioxidant glutathione protected MDA-MB-435 cells from paclitaxel-induced cytotoxicity and reduced ICAM-1 expression. In addition, a selective inhibitor of caspase-3 (Z-DEVD-FMK), as well as a pan-caspase inhibitor (Z-VAD-FMK), partially prevented the decrease in ICAM-1 expression observed following paclitaxel treatment, but did not protect against paclitaxel-induced cytotoxicity. We conclude that the paclitaxel-induced reduction in ICAM-1 expression by MDA-MB-435 breast carcinoma cells is both ROS- and caspase-dependent, whereas paclitaxel-induced cytotoxicity is ROS-dependent and does not involve caspases. Decreased ICAM-1 expression by breast carcinoma cells that survive paclitaxel treatment may negatively impact on cytotoxic lymphocyte-mediated destruction of paclitaxel-resistant breast cancer cells in the context of chemo-immunotherapy or chemo-adoptive immunotherapy.


Assuntos
Apoptose/efeitos dos fármacos , Caspases/metabolismo , Molécula 1 de Adesão Intercelular/metabolismo , Paclitaxel/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Clorometilcetonas de Aminoácidos/farmacologia , Antineoplásicos Fitogênicos/farmacologia , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Caspase 3 , Inibidores de Caspase , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Inibidores de Cisteína Proteinase/farmacologia , Relação Dose-Resposta a Droga , Ativação Enzimática/efeitos dos fármacos , Fibronectinas/metabolismo , Humanos , Membranas Intracelulares/efeitos dos fármacos , Membranas Intracelulares/fisiologia , Potenciais da Membrana/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/fisiologia , Oligopeptídeos/farmacologia , Fatores de Tempo
12.
Exp Mol Pathol ; 79(3): 249-58, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16168410

RESUMO

Cell death caused by the accumulation of extracellular adenosine is believed to contribute to the profound loss of T lymphocytes in patients with severe combined immunodeficiency disease due to adenosine deaminase deficiency. Although adenosine is known to trigger apoptosis in thymocytes and peripheral T cells, the molecular basis of this effect is not understood. In this study, we show that adenosine-induced apoptosis in mouse EL-4 thymoma cells was associated with the generation of reactive oxygen species and a reduction in mitochondrial transmembrane potential. In addition, cell death was by a caspase-independent mechanism because caspase inhibitors did not protect EL-4 cells from adenosine-induced cytotoxicity. Although reverse transcriptase polymerase chain reaction revealed that EL-4 cells expressed A2b and A3 adenosine receptor subtypes, blockade of A2b and A3 adenosine receptors with receptor-selective antagonists did not attenuate adenosine-induced cell death. Nevertheless, the failure of nucleoside transport inhibitors to prevent adenosine cytotoxicity suggested that adenosine was acting through a cell-surface receptor. In addition, adenosine-induced apoptosis was not due to an accumulation of intracellular cyclic adenosine monophosphate (cAMP) since neither forskolin nor 8-Br-cAMP was cytotoxic for EL-4 cells. Adenosine therefore acts through a non-classical receptor at the cell surface to trigger caspase-independent apoptosis in mouse thymoma cells.


Assuntos
Adenosina/fisiologia , Apoptose/fisiologia , Caspases/metabolismo , Receptor A2B de Adenosina/metabolismo , Receptor A3 de Adenosina/metabolismo , Linfócitos T/citologia , 8-Bromo Monofosfato de Adenosina Cíclica/farmacologia , Antagonistas do Receptor A2 de Adenosina , Antagonistas do Receptor A3 de Adenosina , Animais , Inibidores de Caspase , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Colforsina/farmacologia , AMP Cíclico/agonistas , AMP Cíclico/metabolismo , Ativação Enzimática , Potenciais da Membrana , Camundongos , Membranas Mitocondriais/fisiologia , Espécies Reativas de Oxigênio/metabolismo , Linfócitos T/metabolismo , Timoma , Neoplasias do Timo
13.
EMBO J ; 24(17): 2980-8, 2005 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-16079913

RESUMO

Biological membrane fusion is dependent on protein catalysts to mediate localized restructuring of lipid bilayers. A central theme in current models of protein-mediated membrane fusion involves the sequential refolding of complex homomeric or heteromeric protein fusion machines. The structural features of a new family of fusion-associated small transmembrane (FAST) proteins appear incompatible with existing models of membrane fusion protein function. While the FAST proteins function to induce efficient cell-cell fusion when expressed in transfected cells, it was unclear whether they function on their own to mediate membrane fusion or are dependent on cellular protein cofactors. Using proteoliposomes containing the purified p14 FAST protein of reptilian reovirus, we now show via liposome-cell and liposome-liposome fusion assays that p14 is both necessary and sufficient for membrane fusion. Stoichiometric and kinetic analyses suggest that the relative efficiency of p14-mediated membrane fusion rivals that of the more complex cellular and viral fusion proteins, making the FAST proteins the simplest known membrane fusion machines.


