Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Eur J Histochem ; 53(3): 177-82, 2009 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-19864212

RESUMO

Protein kinase C (PKC)-epsilon, a component of the serine/threonine PKC family, has been shown to influence the survival and differentiation pathways of normal hematopoietic cells. Here, we have modulated the activity of PKC-epsilon with specific small molecule activator or inhibitor peptides. PKC-epsilon inhibitor and activator peptides showed modest effects on HL-60 maturation when added alone, but PKC-epsilon activator peptide significantly counteracted the pro-maturative activity of tumor necrosis factor (TNF)-alpha towards the monocytic/macrophagic lineage, as evaluated in terms of CD14 surface expression and morphological analyses. Moreover, while PKC-epsilon inhibitor peptide showed a reproducible increase of TNF-related apoptosis inducing ligand (TRAIL)-induced apoptosis, PKC-epsilon activator peptide potently counteracted the pro-apoptotic activity of TRAIL. Taken together, the anti-maturative and anti-apoptotic activities of PKC-epsilon envision a potentially important proleukemic role of this PKC family member.


Assuntos
Proteína Quinase C-épsilon/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Apoptose/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Ativação Enzimática , Células HL-60 , Humanos , Proteína Quinase C-épsilon/antagonistas & inibidores , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia
2.
Mini Rev Med Chem ; 9(4): 498-509, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19356127

RESUMO

Although the role of serine/threonine protein kinase C (PKC) in malignant transformation is known from decades, an anti-PKC based approach in cancer therapy was hampered for the difficulties in developing pharmacological compounds able to selectively inhibit specific PKC isoforms. In this review, the role of PKC-epsilon and PKC-delta in promoting and counteracting tumor progression in different types of cancer, respectively, will be discussed in relationship with promising therapeutic perspectives based either on small molecule inhibitors or on natural compounds. Among a myriad of molecules able to modulate PKC activity, we will focus on the role of the enzastaurin and briostatin-1, which already entered clinical trials for several human cancers.


Assuntos
Antineoplásicos/uso terapêutico , Proteína Quinase C-delta/antagonistas & inibidores , Proteína Quinase C-delta/fisiologia , Proteína Quinase C-épsilon/antagonistas & inibidores , Proteína Quinase C-épsilon/fisiologia , Inibidores de Proteínas Quinases/uso terapêutico , Animais , Antineoplásicos/farmacologia , Apoptose/fisiologia , Briostatinas/uso terapêutico , Ciclo Celular , Transformação Celular Neoplásica/metabolismo , Ensaios Clínicos como Assunto , Humanos , Indóis/uso terapêutico , Metástase Neoplásica , Neovascularização Patológica , Extratos Vegetais/uso terapêutico , Proteína Quinase C-delta/metabolismo , Proteína Quinase C-épsilon/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-mdm2/antagonistas & inibidores , Proteína Supressora de Tumor p53/metabolismo
3.
Eur J Histochem ; 53(1): 19-24, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19351609

RESUMO

Although several studies have shown that the serum levels of osteoprotegerin (OPG) are significantly elevated in patients affected with atherosclerotic lesions in coronary and peripheral arteries, the cellular source and the role of OPG in the physiopathology of atherosclerosis are not completely defined. Therefore, we aimed to investigate the potential contribution of mesenchymal stem cells in the production/release of OPG. OPG was detectable by immunohistochemistry in aortic and coronary atherosclerotic plaques, within or in proximity of intimal vascular smooth muscle cells (SMC). In addition, bone marrow mesenchymal stem cell (MSC)-derived vascular SMC as well as primary aortic SMC released in the culture supernatant significantly higher levels of OPG with respect to MSC-derived endothelial cells (EC) or primary aortic EC. On the other hand, in vitro exposure to full-length human recombinant OPG significantly increased the proliferation rate of aortic SMC cultures, as monitored by bromodeoxyuridine incorporation. Taken together, these data suggest that OPG acts as an autocrine/paracrine growth factor for vascular SMC, which might contribute to the progression of atherosclerotic lesions.


Assuntos
Células-Tronco Mesenquimais/metabolismo , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/metabolismo , Osteoprotegerina/metabolismo , Aorta/citologia , Aorta/metabolismo , Aterosclerose/metabolismo , Humanos , Músculo Liso Vascular/metabolismo
4.
Eur J Histochem ; 53(1): e3, 2009 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-30256862

RESUMO

Although several studies have shown that the serum levels of osteoprotegerin (OPG) are significantly elevated in patients affected with atherosclerotic lesions in coronary and peripheral arteries, the cellular source and the role of OPG in the physiopathology of atherosclerosis are not completely defined. Therefore, we aimed to investigate the potential contribution of mesenchymal stem cells in the production/release of OPG. OPG was detectable by immunohistochemistry in aortic and coronary atherosclerotic plaques, within or in proximity of intimal vascular smooth muscle cells (SMC). In addition, bone marrow mesenchymal stem cell (MSC)-derived vascular SMC as well as primary aortic SMC released in the culture supernatant significantly higher levels of OPG with respect to MSC-derived endothelial cells (EC) or primary aortic EC. On the other hand, in vitro exposure to full-length human recombinant OPG significantly increased the proliferation rate of aortic SMC cultures, as monitored by bromodeoxyuridine incorporation. Taken together, these data suggest that OPG acts as an autocrine/paracrine growth factor for vascular SMC, which might contribute to the progression of atherosclerotic lesions.

5.
Eur J Histochem ; 53(3): e21, 2009 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-30256868

RESUMO

Protein kinase C (PKC)-ε, a component of the serine/threo-nine PKC family, has been shown to influence the survival and differentiation pathways of normal hematopoietic cells. Here, we have modulated the activity of PKC-ε with specific small molecule activator or inhibitor peptides. PKC-ε inhibitor and activator peptides showed modest effects on HL-60 maturation when added alone, but PKC-ε activator peptide significantly counteracted the pro-maturative activity of tumor necrosis factor (TNF)-α towards the monocytic/macrophagic lineage, as evaluated in terms of CD14 surface expression and morphological analyses. Moreover, while PKC-ε inhibitor peptide showed a reproducible increase of TNF-related apoptosis inducing ligand (TRAIL)-induced apoptosis, PKC-ε activator peptide potently counteracted the pro-apoptotic activity of TRAIL. Taken together, the anti-maturative and anti-apoptotic activities of PKC-ε envision a potentially important proleukemic role of this PKC family member.

6.
Apoptosis ; 9(2): 235-46, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15004520

RESUMO

Erythropoiesis occurs in bone marrow and it has been shown that during in vivo erythroid differentiation some immature erythroblasts undergo apoptosis. In this regard, it is known that immature erythroblasts are FasL- and TRAIL-sensitive and can be killed by cells expressing these ligand molecules. In the present study, we have investigated the cell death phenomenon that occurs during a common unilineage model of erythroid development. Purified CD34+ human haemopoietic progenitors were cultured in vitro in the presence of SCF, IL-3 and erythropoietin. Their differentiation stages and apoptosis were followed by multiple technical approaches. Flow cytometric evaluation of surface and intracellular molecules revealed that glycophorin A appeared at day 3-4 of incubation and about 75% of viable cells co-expressed high density glycophorin A (Gly(bright)) and adult haemoglobin at day 14 of culture, indicating that this system reasonably recapitulates in vivo normal erythropoiesis. Interestingly, when mature (Gly(bright)) erythroid cells reached their higher percentages (day 14) almost half of cultured cells were apoptotic. Morphological studies indicated that the majority of dead cells contained cytoplasmic granular material typical of basophilic stage, and DNA analysis by flow cytometry and TUNEL reaction revealed nuclear fragmentation. These observations indicate that in vitro unilineage erythroid differentiation, as in vivo, is associated with apoptotic cell death of cells with characteristics of basophilic erythroblasts. We suggest that the interactions between different death receptors on immature basophilic erythroblasts with their ligands on more mature erythroblasts may contribute to induce apoptosis in vitro.


Assuntos
Apoptose/fisiologia , Diferenciação Celular/fisiologia , Células Precursoras Eritroides/fisiologia , Antígenos CD34/metabolismo , Células Precursoras Eritroides/citologia , Células Precursoras Eritroides/ultraestrutura , Citometria de Fluxo , Humanos , Microscopia Eletrônica
7.
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
8.
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
9.
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
10.
J Med Virol ; 63(4): 277-83, 2001 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11241458

RESUMO

Chronic gastritis is associated frequently with persistent infection by Helicobacter pylori. However, not all patients with chronic gastritis have evidence of H. pylori infection, suggesting that other factors might contribute to the development of gastritis. The present study was undertaken to evaluate a possible etiologic role of human herpesvirus 7 (HHV-7). HHV-7 DNA was detected in about 80% of gastric biopsies, both in healthy mucosa from individuals without evidence of inflammation and in biopsies from patients with histologically confirmed chronic gastric inflammation. HHV-7 was present also in H. pylori negative samples, was associated specifically with gastric tissue and not with residual blood within the mucosa, and was present with high viral loads. HHV-7 DNA persisted in several patients also after remission of gastric inflammation and the viral presence did not correlate with specific symptoms. Analysis by RT-PCR showed that HHV-7 is transcriptionally inactive in chronic gastritis lesions. These observations show that gastric tissue represents a site of HHV-7 latent infection and a potential reservoir for viral reactivation.


Assuntos
Mucosa Gástrica/virologia , Gastrite/etiologia , Herpesvirus Humano 7/isolamento & purificação , Biópsia , Doença Crônica , DNA Viral/análise , Seguimentos , Mucosa Gástrica/patologia , Gastrite/patologia , Helicobacter pylori , Herpesvirus Humano 7/genética , Humanos , Leucócitos Mononucleares/virologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Carga Viral , Latência Viral
11.
Br J Haematol ; 111(2): 432-40, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11122082

RESUMO

CXC chemokine receptor 4 (CXCR4), the high-affinity receptor for stroma-derived factor 1alpha (SDF-1alpha), shows distinct patterns of expression in human CD34+ haematopoietic progenitor cells induced to differentiate in vitro along the granulocytic and erythroid lineages. In serum-free liquid cultures supplemented with stem cell factor (SCF), interleukin 3 (IL-3) and granulocyte colony-stimulating factor, the expression of surface CXCR4 progressively increased in cells differentiating along the granulocytic lineage. The addition in culture of 200 ng/ml of SDF-1alpha, a concentration which maximally activated intracellular Ca2+ flux, only modestly affected the expression levels of CD15 and CD11b granulocytic antigens, as well as the total number of viable cells. On the other hand, in liquid cultures supplemented with SCF, IL-3 and erythropoietin, SDF-1alpha induced the downregulation of glycophorin A erythroid antigen, accompanied by a progressive decline in the number of viable erythroblasts. Moreover, in semisolid assays, SDF-1alpha significantly reduced the number of plurifocal erythroid colonies (erythroid blast-forming units; BFU-E), whereas it did not affect granulocyte-macrophage colony-forming units (CFU-GM). We also demonstrated that the inhibitory effect of SDF-1alpha on glycophorin A+ erythroid cell development was mediated by the functional upregulation of CD95L in erythroid cultures. These data indicate that SDF-1alpha plays a role as a negative regulator of erythropoiesis.


Assuntos
Antígenos CD34 , Quimiocinas CXC/farmacologia , Eritropoese/efeitos dos fármacos , Células-Tronco Hematopoéticas/metabolismo , Glicoproteínas de Membrana/metabolismo , Receptores CXCR4/metabolismo , Cálcio/metabolismo , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Quimiocina CXCL12 , Depressão Química , Proteína Ligante Fas , Sangue Fetal/citologia , Glicoforinas/metabolismo , Fator Estimulador de Colônias de Granulócitos/farmacologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Humanos , Interleucina-3/farmacologia , Fator de Células-Tronco/farmacologia
13.
Proc Natl Acad Sci U S A ; 95(23): 13911-6, 1998 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-9811900

RESUMO

Human herpesvirus 6 (HHV-6) like other herpesviruses, expresses sequentially immediate early (IE), early, and late genes during lytic infection. Evidence of ability to establish latent infection has not been available, but by analogy with other herpesviruses it could be expected that IE genes that regulate and transactivate late genes would not be expressed. We report that peripheral blood mononuclear cells of healthy individuals infected with HHV-6 express the U94 gene, transcribed under IE conditions. Transcription of other IE genes (U16/17, U39, U42, U81, U89/90, U91) was not detected. To verify that U94 may play a role in the maintenance of the latent state, we derived lymphoid cell lines that stably expressed U94. HHV-6 was able to infect these cells, but viral replication was restricted. No cytopathic effect developed. Furthermore, viral transcripts were present in the first days postinfection and declined thereafter. A similar decline in the level of intracellular viral DNA also was observed. These findings are consistent with the hypothesis that the U94 gene product of HHV-6 regulates viral gene expression and enables the establishment and/or maintenance of latent infection in lymphoid cells.


Assuntos
Regulação Viral da Expressão Gênica , Genes Precoces , Genes Virais , Herpesvirus Humano 6/fisiologia , Leucócitos Mononucleares/virologia , Linfócitos/virologia , Transformação Celular Viral , Células Cultivadas , Humanos , Replicação Viral/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA