Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
1.
Oncogene ; 36(47): 6617-6626, 2017 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-28783166

RESUMO

Dysregulation of microRNAs (miRNAs) plays an important role in the pathogenesis of chronic lymphocytic leukemia (CLL). The Eµ-TCL1 transgenic mouse develops a form of leukemia that is similar to the aggressive type of human B-CLL, and this valuable model has been widely used for testing novel therapeutic approaches. Here, we adopted this model to investigate the potential effects of miR-26a, miR-130an and antimiR-155 in CLL therapy. Improved delivery of miRNA molecules into CLL cells was obtained by developing a novel system based on lipid nanoparticles conjugated with an anti-CD38 monoclonal antibody. This methodology has proven to be highly effective in delivering miRNA molecules into leukemic cells. Short- and long-term experiments showed that miR-26a, miR-130a and anti-miR-155 increased apoptosis after in vitro and in vivo treatment. Of this miRNA panel, miR-26a was the most effective in reducing leukemic cell expansion. Following long-term treatment, apoptosis was readily detectable by analyzing cleavage of PARP and caspase-7. These effects could be directly attributed to miR-26a, as confirmed by significant downregulation of its proven targets, namely cyclin-dependent kinase 6 and Mcl1. The results of this study are relevant to two distinct areas. The first is related to the design of a technical strategy and to the selection of CD38 as a molecular target on CLL cells, both consenting efficient and specific intracellular transfer of miRNA. The original scientific finding inferred from the above approach is that miR-26a can elicit in vivo anti-leukemic activities mediated by increased apoptosis.


Assuntos
ADP-Ribosil Ciclase 1/antagonistas & inibidores , Apoptose/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica , Leucemia Linfocítica Crônica de Células B/tratamento farmacológico , Glicoproteínas de Membrana/antagonistas & inibidores , MicroRNAs/uso terapêutico , ADP-Ribosil Ciclase 1/genética , Animais , Anticorpos Monoclonais Murinos/química , Caspase 7/metabolismo , Linhagem Celular Tumoral , Quinase 6 Dependente de Ciclina/genética , Regulação para Baixo , Sistemas de Liberação de Medicamentos , Feminino , Humanos , Leucemia Linfocítica Crônica de Células B/genética , Lipídeos/química , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Transgênicos , MicroRNAs/administração & dosagem , MicroRNAs/antagonistas & inibidores , Proteína de Sequência 1 de Leucemia de Células Mieloides/genética , Nanopartículas/química , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/genética , Poli(ADP-Ribose) Polimerases/metabolismo , Proteínas Proto-Oncogênicas/genética
2.
Phlebology ; 30(9): 644-7, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24972760

RESUMO

OBJECTIVES: To study the ultrastructure of intraluminal defects found in the internal jugular vein by using a scanning electron microscopy. METHODS: Using a scanning electron microscopy, intraluminal septa and/or defective valves blocking the flow in the distal internal jugular vein of seven patients were studied together with the adjacent wall and compared with control specimen. RESULTS: The internal jugular veins' wall showed a significant derangement of the endothelial layer as compared to controls. Surprisingly, no endothelial cells were found in the defective cusps, and the surface of the structure is covered by a fibro-reticular lamina. CONCLUSIONS: Although the lack of endothelial cells in the internal jugular vein intraluminal obstacles is a further abnormality found in course of chronic cerebrospinal venous insufficiency, our investigation cannot clarify whether this finding is primary or caused by progressive loss of endothelium in relation to altered haemodynamic forces and/or to a past post-thrombotic/inflammatory remodelling.