Assuntos
Fusão de Membrana/fisiologia , Proteínas de Membrana/metabolismo , Proteínas Virais/metabolismo , Animais , Apoptose , Células Cultivadas , Sistemas de Liberação de Medicamentos , Humanos , Lactoferrina/administração & dosagem , Lactoferrina/farmacologia , Lipossomos/química , Proteínas de Membrana/química , Proteínas de Membrana/genética , Microscopia Eletrônica de Transmissão , Peptídeos/farmacologia , Proteolipídeos/química , Reoviridae/química , Proteínas Virais/química , Proteínas Virais/genética
14.
Mol Cancer Ther ; 4(4): 612-24, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15827335

RESUMO

Bovine lactoferricin (LfcinB) is a cationic, amphipathic peptide that is cytotoxic for human and rodent cancer cells. However, the mechanism by which LfcinB causes the death of cancer cells is not well understood. Here, we show that in vitro treatment with LfcinB rapidly induced apoptosis in several different human leukemia and carcinoma cell lines as determined by DNA fragmentation assays and phosphatidylserine headgroup inversion detected by Annexin V binding to the surface of cancer cells. Importantly, LfcinB treatment did not adversely affect the viability of untransformed human lymphocytes, fibroblasts, or endothelial cells. Studies with different LfcinB-derived peptide fragments revealed that the cytotoxic activity of LfcinB resided within the amino acid sequence FKCRRWQWRM. Treatment of Jurkat T leukemia cells with LfcinB resulted in the production of reactive oxygen species followed by caspase-2-induced dissipation of mitochondrial transmembrane potential and subsequent activation of caspase-9 and caspase-3. Selective inhibitors of caspase-2 (Z-VDVAD-FMK), caspase-9 (Z-LEHD-FMK), and caspase-3 (Z-DEVD-FMK) protected both leukemia and carcinoma cells from LfcinB-induced apoptosis. Conversely, a caspase-8 inhibitor (Z-IETD-FMK) had no effect, which argued against a role for caspase-8 and was consistent with the finding that death receptors were not involved in LfcinB-induced apoptosis. Furthermore, Jurkat T leukemia cells that overexpressed Bcl-2 were less sensitive to LfcinB-induced apoptosis, which was characterized by mitochondrial swelling and the release of cytochrome c from mitochondria into the cytosolic compartment. We conclude that LfcinB kills cancer cells by triggering the mitochondrial pathway of apoptosis at least in part through the generation of reactive oxygen species.


Assuntos
Antibacterianos/farmacologia , Apoptose , Carcinoma/tratamento farmacológico , Lactoferrina/fisiologia , Leucemia/tratamento farmacológico , Animais , Anexina A5/química , Antioxidantes/farmacologia , Caspase 2 , Caspase 3 , Caspase 9 , Caspases/metabolismo , Bovinos , Linhagem Celular Tumoral , Sobrevivência Celular , Fragmentação do DNA , Relação Dose-Resposta a Droga , Células Endoteliais/citologia , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Fibroblastos/metabolismo , Humanos , Immunoblotting , Células Jurkat , Lactoferrina/metabolismo , Linfócitos/citologia , Potenciais da Membrana , Microscopia Eletrônica , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Oligopeptídeos/farmacologia , Fosfatidilserinas/química , Espécies Reativas de Oxigênio , Sais de Tetrazólio/farmacologia , Tiazóis/farmacologia , Fatores de Tempo
15.
J Cell Biochem ; 92(2): 387-99, 2004 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-15108363

RESUMO

Ryanodine receptors (RyR) are involved in regulating intracellular Ca(++) mobilization in T lymphocytes. However, the importance of RyR signaling during T cell activation has not yet been determined. In this study, we have used the RyR-selective antagonists, ruthenium red and dantrolene, to determine the effect of RyR blockade on T cell receptor-mediated activation events and cytokine-dependent T cell proliferation. Both ruthenium red and dantrolene inhibited DNA synthesis and cell division, as well as the synthesis of interleukin (IL)-2 by T lymphocytes responding to mitogenic anti-CD3 antibody. Blockade of RyR at initiation of culture or as late as 24 h after T cell receptor stimulation inhibited T cell proliferation, suggesting a requirement for sustained RyR signaling during cell cycle progression. Although flow cytometry revealed that RyR blockade had little effect on activation-induced expression of the alpha chain (CD25) of the high affinity IL-2 receptor, the inhibitory effect of RyR antagonists could not be reversed by the addition of exogenous IL-2 at initiation of culture. In addition, both ruthenium red and dantrolene had a strong inhibitory effect on IL-2-dependent proliferation of CTLL-2 T cells. These data indicate that RyR are involved in regulating IL-2 receptor signaling that drives T cell progression through the cell cycle. We conclude that RyR-associated Ca(++) signaling regulates T cell proliferation by promoting both IL-2 synthesis and IL-2-dependent cell cycle progression.


Assuntos
Complexo CD3/metabolismo , Interleucina-2/biossíntese , Receptores de Interleucina-2/metabolismo , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Transdução de Sinais , Linfócitos T/citologia , Linfócitos T/metabolismo , Animais , Anticorpos/imunologia , Anticorpos/farmacologia , Cálcio/metabolismo , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , DNA/biossíntese , Feminino , Interleucina-2/imunologia , Interleucina-2/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Rutênio Vermelho/farmacologia , Transdução de Sinais/efeitos dos fármacos , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia
16.
Oncol Rep ; 11(4): 893-7, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15010891

RESUMO

Paclitaxel (Taxol) is a drug that is commonly used in the treatment of metastatic breast cancer. In this study, we investigated the effect of prior exposure to submaximal cytotoxic concentrations (EC(25) and EC(50)) of paclitaxel on the subsequent ability of human Jurkat T lymphocytes to adhere to monolayers of MDA-MB-435 human breast carcinoma cells. Jurkat T cells that survived culture for 24 h in the presence of paclitaxel (0.1 or 3 micro g/ml) exhibited a diminished ability to adhere to MDA-MB-435 breast cancer cell monolayers. Flow cytometric analysis of paclitaxel-treated Jurkat T cells revealed reduced surface expression of alphaL, alpha4, alpha5, and beta7 integrins, but normal beta1 integrin expression. These data suggest that paclitaxel interferes with the expression of alphaLbeta2 (LFA-1), alpha4beta1 (VLA-4), alpha5beta1 (VLA-5), and alpha4beta7 (LPAM-1) adhesion molecules by surviving T cells. Blocking experiments with antibodies against alphaL, alpha4, alpha5, beta1, and beta7 integrins revealed that adhesion of Jurkat T cells to MDA-MB-435 breast cancer cell monolayers was dependent upon alphaL, alpha4, and beta7 but not alpha5 or beta1 integrins. We conclude that prior exposure to paclitaxel caused a reduction in Jurkat T cell adhesion to MDA-MB-435 breast carcinoma cells by preventing optimal alphaLbeta2 and alpha4beta7 interactions with their corresponding ligands on tumor cells. A similar effect by paclitaxel on T lymphocytes of breast cancer patients may compromise cell-mediated anti-tumor immune responses.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Neoplasias da Mama/imunologia , Carcinoma/imunologia , Integrinas/metabolismo , Paclitaxel/farmacologia , Linfócitos T/imunologia , Neoplasias da Mama/metabolismo , Carcinoma/metabolismo , Adesão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Citometria de Fluxo , Humanos , Células Jurkat , Linfócitos T/efeitos dos fármacos , Linfócitos T/metabolismo
17.
J Cell Biochem ; 89(5): 975-91, 2003 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-12874832

