Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Br J Dermatol ; 164(5): 1061-70, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21198538

RESUMO

BACKGROUND: Vascular endothelial growth factor (VEGF)-A, placenta growth factor (PlGF) and their corresponding membrane receptors are involved in autocrine and paracrine regulation of melanoma growth and metastasis. Besides the membrane receptors, a soluble form of the VEGF receptor (VEGFR)-1 (sVEGFR-1) has been identified, that behaves both as a decoy receptor, sequestering VEGF-A and PlGF, and as an extracellular matrix (ECM) molecule, promoting endothelial cell adhesion and migration through the interaction with α5ß1 integrin. OBJECTIVES: To analyse whether sVEGFR-1 plays a role during melanoma progression. METHODS: sVEGFR-1 expression was evaluated in a panel of 36 melanoma cell lines and 11 primary human melanocyte cultures by quantitative real-time polymerase chain reaction analysis and in specimens of primary or metastatic melanoma lesions from 23 patients by immunohistochemical analysis. RESULTS: sVEGFR-1 expression was highly upregulated in melanoma cell lines with respect to human melanocytes. Interestingly, cell lines obtained from cutaneous metastases showed a significant reduction of sVEGFR-1 expression, as compared with cell lines derived from primary tumours. These results were confirmed by immunohistochemical analysis of sections from primary skin melanomas and the corresponding cutaneous metastases, suggesting that modulation of sVEGFR-1 expression influences ECM invasion by melanoma cells and metastasis localization. Moreover, we provide evidence that adhesion of melanoma cells to sVEGFR-1 is favoured by the activation of a VEGF-A/VEGFR-2 autocrine loop. CONCLUSIONS: Our data strongly suggest that sVEGFR-1 plays a role in melanoma progression and that low sVEGFR-1/VEGF-A and sVEGFR-1/transmembrane VEGFR-1 ratios might predict a poor outcome in patients with melanoma.


Assuntos
Melanoma/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias Cutâneas/metabolismo , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Linhagem Celular Tumoral , Progressão da Doença , Humanos , Imuno-Histoquímica , Melanoma/secundário , Reação em Cadeia da Polimerase , Neoplasias Cutâneas/secundário
2.
Pharmacol Ther ; 213: 107579, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32442437

RESUMO

Ubiquitin Proteasome System (UPS) is an adaptable and finely tuned system that sustains proteostasis network under a large variety of physiopathological conditions. Its dysregulation is often associated with the onset and progression of human diseases; hence, UPS modulation has emerged as a promising new avenue for the development of treatments of several relevant pathologies, such as cancer and neurodegeneration. The clinical interest in proteasome inhibition has considerably increased after the FDA approval in 2003 of bortezomib for relapsed/refractory multiple myeloma, which is now used in the front-line setting. Thereafter, two other proteasome inhibitors (carfilzomib and ixazomib), designed to overcome resistance to bortezomib, have been approved for treatment-experienced patients, and a variety of novel inhibitors are currently under preclinical and clinical investigation not only for haematological malignancies but also for solid tumours. However, since UPS collapse leads to toxic misfolded proteins accumulation, proteasome is attracting even more interest as a target for the care of neurodegenerative diseases, which are sustained by UPS impairment. Thus, conceptually, proteasome activation represents an innovative and largely unexplored target for drug development. According to a multidisciplinary approach, spanning from chemistry, biochemistry, molecular biology to pharmacology, this review will summarize the most recent available literature regarding different aspects of proteasome biology, focusing on structure, function and regulation of proteasome in physiological and pathological processes, mostly cancer and neurodegenerative diseases, connecting biochemical features and clinical studies of proteasome targeting drugs.


Assuntos
Neoplasias/fisiopatologia , Doenças Neurodegenerativas/fisiopatologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Inibidores de Proteassoma/farmacologia , Ubiquitina/metabolismo , Quinases Ciclina-Dependentes/metabolismo , Resistência a Medicamentos/fisiologia , Fator de Transcrição E2F4/metabolismo , Holoenzimas , Humanos , Gotículas Lipídicas/metabolismo , Chaperonas Moleculares/metabolismo , Proteínas Musculares/metabolismo , NF-kappa B/metabolismo , Neoplasias/tratamento farmacológico , Doenças Neurodegenerativas/tratamento farmacológico , Complexo de Endopeptidases do Proteassoma/efeitos dos fármacos , Inibidores de Proteassoma/uso terapêutico , Proteostase/fisiologia , Proteína Supressora de Tumor p53/metabolismo
3.
Mol Cell Biol ; 10(7): 3806-9, 1990 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-2162481

RESUMO

Recent studies have shown that ligand-activated growth factor receptors as well as transforming versions of nonreceptor protein-tyrosine kinases physically associate with phosphatidylinositol-3 kinase (PI-3 kinase). Reasoning that PI-3 kinase might also play a role in the normal functions of nonreceptor kinases, we sought to determine whether association with PI-3 kinase might serve as a measure of nonreceptor protein-tyrosine kinase activation under physiological conditions. We found that p60c-src as well as p59fyn, the product of another member of the src family of proto-oncogenes, physically associated with a PI kinase activity within 5 s after exposure to thrombin. Furthermore, PI kinase reaction products generated in p60v-src, p60c-src or p59fyn containing immunoprecipitates were indistinguishable, demonstrating the identity of the associated enzyme as PI-3 kinase. These findings demonstrate a thrombin-dependent interaction between p60c-src or p59fyn and PI-3 kinase and suggest a role for nonreceptor protein-tyrosine kinases in human platelet signal transduction.