Assuntos
Veias Jugulares/ultraestrutura , Insuficiência Venosa/fisiopatologia , Válvulas Venosas/diagnóstico por imagem , Adulto , Idoso , Doença Crônica , Feminino , Voluntários Saudáveis , Hemodinâmica , Humanos , Inflamação , Masculino , Microscopia Eletrônica de Varredura , Pessoa de Meia-Idade , Esclerose Múltipla/complicações , Perfusão , Tomografia Computadorizada de Emissão de Fóton Único , Ultrassonografia Doppler em Cores , Insuficiência Venosa/complicações
3.
Curr Med Chem ; 20(21): 2712-22, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23627936

RESUMO

The small molecule Nutlin-3 is a potent antagonist of the murine double minute 2 (MDM2)/p53 interaction exhibiting promising therapeutic anti-cancer activity. Nutlin-3 has been proposed as an anti-neoplastic agent for the treatment of onco-hematological diseases characterized by a lower incidence of p53 mutation with respect to solid tumors. Indeed, based on its selective non-genotoxic p53 activation, Nutlin-3 might represent an alternative to the current cytotoxic chemotherapy. To overcome the poor bioavailability of Nutlin-3, we have assessed the potential efficacy of Nutlin-3 loaded poly(lactide-co-glycolide) (PLGA) nanoparticles (NP) against hematological malignancies. To test the specificity of the anti-leukemic activity, we have used leukemic cell lines characterized by different p53 status (JVM-2 and BJAB). NP loaded with Nutlin-3 (NP-Nutlin) were rapidly taken up by the leukemic cells and were as effective as native Nutlin-3 in promoting both induction of apoptosis and cell cycle arrest in p53(wild-type) JVM-2 cells, but not in p53(mutated) BJAB cells. Moreover, injection of NP-Nutlin, but not of free Nutlin-3, in a JVM-2-derived xenograft mouse model, reduced the subcutaneous tumor volume and promoted induction of apoptosis in the tumor mass. Overall, the chemical and structural characteristics of the NP-Nutlin-3, as well as their biological activity in vitro and in vivo, made them promising for further preclinical evaluations as potentially useful anti-leukemic carriers.


Assuntos
Antineoplásicos/farmacologia , Imidazóis/farmacologia , Leucemia/genética , Leucemia/patologia , Nanopartículas/química , Piperazinas/farmacologia , Poliglactina 910/química , Proteína Supressora de Tumor p53/genética , Animais , Antineoplásicos/química , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Humanos , Imidazóis/química , Leucemia/tratamento farmacológico , Piperazinas/química , Proteína Supressora de Tumor p53/metabolismo
4.
Curr Med Chem ; 20(17): 2226-36, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23458617

RESUMO

Since the discovery of p53 as "guardian of the genome", a large number of efforts have been put in place in order to find molecular strategies aiming to restore p53 wild-type functions, particularly in the light of the fact that its pathway results ineffective in most tumors even though they have non-mutated p53. In this context, pediatric cancers, that are mostly p53 wild-type at the time of diagnosis, represent an ideal target for such therapeutic approach. Within the several mechanisms and proteins ruling p53 activity, the murine double minute 2 (MDM2) is its crucial negative regulator, frequently found overexpressed in p53-wild-type tumors. The development of new technologies such as nuclear magnetic resonance structure analyses, computational structure-based design studies, and library peptides screening have recently led to the discovery and characterization of a large number of compounds belonging to different chemical families that are able to target the interaction p53-MDM2, rescuing the p53 wild-type pathway with an overall pro-apoptotic and anticancer activity. Within the preclinical assessment of these molecules, the cis-imidazoline analogue Nutlin-3 has definitely attracted great interest for its in vitro and in vivo antitumor activity in several pediatric cancer models, either as single agent on in combination with standard chemotherapy. In this light, the aim of this review is to summarize the main preclinical evidences of the potential of MDM2 inhibitors for the treatment of childhood cancers and the key suggestions coming from their assessment in the treatment of adult cancers as proof of concept for future pediatric clinical studies.