RESUMO

Adenosine is an immunosuppressive molecule that is associated with the microenvironment of solid tumors. Mouse T cells activated with anti-CD3 antibody in the presence of adenosine with or without coformycin (to prevent adenosine breakdown by adenosine deaminase) exhibited decreased tyrosine phosphorylation of some intracellular proteins and were inhibited in their ability to proliferate and synthesize interleukin (IL)-2. In addition, adenosine interfered with activation-induced expression of the co-stimulatory molecules CD2 and CD28. Activation-induced CD2 and CD28 expression was also diminished when T cells were activated in the presence of anti-IL-2 and anti-CD25 antibodies to neutralize IL-2 bioactivity. Collectively, these data suggest that CD2 and CD28 up-regulation following T cell activation is IL-2-dependent; and that adenosine inhibits activation-induced T cell expression of CD2 and CD28 by interfering with IL-2-dependent signaling. The inhibitory effect of adenosine on activation-induced CD2 and CD28 expression could not be attributed to cyclic AMP (cAMP) accumulation resulting from the stimulation of adenylyl cyclase-coupled adenosine receptors, even though cAMP at concentrations much higher than those generated following adenosine stimulation was inhibitory for both CD2 and CD28 expression. We conclude that adenosine interferes with IL-2-dependent T cell expression of co-stimulatory molecules via a mechanism that does not involve the accumulation of intracellular cAMP.


Assuntos
Adenosina/análogos & derivados , Adenosina/farmacologia , Antígenos CD2/biossíntese , Antígenos CD28/biossíntese , Cafeína/análogos & derivados , AMP Cíclico/fisiologia , Interleucina-2/fisiologia , Linfócitos T/metabolismo , Adenosina/antagonistas & inibidores , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacologia , Complexo CD3/imunologia , Cafeína/farmacologia , Ciclo Celular/efeitos dos fármacos , Colforsina/farmacologia , AMP Cíclico/metabolismo , Feminino , Flavinas/farmacologia , Interleucina-2/imunologia , Interleucina-2/metabolismo , Ativação Linfocitária/efeitos dos fármacos , Ativação Linfocitária/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Fenetilaminas/farmacologia , Receptores de Interleucina-2/imunologia , Receptores Purinérgicos P1/efeitos dos fármacos , Receptores Purinérgicos P1/metabolismo , Transdução de Sinais/fisiologia , Linfócitos T/citologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Tirosina/metabolismo
18.
J Immunol ; 171(1): 69-77, 2003 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-12816984

RESUMO

Cross-linking of the GPI-anchored protein Thy-1 results in T cell proliferation and IL-2 synthesis. However, the exact function of Thy-1 in the process of T cell activation remains unknown, as does the effect of costimulation on Thy-1-driven T cell responses. In this study, we have investigated the ability of Thy-1 to substitute for traditional signal 1 in the context of costimulation provided by dendritic cells. Dendritic cells dramatically enhanced T cell proliferation and IL-2 synthesis in response to Thy-1 triggering by anti-Thy-1 mAb. This effect was not dependent on dendritic cell Fcgamma receptors, but was a result of B7-mediated costimulation (signal 2). T cells were also activated when microbeads coated with a combination of anti-Thy-1 and anti-CD28 mAbs were used to supply signals 1 and 2, respectively. Thy-1-stimulated T cells adhere to target cells and express perforin, granzyme B, and Fas ligand, but fail to kill target cells due to an inability to reorganize their secretion machinery. Moreover, in contrast to TCR signaling, Thy-1 triggering failed to induce cytotoxicity in redirected lysis assays. We conclude that Thy-1 triggering can partially substitute for signal 1, which, in combination with a strong signal 2, leads to robust T cell proliferation, IL-2 synthesis, and cytotoxic effector molecule expression, but does not induce cytolytic function. The block at the level of cytotoxic effector function that results when T cells are activated in the absence of a classical, Ag-specific signal 1 may constitute a mechanism to ensure the specificity of CTL responses and prevent potentially harmful promiscuous cytotoxicity.


Assuntos
Citotoxicidade Imunológica , Células Dendríticas/imunologia , Interleucina-2/biossíntese , Ativação Linfocitária , Transdução de Sinais/imunologia , Linfócitos T Citotóxicos/imunologia , Antígenos Thy-1/fisiologia , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/farmacologia , Antígenos CD28/fisiologia , Testes Imunológicos de Citotoxicidade/métodos , Citotoxicidade Imunológica/genética , Células Dendríticas/metabolismo , Ativação Linfocitária/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Camundongos Transgênicos , Dados de Sequência Molecular , Complexo Receptor-CD3 de Antígeno de Linfócitos T/genética , Complexo Receptor-CD3 de Antígeno de Linfócitos T/fisiologia , Receptores de IgG/deficiência , Receptores de IgG/genética , Receptores de IgG/fisiologia , Transdução de Sinais/genética , Linfócitos T Citotóxicos/metabolismo , Antígenos Thy-1/imunologia , Células Tumorais Cultivadas , Regulação para Cima/genética , Regulação para Cima/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...