Assuntos
Plaquetas/enzimologia , Proteína Oncogênica pp60(v-src)/sangue , Fosfotransferases/sangue , Proteínas Tirosina Quinases/sangue , Proteínas Proto-Oncogênicas/sangue , Trombina/farmacologia , Plaquetas/efeitos dos fármacos , Humanos , Fosfatos de Inositol/sangue , Fosfatos de Inositol/isolamento & purificação , Cinética , Proteína Oncogênica pp60(v-src)/biossíntese , Proteína Oncogênica pp60(v-src)/isolamento & purificação , Fosfatidilinositol 3-Quinases , Fosfotransferases/biossíntese , Fosfotransferases/isolamento & purificação , Proteínas Tirosina Quinases/biossíntese , Proteínas Tirosina Quinases/isolamento & purificação , Proteínas Proto-Oncogênicas/biossíntese , Proteínas Proto-Oncogênicas/isolamento & purificação
4.
Oncogene ; 36(31): 4508-4515, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28368402

RESUMO

Melanoma is a dangerous form of skin cancer derived from the malignant transformation of melanocytes. The transcription factor SOX2 is not expressed in melanocytes, however, it has been shown to be differentially expressed between benign nevi and malignant melanomas and to be essential for melanoma stem cell maintenance and expansion in vitro and in xenograft models. By using a mouse model in which BRafV600E mutation cooperates with Pten loss to induce the development of metastatic melanoma, we investigated if Sox2 is required during the process of melanomagenesis, melanoma growth and metastasis and in the acquisition of resistance to BRAF inhibitors (BRAFi) treatments. We found that deletion of Sox2 specifically in Pten null and BRafV600E-expressing melanocytes did not prevent tumor formation and did not modify the temporal kinetics of melanoma occurrence compared to Sox2 wt mice. In addition, tumor growth was similar between Sox2 wt and Sox2 deleted (del) melanomas. By querying publicly available databases, we did not find statistically significant differences in SOX2 expression levels between benign nevi and melanomas, and analysis on two melanoma patient cohorts confirmed that Sox2 levels did not significantly change between primary and metastatic melanomas. Melanoma cell lines derived from both Sox2 genotypes showed a similar sensitivity to vemurafenib treatment and the same ability to develop vemurafenib resistance in long-term cultures. Development of vemurafenib resistance was not dependent on SOX2 expression also in human melanoma cell lines in vitro. Our findings exclude an oncogenic function for Sox2 during melanoma development and do not support a role for this transcription factor in the acquisition of resistance to BRAFi treatments.


Assuntos
Melanoma/etiologia , Fatores de Transcrição SOXB1/fisiologia , Animais , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos , Humanos , Indóis/uso terapêutico , Melanoma/tratamento farmacológico , Melanoma/patologia , Melanoma/secundário , Camundongos , PTEN Fosfo-Hidrolase/genética , PTEN Fosfo-Hidrolase/fisiologia , Proteínas Proto-Oncogênicas B-raf/antagonistas & inibidores , Proteínas Proto-Oncogênicas B-raf/genética , Proteínas Proto-Oncogênicas B-raf/fisiologia , Sulfonamidas/uso terapêutico , Vemurafenib
5.
Oncogene ; 2(6): 533-7, 1988 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-2455265

RESUMO

Ras p21 proteins exert their biological functions when associated to the inner surface of the plasma membrane. This association is mediated by a lipid molecule which is covalently attached to the protein by a thioester bond through a cysteine at residue 186, at the carboxy end of the molecule. Deletion or substitution of the critical Cys186 residue of the Harvey-ras protein leads to ras-p21 mutants lacking the ability to translocate to the membrane and devoid of transforming activity (Willumsen et al., 1984a, 1984b). We have been able to regenerate both localization to the plasma membrane as well as transforming activity of such mutant ras p21 proteins by fusion of the amino-terminal 15 residues of the v-p60src protein, responsible for the covalent binding of myristic acid and its membrane association. Thus, while translocation to the plasma membrane is necessary for function of the transforming Harvey-ras p21 protein, it appears to be independent of a specific membrane insertion mechanism.