Assuntos
Neoplasias/tratamento farmacológico , Proteínas Proto-Oncogênicas c-mdm2/antagonistas & inibidores , Criança , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/uso terapêutico , Humanos , Imidazóis/química , Imidazóis/farmacologia , Imidazóis/uso terapêutico , Piperazinas/química , Piperazinas/farmacologia , Piperazinas/uso terapêutico , Ligação Proteica/efeitos dos fármacos , Domínios e Motivos de Interação entre Proteínas/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia , Ligante Indutor de Apoptose Relacionado a TNF/uso terapêutico , Proteína Supressora de Tumor p53/metabolismo
6.
Curr Med Chem ; 19(28): 4875-84, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22934770

RESUMO

The bi-aryl urea multi-kinase inhibitor Sorafenib (BAY 43-9006, Nexavar) was initially approved for the treatment of unresectable hepatocellular carcinoma and advanced renal cell carcinoma. Eleven years after its first description in PubMed, the therapeutic potential of Sorafenib has been evaluated in an increasing number of studies, mainly focused on solid tumors. More recently, the potential usefullness of Sorafenib has started to emerge also against hematological malignancies. At the molecular level, besides the RAF kinase pathway, which represents the first therapeutic target of Sorafenib, additional kinases, in particular the vascular endothelial growth factor receptor, have been identified as important targets of Sorafenib. A great interest for the potential use of Sorafenib against acute myeloid leukemia (AML) arose when it was demonstrated that a specific mutation of a kinase gene, called FMS-like tyrosin-kinase-3- internal tandem duplication (FLT-3-ITD) and occurring in more than 30% of AML, represents a molecular target of Sorafenib. However, recent phase I and II clinical studies showed that, in spite of its ability to suppress the activity of this mutated kinase, resistence to Sorafenib rapidly occurs in AML, suggesting that Sorafenib will be more effective in combined therapy than used as single drug. Another critical molecular target of Sorafenib is the anti-apoptotic protein Mcl-1. The ability of Sorafenib to rapidly shut-off Mcl-1 in virtually all the hematological malignancies investigated, including the B-chronic lymphocytic leukemia, represents a key element for its antileukemic activity as well as for therapeutic combinations based on Sorafenib. In this respect, it is of particular interest that many chemotherapeutic drugs or innovative anti-neoplastic compounds, such as recombinant TRAIL or inibitors of MDM2 protein, are either unable to down-regulate Mcl-1 or in some instances promote a paradoxical induction of Mcl-1. In this review, the growing evidences for the role of Mcl-1 in mediating the anti-leukemic activity of Sorafenib will be discussed in relationship with promising therapeutic perspectives.


Assuntos
Neoplasias Hematológicas/tratamento farmacológico , Niacinamida/análogos & derivados , Compostos de Fenilureia/uso terapêutico , Apoptose/efeitos dos fármacos , Ensaios Clínicos como Assunto , Humanos , Leucemia Mieloide Aguda/tratamento farmacológico , Proteína de Sequência 1 de Leucemia de Células Mieloides , Niacinamida/química , Niacinamida/uso terapêutico , Niacinamida/toxicidade , Compostos de Fenilureia/química , Compostos de Fenilureia/toxicidade , Proteínas Proto-Oncogênicas c-bcl-2/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteínas Proto-Oncogênicas c-mdm2/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Receptores de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Receptores de Fatores de Crescimento do Endotélio Vascular/metabolismo , Sorafenibe , Ligante Indutor de Apoptose Relacionado a TNF/genética , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/uso terapêutico , Tirosina Quinase 3 Semelhante a fms/genética , Tirosina Quinase 3 Semelhante a fms/metabolismo
10.
Curr Cancer Drug Targets ; 9(4): 510-8, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19519319

RESUMO

By analyzing the cDNA obtained from 16 B-cell chronic lymphocytic leukemia (B-CLL) patient samples, we found that Nutlin-3, a small molecule inhibitor of MDM2/p53 interaction, induced a characteristic gene expression profile (GEP) signature in 13 out of 16 B-CLL samples. The lack of Nutlin-3-induced GEP signature in 3 out of 16 B-CLL samples was not due to p53 deletion and/or mutation, as demonstrated by FISH analysis and p53 sequencing. Of note, the 3 B-CLL samples in which Nutlin-3 did not elicit the GEP signature were also less susceptible to Nutlin-3-mediated cytotoxicity with respect to the remaining 13 B-CLL samples. However, the partial lack of response in these p53 wild-type B-CLL samples was not due to defects in the ability of Nutlin-3 to promote p53 induction, as confirmed by the rapid accumulation of p53 protein at Western blot analysis in response to Nutlin-3 in all samples examined. Upon exposure to Nutlin-3, the genes up-regulated with the highest score in the majority of B-CLL cells were all known p53-target genes, including genes involved in apoptotic pathways, such as FAS and BAX, as well as MDM2. Taken together, our data indicate that the ability of Nutlin-3 to induce a characteristic GEP signature correlates with its cytotoxic potential in p53 wild-type B-CLL cells. However, in some p53 wild-type B-CLL samples, the response to Nutlin-3 cannot be predicted on the basis of FISH analysis or p53 sequencing.


Assuntos
Regulação da Expressão Gênica/efeitos dos fármacos , Imidazóis/farmacologia , Leucemia Linfocítica Crônica de Células B/genética , Piperazinas/farmacologia , Proteína Supressora de Tumor p53/metabolismo , Morte Celular/efeitos dos fármacos , Células Cultivadas , Perfilação da Expressão Gênica , Humanos , Leucemia Linfocítica Crônica de Células B/metabolismo , Proteínas Proto-Oncogênicas c-mdm2/antagonistas & inibidores , Proteína Supressora de Tumor p53/genética
11.
Int J Immunopathol Pharmacol ; 22(1): 73-83, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19309554

RESUMO

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a promising natural anticancer therapeutic agent because through its death receptors, TRAIL-R1 and TRAIL-R2, it induces apoptosis in many transformed tumor cells, but not in the majority of normal cells. Hence, agonistic compounds directed against TRAIL death receptors have the potential of being excellent cancer therapeutic agents, with minimal cytotoxicity in normal tissues. Here, we report the selection and characterization of a new single-chain fragment variable (scFv) to TRAIL-R2 receptor isolated from a human phage-display library, produced as minibody (MB), and characterized for the in vitro anti-leukemic tumoricidal activity. The anti-TRAIL-R2 MB2.23 efficiently and specifically bound to membrane-associated TRAIL-R2 on different leukemic cell lines and could act as a direct agonist in vitro, initiating apoptotic signaling as well as complement-dependent cytotoxicity and antibody-dependent cell cytotoxicity, providing a rationale for further investigations of MB2.23 in anticancer therapy.


Assuntos
Fragmentos de Imunoglobulinas/uso terapêutico , Leucemia/tratamento farmacológico , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/agonistas , Animais , Citotoxicidade Celular Dependente de Anticorpos , Apoptose/efeitos dos fármacos , Células CHO , Cricetinae , Cricetulus , Humanos , Leucemia/patologia , Biblioteca de Peptídeos , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/imunologia , Proteínas Recombinantes/uso terapêutico
12.
Differentiation ; 77(4): 369-76, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19281785

RESUMO

Extensive apoptotic oocyte reduction occurs during fetal ovarian development. The regulatory pathways responsible for oocyte selection to programmed cell death are, however, poorly understood. The aim of this study was to investigate the potential involvement of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and its death receptors TRAIL-R1/DR4 and TRAIL-R2/DR5 and decoy receptors TRAIL-R3/DcR1 and TRAIL-R4/DcR2 in the apoptotic process characterizing human fetal and adult ovaries. For this purpose, in situ hybridization and immunohistochemistry were applied to human fetal and adult ovarian samples to study the mRNA and protein expression of TRAIL pathway components, and a human granulosa cell tumor-derived cell line (KGN) was used to elucidate functional effects of TRAIL on apoptosis. TRAIL was expressed in human fetal ovary from the 11th week until term. The pro-apoptotic TRAIL-R2/DR5 and the anti-apoptotic TRAIL-R4/DcR2 were also expressed in human ovaries throughout the fetal period. Among the different ovarian cell types, these TRAIL pathway components were mainly localized in the oocytes, and their expression increased towards term. Expression of TRAIL-R1/DR4 and TRAIL-R3/DcR1 was negligible in all of the fetal ovaries studied. Adult ovaries expressed TRAIL, TRAIL-R2/DR5, TRAIL-R3/DcR1 and TRAIL-R4/DcR2 in granulosa cells and oocytes of small primary/secondary follicles as well as in granulosa and theca cells of more developed antral follicles. In KGN cells, TRAIL efficiently induced apoptosis in a dose-dependent manner, and this was blocked by a caspase inhibitor. The results indicate a role of the TRAIL pathway components in the regulation of granulosa cell apoptosis in in vitro and suggest that these factors may have a role in regulating ovarian apoptosis also in vivo.


Assuntos
Apoptose , Células da Granulosa/metabolismo , Ovário/citologia , Ovário/fisiologia , Ligante Indutor de Apoptose Relacionado a TNF/fisiologia , Feminino , Feto/citologia , Feto/fisiologia , Regulação da Expressão Gênica , Células da Granulosa/citologia , Células da Granulosa/efeitos dos fármacos , Humanos , Imuno-Histoquímica , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia
13.
Cell Mol Life Sci ; 66(5): 841-51, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19011755

RESUMO

Osteoprotegerin (OPG) is a soluble tumor necrosis factor receptor family member, which potently inhibits RANKL-mediated osteoclastogenesis. Numerous constructs have been created for therapeutic purposes in which the heparin-binding and death homology domains of OPG were removed and the remaining peptide (amino acids 22-194) was fused to the Fc domain of human IgG1 (OPG-Fc). The administration of OPG-Fc efficiently counteracted bone loss in a variety of preclinical models of cancers. However, several in vitro studies have shown that native or recombinant full-length OPG not only neuralizes RANKL, but also the death-inducing ligand TRAIL, suggesting that OPG might potentially counteract the anti-tumor activity of TRAIL. Additional evidence suggests that full-length OPG possesses RANKL- and TRAIL-independent biological properties, mainly related to the promotion of endothelial cell survival and angiogenesis. Finally, breast tumor cells overexpressing OPG have shown increased bone metastatic potential in vivo. The relevance of these apparently conflicting findings in tumor cell biology is highlighted.


Assuntos
Neoplasias/metabolismo , Osteoprotegerina/metabolismo , Sobrevivência Celular , Células Endoteliais/citologia , Células Endoteliais/fisiologia , Humanos , Imunoglobulina G/genética , Imunoglobulina G/metabolismo , Neovascularização Fisiológica , Osteoclastos/citologia , Osteoclastos/fisiologia , Osteoprotegerina/genética , Ligante RANK/genética , Ligante RANK/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo
15.
Lupus ; 16(7): 479-82, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17670845

RESUMO

The objective of this study was to determine potential relationship between the levels of serum TNF-related apoptosis inducing ligand (TRAIL) and osteoprotegerin (OPG) and clinical markers in systemic lupus erythematosus (SLE) patients. Forty SLE patients with inactive disease were enrolled in this study. For comparison, 20 Sjögren's syndrome (SS) patients and 30 normal controls were also analysed. Serum levels of TRAIL and OPG were determined by ELISA. Serum TRAIL and OPG concentrations in SLE patients were significantly (P < 0.05) higher than those in healthy volunteers. Of note, serum TRAIL but not OPG was significantly (P < 0.05) higher in the SLE patient subset characterized by the presence of anti-SSA/SSB antibodies. The relationship between high levels of TRAIL and SSA/SSB antibodies was further supported by the analysis of SS patients characterized by SSA/SSB antibodies positivity, in which TRAIL levels resulted comparable to the subgroup of anti-SSA/SSB positive SLE patients. The presence of SSA/SSB antibodies, targeting a specific subset of SLE and SS patients, is related to increased serum levels of TRAIL but not of OPG.


Assuntos
Autoanticorpos/sangue , Autoantígenos/imunologia , Lúpus Eritematoso Sistêmico/sangue , Fragmentos de Peptídeos/imunologia , Ligante Indutor de Apoptose Relacionado a TNF/sangue , Adulto , Apoptose , Biomarcadores/sangue , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Lúpus Eritematoso Sistêmico/imunologia , Lúpus Eritematoso Sistêmico/patologia , Masculino , Pessoa de Meia-Idade , Osteoprotegerina/sangue , Estudos Prospectivos , Índice de Gravidade de Doença
16.
Cell Mol Life Sci ; 61(15): 1965-74, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15289937

RESUMO

Human and rat primary sub-cultured vascular smooth muscle cells (VSMCs) showed clear expression of the death receptors TRAIL-R1 and TRAIL-R2; however, recombinant soluble TRAIL did not induce cell death when added to these cells. TRAIL tended to protect rat VSMCs from apoptosis induced either by inflammatory cytokines tumor necrosis factor-alpha + interleukin-1beta + interferon-gamma or by prolonged serum withdrawal, and promoted a significant increase in VSMC proliferation and migration. Of note, all the biological effects induced by TRAIL were significantly inhibited by pharmacological inhibitors of the ERK pathway. Western blot analysis consistently showed that TRAIL induced a significant activation of ERK1/2, and a much weaker phosphorylation of Akt, while it did not affect the p38/MAPK pathway. Taken together, these data strengthen the notion that the TRAIL/TRAIL-R system likely plays a role in the biology of the vascular system by affecting the survival, migration and proliferation of VSMCs.


Assuntos
Movimento Celular/fisiologia , Glicoproteínas de Membrana/metabolismo , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Animais , Apoptose/fisiologia , Proteínas Reguladoras de Apoptose , Artérias/metabolismo , Divisão Celular/fisiologia , Sobrevivência Celular/fisiologia , Humanos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Músculo Liso Vascular/citologia , Ratos , Receptores do Ligante Indutor de Apoptose Relacionado a TNF , Receptores do Fator de Necrose Tumoral/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF
17.
Blood ; 98(8): 2474-81, 2001 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-11588045

RESUMO

Human herpesvirus 7 (HHV-7) is endemic in the adult human population. Although HHV-7 preferentially infects activated CD4(+) T lymphocytes, the consequence of T-cell infection for viral pathogenesis and immunity are still largely unknown. HHV-7 infection induces apoptosis mostly in uninfected bystander cells but not in productively infected CD4(+) T cells. To dissect the underlying molecular events, the role of death-inducing ligands belonging to the tumor necrosis factor (TNF) cytokine superfamily was investigated. HHV-7 selectively up-regulated the expression of TNF-related apoptosis-inducing ligand (TRAIL), but not that of CD95 ligand or TNF-alpha in lymphoblastoid (SupT1) or primary activated CD4(+) T cells. Moreover, in a cell-to-cell-contact assay, HHV-7-infected CD4(+) T lymphocytes were cytotoxic for bystander uninfected CD4(+) T cells through the TRAIL pathway. By contrast, HHV-7 infection caused a marked decrease of surface TRAIL-R1, but not of TRAIL-R2, CD95, TNF-R1, or TNF-R2. Of note, the down-regulation of TRAIL-R1 selectively occurred in cells coexpressing HHV-7 antigens that became resistant to TRAIL-mediated cytotoxicity. These findings suggest that the TRAIL-mediated induction of T-cell death may represent an important immune evasion mechanism of HHV-7, helping the virus to persist in the host organism throughout its lifetime.


Assuntos
Apoptose , Linfócitos T CD4-Positivos/imunologia , Regulação da Expressão Gênica , Herpesvirus Humano 7/imunologia , Glicoproteínas de Membrana/fisiologia , Fator de Necrose Tumoral alfa/fisiologia , Proteínas Reguladoras de Apoptose , Western Blotting , Antígenos CD4/genética , Linfócitos T CD4-Positivos/virologia , Linhagem Celular , Citotoxicidade Imunológica , Humanos , Glicoproteínas de Membrana/genética , Proteínas Recombinantes/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF , Fator de Necrose Tumoral alfa/genética , Receptor fas/imunologia
18.
Blood ; 98(7): 2220-8, 2001 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-11568010

RESUMO

Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) induced both cytotoxic (apoptosis) and cytostatic (cell cycle perturbation) effects on the human myeloid K562 cell line. TRAIL stimulated caspase 3 and nitric oxide synthase (NOS) activities, and both pathways cooperate in mediating inhibition of K562 survival/growth. This was demonstrated by the ability of z-VAD-fmk, a broad inhibitor of effector caspases, and N-nitro-L-arginine methyl ester (L-NAME), an NOS pharmacologic inhibitor, to completely (z-VAD-fmk) or partially (L-NAME) suppress the TRAIL-mediated inhibitory activity. Moreover, z-VAD-fmk was able to block TRAIL-mediated apoptosis and cell cycle abnormalities and increase of NOS activity. The addition of the NO donor sodium nitroprusside (SNP) to K562 cells reproduced the cytostatic effect of TRAIL without inducing apoptosis. When TRAIL was associated to SNP, a synergistic increase of apoptosis and inhibition of clonogenic activity was observed in K562 cells as well as in other myeloblastic (HEL, HL-60), lymphoblastic (Jurkat, SupT1), and multiple myeloma (RPMI 8226) cell lines. Although SNP greatly augmented TRAIL-mediated antileukemic activity also on primary leukemic blasts, normal erythroid and granulocytic cells were less sensitive to the cytotoxicity mediated by TRAIL with or without SNP. These data indicate that TRAIL promotes cytotoxicity in leukemic cells by activating effector caspases, which directly lead to apoptosis and stimulate NO production, which mediates cell cycle abnormalities. Both mechanisms seem to be essential for TRAIL-mediated cytotoxicity.


Assuntos
Apoptose/efeitos dos fármacos , Neoplasias Hematológicas/enzimologia , Glicoproteínas de Membrana/farmacologia , Óxido Nítrico Sintase/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Proteínas Reguladoras de Apoptose , Inibidores de Caspase , Caspases/metabolismo , Ciclo Celular/efeitos dos fármacos , Sinergismo Farmacológico , Inibidores Enzimáticos/farmacologia , Neoplasias Hematológicas/metabolismo , Neoplasias Hematológicas/patologia , Humanos , Células K562/efeitos dos fármacos , Glicoproteínas de Membrana/antagonistas & inibidores , Células-Tronco Neoplásicas/efeitos dos fármacos , Óxido Nítrico Sintase/efeitos dos fármacos , Óxido Nítrico Sintase/farmacologia , Nitroprussiato/farmacologia , Ligante Indutor de Apoptose Relacionado a TNF , Células Tumorais Cultivadas/efeitos dos fármacos , Fator de Necrose Tumoral alfa/antagonistas & inibidores
19.
Blood ; 97(9): 2596-603, 2001 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-11313247

RESUMO

Cytotoxic activity of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL/Apo-2 ligand), used alone or in different combinations with either a low (1.5 Gy) or a high (15 Gy) single dose of ionizing radiation (IR), was investigated on erythroleukemic cells (K562, HEL, Friend, primary leukemic erythroblasts) and on primary CD34(+)-derived normal erythroblasts. Human recombinant TRAIL alone variably affected the survival/growth of erythroleukemic cells; K562 cells were the most sensitive. Moreover, all erythroleukemic cells were radio-resistant, as demonstrated by the fact that cytotoxicity was evident only after treatment with high-dose (15 Gy) IR. Remarkably, when IR and TRAIL were used in combination, an additive effect was noticed in all erythroleukemic cells. Augmentation of TRAIL-induced cell death by IR was observed with both low and high IR doses and required the sequential treatment of IR 3 to 6 hours before the addition of TRAIL. Conversely, both TRAIL and IR showed a moderate cytotoxicity on primary CD34(+)-derived normal erythroblasts when used alone, but their combination did not show any additive effect. Moreover, the cytotoxicity of IR plus TRAIL observed in erythroleukemic cells was accompanied by the selective up-regulation of the surface expression of TRAIL-R1 (DR4), and it was completely blocked by the z-Val-Ala-Asp (OMe)-CH(2) (z-VAD-fmk) caspase inhibitor. On the other hand, the surface expression of TRAIL-R1 in CD34(+)-derived normal erythroblasts was unaffected by IR, which induced the up-regulation of the decoy TRAIL-R3. These data demonstrate that treatment with IR provides an approach to selectively sensitize erythroleukemic cells, but not normal erythroblasts, to TRAIL-induced apoptosis through the functional up-regulation of TRAIL-R1.


Assuntos
Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Eritroblastos/efeitos dos fármacos , Eritroblastos/patologia , Eritroblastos/efeitos da radiação , Glicoproteínas de Membrana/farmacologia , Fator de Necrose Tumoral alfa/farmacologia , Animais , Proteínas Reguladoras de Apoptose , Eritroblastos/metabolismo , Humanos , Leucemia Eritroblástica Aguda/patologia , Camundongos , Receptores do Ligante Indutor de Apoptose Relacionado a TNF , Receptores do Fator de Necrose Tumoral/agonistas , Transdução de Sinais , Ligante Indutor de Apoptose Relacionado a TNF , Células Tumorais Cultivadas , Regulação para Cima
20.
J Leukoc Biol ; 69(2): 263-70, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11272277

RESUMO

Stromal-derived factor-1alpha (SDF-1alpha), the high-affinity ligand of CXC-chemokine receptor 4 (CXCR4), induced a progressive increase of apoptosis when added to the Jurkat CD4+/CXCR4+ T cell line. The SDF-1alpha-mediated Jurkat cell apoptosis was observed in serum-free or serum-containing cultures, peaked at SDF-1alpha concentrations of 10-100 ng/ml, required 3 days to take place, and was completely blocked by the z-VAD-fmk tripeptide caspase inhibitor. Although SDF-1alpha did not modify the expression of TNF-alpha or that of TNF-RI and TNF-RII, it increased the expression of surface Fas/APO-1 (CD95) and intracellular Fas ligand (CD95L) significantly. Moreover, the ability of SDF-1alpha to induce apoptosis was inhibited by an anti-CD95 Fab' neutralizing antibody. These findings suggest a role for SDF-1alpha in the homeostatic control of CD4+ T-cell survival/apoptosis mediated by the CD95-CD95L pathway.


Assuntos
Apoptose/imunologia , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/imunologia , Quimiocinas CXC/fisiologia , Glicoproteínas de Membrana/biossíntese , Transdução de Sinais/imunologia , Regulação para Cima/imunologia , Receptor fas/biossíntese , Clorometilcetonas de Aminoácidos/farmacologia , Anticorpos Bloqueadores/farmacologia , Apoptose/efeitos dos fármacos , Linfócitos T CD4-Positivos/metabolismo , Inibidores de Caspase , Linhagem Celular Transformada , Membrana Celular/imunologia , Membrana Celular/metabolismo , Quimiocina CXCL12 , Quimiocinas CXC/antagonistas & inibidores , Inibidores de Cisteína Proteinase/farmacologia , Proteína Ligante Fas , Humanos , Soros Imunes/farmacologia , Líquido Intracelular/imunologia , Líquido Intracelular/metabolismo , Células Jurkat/citologia , Células Jurkat/efeitos dos fármacos , Células Jurkat/imunologia , Células Jurkat/metabolismo , Ligantes , Ativação Linfocitária , Glicoproteínas de Membrana/antagonistas & inibidores , Glicoproteínas de Membrana/fisiologia , Receptores do Fator de Necrose Tumoral/biossíntese , Células Estromais/imunologia , Fator de Necrose Tumoral alfa/biossíntese , Receptor fas/imunologia , Receptor fas/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...