Assuntos
Transformação Celular Neoplásica/fisiopatologia , Genes ras , Proteínas de Membrana/fisiologia , Ácidos Mirísticos/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Animais , Compartimento Celular , Linhagem Celular , Camundongos , Ácido Mirístico , Processamento de Proteína Pós-Traducional , Proteínas Proto-Oncogênicas pp60(c-src) , Relação Estrutura-Atividade
6.
Leukemia ; 9(11): 1888-95, 1995 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-7475280

RESUMO

Previous studies demonstrated that triazene compounds (TZC) possess antitumor, antimetastatic and immunosuppressive activity, and induce novel antigenic properties in neoplastic cells. Moreover, TZC showed marked antitumor activity in patients with acute myelogenous leukemias (AML). In most cases leukemic blasts with low levels of the repair enzyme O6-alkyl-guanine-DNA alkyltransferase (OGAT) were highly susceptible to TZC. Therefore the cytotoxic effects of TZC against human leukemic cells and the influence of OGAT modulation were investigated. Five leukemia cell lines were treated with the in vitro active derivative of dacarbazine: 5-(3-methyl-1-triazeno) imidazole-4-carboxamide (MTIC), or with temozolomide (TZM), which is readily cleaved to form the linear triazene MTIC in aqueous solution. The results showed that treatment with TZC at concentrations ranging between 62.5 and 250 microM significantly inhibited cell growth of U-937 and K-562 leukemia cell lines, both with undetectable OGAT activity. Growth inhibition was accompanied by DNA fragmentation and reduction of cell volume characteristic of cell undergoing apoptosis. In contrast, Daudi, HL-60 and Jurkat leukemia cell lines, characterized by high levels of the repair enzyme, were resistant to concentrations of TZC up to 500 microM. Treatment of resistant lines with O6-benzylguanine (BG, a specific inhibitor of OGAT) rendered HL-60 and Daudi but not Jurkat cells sensitive to cytotoxic effects and apoptosis mediated by MTIC. The results presented suggest that: (1) apoptosis is involved in cytotoxic activity of TZC; (2) OGAT could have a role in preventing programmed cell death induced by TZC; and (3) treatment with BG could potentiate cytotoxic and apoptotic effects of TZC on leukemic cell lines when high level of OGAT activity is the main factor involved in resistance to TZC.


Assuntos
Antineoplásicos Alquilantes/farmacologia , Apoptose/efeitos dos fármacos , Dacarbazina/análogos & derivados , Metiltransferases/deficiência , Dacarbazina/farmacologia , Expressão Gênica , Humanos , Leucemia/tratamento farmacológico , Leucemia/enzimologia , Metiltransferases/genética , O(6)-Metilguanina-DNA Metiltransferase , RNA Mensageiro/genética , Temozolomida , Células Tumorais Cultivadas
7.
Leukemia ; 14(8): 1451-9, 2000 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10942242

RESUMO

Mismatch repair deficiency contributes to tumor cell resistance to O6-guanine methylating compounds and to other antineoplastic agents. Here we demonstrate that MeOSO2(CH2)2-lexitropsin (Me-Lex), a DNA minor groove alkylating compound which generates mainly N3-methyladenine, has cytotoxic and clastogenic effects in mismatch repair-deficient leukemic cells. Moreover, MT-1 cells, which express p53 upon drug treatment and possess low levels of 3-methylpurine DNA glycosylase activity, are more susceptible to cytotoxicity induced by Me-Lex, with respect to p53-null and 3-methylpurine DNA glycosylase-proficient Jurkat cells. In both cell lines, the poly(ADP-ribose) polymerase inhibitor 3-aminobenzamide, which inhibits base excision repair capable of removing N-methylpurines, increases cytotoxicity and clastogenicity induced by Me-Lex or by temozolomide, which generates low levels of N3-methyl adducts. The enhancing effect is more evident at low Me-Lex concentrations, which induce a level of DNA damage that presumably does not saturate the repair ability of the cells. Nuclear fragmentation induced by Me-Lex + 3-aminobenzamide occurs earlier than in cells treated with the single agent. Treatment with Me-Lex and 3-aminobenzamide results in augmented expression of p53 protein and of the X-ray repair cross-complementing 1 transcript (a component of base excision repair). These results indicate that N3-methyladenine inducing agents, alone or combined with poly(ADP-ribose) polymerase inhibitors, could open up novel chemotherapeutic strategies to overcome drug resistance in mismatch repair-deficient leukemic cells.


Assuntos
Antineoplásicos/farmacologia , DNA de Neoplasias/efeitos dos fármacos , Mutagênicos/farmacologia , Netropsina/análogos & derivados , Apoptose , Aberrações Cromossômicas , Proteínas de Ligação a DNA/genética , Inibidores Enzimáticos/farmacologia , Regulação da Expressão Gênica , Células HT29 , Humanos , Células Jurkat , Netropsina/farmacologia , Inibidores de Poli(ADP-Ribose) Polimerases , Proteína Supressora de Tumor p53/biossíntese , Proteína 1 Complementadora Cruzada de Reparo de Raio-X
8.
Neoplasia ; 1(1): 42-9, 1999 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10935469

RESUMO

The expression of telomerase activity and the in situ localization of the human telomerase RNA component (hTR) in melanocytic skin lesions was evaluated in specimens from sixty-three patients. Specimens of melanocytic nevi, primary melanomas and subcutaneous metastases of melanoma were obtained from fifty-eight patients, whereas metastasized lymph nodes were obtained from five patients. Telomerase activity was determined in these specimens by using a Polymerase Chain Reaction-based assay (TRAP). High relative mean telomerase activity levels were detected in metastatic melanoma (subcutaneous metastases = 54.5, lymph node metastases = 56.5). Much lower levels were detected in primary melanomas, which increased with advancing levels of tumor cell penetration (Clark II = 0.02, Clark III = 1.1, and Clark IV = 1.9). Twenty-six formalin-fixed, paraffin-embedded melanocytic lesions were sectioned and analyzed for telomerase RNA with a radioactive in situ hybridization assay. In situ hybridization studies with a probe to the template RNA component of telomerase confirmed that expression was almost exclusively confined to tumor cells and not infiltrating lymphocytes. These results indicate that levels of telomerase activity and telomerase RNA in melanocytic lesions correlate well with clinical stage and could potentially assist in the diagnosis of borderline lesions.


Assuntos
Melanoma/enzimologia , Nevo Pigmentado/enzimologia , Neoplasias Cutâneas/enzimologia , Telomerase/biossíntese , Humanos , Hibridização In Situ , Melanoma/patologia , Melanoma/secundário , Mitose , Invasividade Neoplásica , Nevo Pigmentado/patologia , RNA/análise , Telomerase/genética
9.
J Invest Dermatol ; 115(6): 1000-7, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11121133

RESUMO

The vascular endothelial growth factor is produced by a large variety of human tumors, including melanoma, in which it appears to play an important role in the process of tumor-induced angiogenesis. Little information is available on the role of placenta growth factor, a member of the vascular endothelial growth factor family of cytokines, in tumor angiogenesis, even though placenta growth factor/vascular endothelial growth factor heterodimers have been recently isolated from tumor cells. To investigate the role of placenta growth factor and vascular endothelial growth factor homodimers and heterodimers in melanoma angiogenesis and growth, 19 human melanoma cell lines derived from primary or metastatic tumors were characterized for the expression of these cytokines and their receptors. Release of placenta growth factor and vascular endothelial growth factor polypeptides into the supernatant of human melanoma cells was demonstrated. Reverse transcriptase polymerase chain reaction analysis showed the presence of mRNAs encoding at least three different vascular endothelial growth factor isoforms (VEGF(121), VEGF(165), and VEGF(189)) and transcripts for two placenta growth factor isoforms (PlGF-1 and PlGF-2) in human melanoma cells. In addition, placenta growth factor expression in human melanoma in vivo was detected by immunohistochemical staining of tumor specimens. Both primary and metastatic melanoma cells were found to express the mRNAs encoding for vascular endothelial growth factor and placenta growth factor receptors (KDR, Flt-1, neuropilin-1, and neuropilin-2), and exposure of melanoma cells to these cytokines resulted in a specific proliferative response, supporting the hypothesis of a role of these angiogenic factors in melanoma growth. J Invest Dermatol 115:1000-1007 2000


Assuntos
Fatores de Crescimento Endotelial/farmacologia , Linfocinas/farmacologia , Melanoma/metabolismo , Melanoma/patologia , Dimerização , Humanos , Fator de Crescimento Placentário , Proteínas da Gravidez/farmacologia , Isoformas de Proteínas , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/patologia , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
10.
J Chemother ; 16(5): 479-86, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15565916

RESUMO

Preclinical studies based on a "simulation design", were performed with cultured melanoma cells prelabeled with 51Cr, added to normal blood and subjected to separation and recognition steps. Mononuclear cells (MNC) were isolated on ficollhypaque gradient, and melanoma cells were separated from lymphocytes using anti-CD45 immunomagnetic beads. Malignant cells were then recognized by measuring telomerase activity (TRAP and TRAP-ELISA assays). It was found that: (a)recovery of prelabeled cells present in MNC did not exceed 75%; (b) further recovery of prelabeled cells after separation from lymphocytes did not exceed 68%. Therefore, the overall recovery of prelabeled cells did not exceed 48%; (c) the entire procedure was able to reliably detect as few as 30 malignant cells added to normal blood, providing a telomerase signal significantly higher than that found in absence of melanoma cells. These results furnish the technical bases for developing a tumor detection assay in the blood of melanoma patients.


Assuntos
Biomarcadores Tumorais/sangue , Melanoma/diagnóstico , Células Neoplásicas Circulantes , Neoplasias Cutâneas/diagnóstico , Telomerase/sangue , Linhagem Celular Tumoral , Humanos , Melanoma/patologia , Valor Preditivo dos Testes , Sensibilidade e Especificidade , Neoplasias Cutâneas/patologia
11.
J Chemother ; 15(2): 173-83, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12797396

RESUMO

O6-alkylguanine-DNA alkyltransferase (OGAT) and the mismatch repair system (MRS) play a crucial role in the susceptibility of tumor cells to the cytotoxic effects of agents that generate O6-methylguanine in DNA, including the triazene compound temozolomide (TMZ). Studies performed with peripheral blood mononuclear cells (MNC) showed that TMZ was scarcely active on lymphocyte functions not dependent on cell proliferation (e.g. NK activity and cytokine-mediated induction of CD1b molecule in adherent MNC). In contrast, TMZ depressed proliferation and lymphokine activated killer (LAK) cell generation in response to IL-2. In this case, a reasonably good inverse relationship was found between OGAT levels of MNC and their susceptibility to TMZ. This study also analyzed the ratio of the toxic effect of TMZ on MNC and on tumor cells (i.e. "Tumor-Immune Function Toxicity Index", TIFTI). A particularly favorable TIFTI can be obtained when OGAT levels are extremely high in MNC and markedly low in tumor cells. This holds true for MRS-proficient neoplastic cells, but not for MRS-deficient tumors. In conclusion, strategies aimed at modulating OGAT and MRS may improve the clinical response to TMZ. However, the use of OGAT inhibitors to potentiate the antitumor activity of TMZ might result in a concomitant increase of the immunosuppressive effects of the drug, thus reducing the relative TIFTI.


Assuntos
Antineoplásicos Alquilantes/farmacologia , Reparo do DNA , Dacarbazina/análogos & derivados , Dacarbazina/farmacologia , Leucócitos Mononucleares/efeitos dos fármacos , O(6)-Metilguanina-DNA Metiltransferase/farmacologia , Linfoma de Burkitt/patologia , Divisão Celular , Dano ao DNA , Resistencia a Medicamentos Antineoplásicos , Humanos , Interleucina-2/farmacologia , Células Matadoras Ativadas por Linfocina , Leucemia Eritroblástica Aguda/patologia , Leucócitos Mononucleares/fisiologia , Linfócitos/fisiologia , Melanoma/patologia , O(6)-Metilguanina-DNA Metiltransferase/efeitos dos fármacos , Neoplasias Cutâneas/patologia , Temozolomida , Células Tumorais Cultivadas
12.
J Immunol ; 148(1): 161-8, 1992 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-1309367

RESUMO

A mouse IgM mAb termed P1E3 was raised against resting human peripheral blood neutrophils and has been shown to recognize a cell-surface Ag with an apparent molecular mass of 155 kDa, as assessed by immunoprecipitation analysis. In addition to the main 155-kDa protein, an additional band of about 210 kDa was also recognized by P1E3 in Western blot analysis. Sequential immunoprecipitation assays showed that the Ag recognized by P1E3 differed from the CD29 and CD45 Ag. However, sequential immunoprecipitation assays carried out with two distinct anti-CD15 mAb and P1E3 showed that P1E3 reacted with CD15 or with a CD15-like Ag. P1E3 stained strongly resting human peripheral blood neutrophils, hardly reacted with peripheral blood monocytes and did not react with PBL and platelets, as assessed by immunofluorescence flow cytometry. P1E3 inhibited the respiratory burst induced by PMA or FMLP, but not the oxidative response induced by Con A or the calcium ionophores A23187 or ionomycin. Furthermore, P1E3 inhibited the activation of the Na+/H+ antiporter in response to PMA or FMLP and the phosphorylation of a protein of about 50 kDa in response to PMA. However, preincubation of neutrophils with P1E3 did not affect the increase in cytosolic free calcium concentration induced by FMLP. These data suggest that the Ag recognized by P1E3 may play a role in modulating the activation of the respiratory burst induced by PMA or FMLP, and that P1E3 seems to affect protein kinase C-mediated signal transduction mechanisms coupled to the induction of the respiratory burst.


Assuntos
Anticorpos Monoclonais/imunologia , Antígenos de Superfície/imunologia , Neutrófilos/imunologia , Explosão Respiratória , Antígenos de Superfície/química , Cálcio/metabolismo , Proteínas de Transporte/metabolismo , Membrana Celular/imunologia , Citosol/fisiologia , Humanos , Concentração de Íons de Hidrogênio , Proteínas de Membrana/química , Proteínas de Membrana/imunologia , Peso Molecular , N-Formilmetionina Leucil-Fenilalanina/farmacologia , Fosfoproteínas/metabolismo , Fosforilação , Trocadores de Sódio-Hidrogênio , Superóxidos/metabolismo , Acetato de Tetradecanoilforbol/farmacologia
13.
J Pharmacol Exp Ther ; 279(1): 416-22, 1996 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-8859021

RESUMO

The cytotoxic and mutagenic properties of antitumor triazene compounds (TZC) have been mainly attributed to their ability to form DNA adducts at the O6 position of guanine. Repair of these lesions is mediated by O6-alkylguanine DNA alkyltransferase (OGAT) in an autoinactivating reaction. Therefore when lesion repair has occurred, cells are depleted of OGAT until synthesis of new enzyme molecules takes place. In this study, we have evaluated the ability of DNA alkylated by different TZC to deplete OGAT activity. Moreover, we have also investigated whether these compounds might inactivate the OGAT enzyme by a direct reaction with the protein. Human OGAT protein was partially purified from insect cells infected with a recombinant baculovirus containing the human OGAT coding sequences. Thereafter human OGAT protein was exposed directly to TZC or to TZC-alkylated DNA. Among the TZC tested, p-(3-methyl-1-triazeno)benzoic acid was the most effective OGAT inactivator by direct interaction with the protein. Moreover DNA substrates treated with methylating TZC, such as temozolomide or p-(3-methyl-1-triazeno)benzoic acid, were more effective in depleting the repair enzyme, compared to DNA pretreated with the chloroethylating TZC mitozolomide. In conclusion, our results show that TZC inactivate in vitro OGAT activity by either direct or indirect mechanisms. Therefore TZC are good candidates for 1) increasing their own cytotoxicity, if used according to appropriate dose and treatment schedules and 2) reversing tumor cell resistance to O6-guanine alkylating agents.


Assuntos
Antineoplásicos Alquilantes/farmacologia , Metiltransferases/antagonistas & inibidores , Animais , DNA/metabolismo , Dacarbazina/análogos & derivados , Dacarbazina/farmacologia , Resistência a Medicamentos , Guanina/análogos & derivados , Guanina/farmacologia , Humanos , O(6)-Metilguanina-DNA Metiltransferase , Proteínas Recombinantes/antagonistas & inibidores , Spodoptera , Temozolomida , Triazenos/farmacologia , Células Tumorais Cultivadas
14.
J Pharmacol Exp Ther ; 271(2): 1086-95, 1994 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-7965770

RESUMO

Human myeloid cell lines at different stages of differentiation (K562, HL60 and U937) were used to analyze the permissivity of the myelomonocytic lineage to acute infection with human T-cell leukemia virus type-I (HTLV-I) after cell-to-cell transmission and to evaluate the effect of cyclopentenone prostaglandins (PG)A1 and PGJ2 on virus transmission, proliferation of recipient cells and cell-mediated cytotoxicity against virus-donor cells. Exposure to HTLV-I delayed the growth rate of recipient cells, especially in U937 cells. This effect was related to the phase of cell cycle when cells were exposed to HTLV-I. Treatment of control and virus-exposed cells with these PGs, both inducing growth arrest prevalently at the G1/S interphase of the cell cycle, inhibited cell proliferation in a concentration-dependent way. The antiproliferative effect of both PGs increased progressively from pluripotent K562 to promyelocytic HL60 and monoblastoid U937 cells, suggesting that differentiated cells were more susceptible to PG-mediated inhibition of growth than pluripotent cells. PG treatment influenced the permissivity of recipient cells to HTLV-I, with different effects on less differentiated myeloid cells in comparison with more differentiated monoblastoid cells. In fact, the percentage of cells positive for the p19gag protein was increased among PG-treated K562 or HL60 cells, although it was reduced in PG-treated U937 cells. To this respect, PGA1 was more effective on asynchronous and PGJ2 on synchronous U937 cells.(ABSTRACT TRUNCATED AT 250 WORDS)


Assuntos
Vírus Linfotrópico T Tipo 1 Humano/fisiologia , Prostaglandina D2/análogos & derivados , Prostaglandinas A/farmacologia , Diferenciação Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , DNA Viral/análise , Proteínas de Choque Térmico HSP70/biossíntese , Humanos , Prostaglandina D2/farmacologia , Timidina/metabolismo , Células Tumorais Cultivadas
15.
J Pharmacol Exp Ther ; 271(2): 1096-102, 1994 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-7965771

RESUMO

Differentiation of cells of myelomonocitic lineage influences both cellular permissivity to infection with human T-cell leukemia virus type I after cell-to-cell virus transmission and sensitivity to the antiproliferative effect of cyclopentenone prostaglandins (PG)A1 and PGJ2. Growth inhibition and control of infection were found to be associated with high intracellular levels of inducible p72 heat shock protein (HSP70). Pluripotent K562 cells produced higher HSP70 base-line levels than promyelocytic HL60 or monoblastoid U937 cells. Treatment with PGA1 and especially with PGJ2 enhanced the synthesis of HSP70 in all these cells. Notably, HSP70 accumulated in virus-exposed U937 cells (but not in K562 or HL60 cells). Because in lethally irradiated virus-donor cells HSP70 production was barely detectable, expression of this protein in cocultured U937 cells can be prevalently attributed to virus-recipient cells. Treatment with PGA1 and even more with PGJ2 remarkably enhanced the synthesis of HSP70 in virus-exposed U937 cells, thus resulting in persistently high levels of HSP70 protein in the cells. As shown previously, in U937 cells treatment with PGs was associated with reduced percentages of virus p19gag positive cells and enhanced specific lysis of virus-donor cells at early time points after cell-to-cell transmission. Because the HSP70 protein family is involved in the control of cell proliferation as well as in antigen processing function during the immune response to pathogens, it is possible that persistent high expression levels of HSP70 in PG-treated cells play a critical role in regulating both cell cycling and antiviral cellular responses.


Assuntos
Proteínas de Choque Térmico HSP70/biossíntese , Vírus Linfotrópico T Tipo 1 Humano/fisiologia , Prostaglandina D2/análogos & derivados , Prostaglandinas A/farmacologia , Diferenciação Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Humanos , Prostaglandina D2/farmacologia , Biossíntese de Proteínas , Células Tumorais Cultivadas
16.
Scand J Immunol ; 34(1): 33-43, 1991 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-1676856

RESUMO

Components involved in superoxide anion production (cytochrome b) and in cell adhesion processes (CD11b, CD11c, CD18), two early functional responses of neutrophils during acute inflammation, are intracellularly located in resting human neutrophils. We have found a correlation between secretion of gelatinase and overexpression in the plasma membrane of CD11b, CD11c, CD18 and cytochrome b upon cell activation. Gelatinase and lactoferrin were parallely released after cell activation with different stimuli, but a better correlation between antigen up-regulation and gelatinase release was obtained. Total translocation of the intracellular pool of these mobilizable molecules to plasma membrane was achieved under conditions that induced total degranulation of the gelatinase-rich granule population, whereas 50% and 90% of the lactoferrin-containing secondary granules and peroxidase-containing primary granules, respectively, remained unfused. These results suggest a mechanism by which neutrophil function can be regulated through mobilization of gelatinase-rich granules, which can be considered as a subpopulation of secondary granules.


Assuntos
Grânulos Citoplasmáticos/imunologia , Neutrófilos/imunologia , Pepsina A/imunologia , Antígenos CD/imunologia , Antígenos CD11 , Antígenos CD18 , Membrana Celular/imunologia , Grupo dos Citocromos b/imunologia , Citometria de Fluxo , Imunofluorescência , Gelatinases , Humanos , Lactoferrina/imunologia , Ativação Linfocitária
17.
Biochem J ; 268(3): 707-12, 1990 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-1973035

RESUMO

We have found that an anti-CD11c monoclonal antibody (MAb) inhibits the respiratory burst induced in phorbol 12-myristate 13-acetate (PMA)-differentiated U937 cells as well as in human peripheral blood monocytes and neutrophils upon cell stimulation with concanavalin A. The MAb had no effect, however, when the added stimulus was fMet-Leu-Phe or PMA. Flow cytometry analyses indicated that concanavalin A was able to interact with CD11c. The anti-CD11c MAb inhibited significantly concanavalin A binding to differentiated U937 cells, and concanavalin A blocked binding of anti-CD11c MAb to the cells. Binding of labelled concanavalin A to membrane proteins which were separated by PAGE and transferred to nitrocellulose paper indicated that proteins with apparent molecular masses similar to those of CD11c (150 kDa) and CD18 (95 kDa) molecules were the main concanavalin A-binding proteins in differentiated U937 cells as well as in mature neutrophils. Similar experiments carried out in the presence of the anti-CD11c MAb showed a specific and significant inhibition of concanavalin A binding to the CD11c molecule. These results indicate that concanavalin A binds to the CD11c molecule and this binding is responsible for the concanavalin A-induced respiratory burst in PMA-differentiated U937 cells as well as in human mature monocytes and neutrophils.


Assuntos
Antígenos CD/metabolismo , Antígenos de Diferenciação/metabolismo , Concanavalina A/metabolismo , Fagócitos/metabolismo , Superóxidos/metabolismo , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacologia , Antígenos CD/imunologia , Antígenos de Diferenciação/imunologia , Antígenos CD11 , Linhagem Celular , Concanavalina A/antagonistas & inibidores , Concanavalina A/farmacologia , Citometria de Fluxo , Humanos , Monócitos/metabolismo , Neutrófilos/metabolismo , Fagocitose , Estimulação Química
18.
Mol Pharmacol ; 52(2): 249-58, 1997 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9271347

RESUMO

High levels of expression of the DNA repair enzyme O6-alkylguanine DNA-alkyltransferase (OGAT) (EC 2.1.1.63) account for tumor cell resistance to methylating agents. Previous studies suggested that methylating triazenes might have a potential role for the treatment of acute leukemias with low levels of OGAT. In the current study, we transduced the human OGAT cDNA in OGAT-deficient leukemia cell clones. OGAT-transduced cells were more resistant than their OGAT-deficient counterparts to apoptosis triggered by the methylating triazene temozolomide (TZM), as indicated by the results of flow cytometry, terminal deoxynucleotidyl transferase assay, and analysis of DNA fragmentation. Depletion of OGAT activity by O6-benzylguanine increased leukemia cell sensitivity to TZM-mediated apoptosis. Moreover, combined treatment of cells with TZM and benzamide, an inhibitor of the poly(ADP-ribose) polymerase (EC 2.4.2.30), increased the apoptosis induced by the methylating agent. These results demonstrate for the first time that methyl adducts at the O6 position of guanine, which are specifically removed by OGAT, are the principal DNA lesions responsible for the induction of apoptosis on treatment of leukemic cells with the methylating triazene TZM. This study also supports the possible use of TZM for the treatment of acute leukemias and suggests new strategies to increase the susceptibility of tumor cells to methylating triazenes in the clinic.


Assuntos
Antineoplásicos Alquilantes/farmacologia , Apoptose/efeitos dos fármacos , Reparo do DNA , Dacarbazina/análogos & derivados , Leucemia/tratamento farmacológico , Metiltransferases/antagonistas & inibidores , Inibidores de Poli(ADP-Ribose) Polimerases , Fragmentação do DNA/efeitos dos fármacos , Metilação de DNA , DNA de Neoplasias/metabolismo , Dacarbazina/farmacologia , Humanos , Leucemia/patologia , O(6)-Metilguanina-DNA Metiltransferase , Temozolomida , Células Tumorais Cultivadas
19.
J Pharmacol Exp Ther ; 285(2): 884-93, 1998 May.
Artigo em Inglês | MEDLINE | ID: mdl-9580640

RESUMO

The DNA repair enzyme O6-alkylguanine DNA-alkyltransferase (OGAT) and a deficient mismatch repair system play a critical role in the resistance to chemotherapeutic agents that generate adducts at the O6-position of guanine. However, DNA adducts different from O6-methylguanine might be also involved in cytotoxicity induced by methylating agents. Because the loss of p53 function is generally associated with tumor cell resistance to anticancer chemotherapy, we have investigated whether wild-type p53 might affect chemosensitivity of leukemia cells endowed with high OGAT levels to the methylating agent temozolomide (TZM). The effect of poly(ADP-ribose) polymerase (PADPRP) inhibition, which potentiates the cytotoxic effects of N7-methylguanine and N3-methylguanine, was also assessed in OGAT-proficient cells, either susceptible or tolerant to O6-methylguanine. OGAT-proficient and p53 null HL60 cells were transfected with the human p53 cDNA (p53+ cells). Treatment with TZM concentrations not toxic for the cells transduced with the control vector (p53-cells), induced apoptosis in p53+ cells. These cells were characterized by a lower level of bcl-2 protein than p53- cells, whereas bax and OGAT expression was comparable in both lines. Inhibition of PADPRP potentiated the cytotoxic and apoptotic effects of TZM in either p53- or p53+ HL60 cells. Furthermore, PADPRP inhibitors potentiated apoptosis induced by TZM in Jurkat cells, which possess a mutated p53 gene and are tolerant to O6-methylguanine adducts. The analysis of cell cycle indicated that the drug combination of TZM and PADPRP inhibitors provoked G1 arrest only in p53+ cells. Conversely, G1 arrest was not observed in p53+ cells exposed to TZM alone. It is possible to speculate that PADPRP inhibitors might affect the repair of DNA adducts that are processed differently from O6 methylguanine and induce a different pattern of cell cycle distribution. In conclusion, the results show that p53 increases apoptosis by TZM in OGAT-proficient cells and suggest the potential role of PADPRP inhibitors in enhancing TZM activity against leukemias independently of DNA repair systems.


Assuntos
Antineoplásicos Alquilantes/farmacologia , Dacarbazina/análogos & derivados , Inibidores Enzimáticos/farmacologia , Inibidores de Poli(ADP-Ribose) Polimerases , Proteína Supressora de Tumor p53/fisiologia , Apoptose , Benzamidas/farmacologia , Dacarbazina/farmacologia , Guanina/análogos & derivados , Guanina/farmacologia , Células HL-60 , Humanos , O(6)-Metilguanina-DNA Metiltransferase/genética , O(6)-Metilguanina-DNA Metiltransferase/fisiologia , Temozolomida
20.
J Pharmacol Exp Ther ; 291(3): 1292-300, 1999 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-10565854

RESUMO

Temozolomide (TMZ) is a new cytotoxic triazene compound of clinical interest that is able to generate methyl adducts at the O(6)-guanine of DNA, which can be repaired by O(6)-alkylguanine-DNA alkyltransferase (OGAT). It was previously found that triazene compounds are highly immunosuppressive in mice. In the present study, we investigate whether TMZ could affect immune functions of human competent cells and whether methylation of O(6)-guanine could be involved in the immunosuppressive activity of the drug. Mononuclear cells (MNCs) obtained from peripheral blood of healthy donors were tested for OGAT activity and treated with TMZ alone or combined with the OGAT inhibitor O(6)-benzylguanine. Control or drug-treated MNCs were then assayed for natural killer activity and for the ability to proliferate and to generate cytotoxic effector cells in response to interleukin-2 or allogeneic MT-2 tumor cells. The results show that TMZ inhibited both proliferation and induction of lytic activity in response to interleukin-2 or allogeneic MT-2 cells. Moreover, an inverse correlation was found between the OGAT activity of MNCs and their sensitivity to TMZ. The involvement of O(6)-guanine methylation in the immunosuppressive effects of TMZ was further confirmed by the finding that O(6)-benzylguanine increased the activity of the drug. On the other hand, the natural killer activity of MNCs was only moderately affected by TMZ, and no relationship was observed between OGAT levels and sensitivity to the drug. These data suggest that in patients with tumors who are undergoing TMZ treatment, the drug may impair immune responses involving cell proliferation, depending on OGAT levels of MNCs, and that O(6)-benzylguanine may potentiate this activity.


Assuntos
Antineoplásicos Alquilantes/toxicidade , Dacarbazina/análogos & derivados , Inibidores Enzimáticos/farmacologia , Guanina/análogos & derivados , Imunidade Celular/efeitos dos fármacos , Imunidade Inata/efeitos dos fármacos , Imunossupressores/toxicidade , Citotoxicidade Celular Dependente de Anticorpos/efeitos dos fármacos , Linhagem Celular , Dacarbazina/toxicidade , Guanina/farmacologia , Humanos , Interleucina-2/farmacologia , Monócitos/efeitos dos fármacos , Monócitos/imunologia , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/imunologia , Temozolomida , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA