Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Exp Dermatol ; 23(3): 202-4, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24495013

RESUMO

Myeloid-derived suppressor cells (MDSC) are a heterogeneous cell population characterized by immunosuppressive activity. Elevated levels of MDSC in peripheral blood are found in inflammatory diseases as well as in malignant tumors where they are supposed to be major contributors to mechanisms of tumor-associated tolerance. We investigated the frequency and function of MDSC in peripheral blood of melanoma patients and observed an accumulation of CD11b(+) CD33(+) CD14(+) HLA-DR(low) MDSC in all stages of disease (I-IV), including early stage I patients. Disease progression and enhanced tumor burden did not result in a further increase in frequencies or change in phenotype of MDSC. By investigation of specific MDSC-associated cytokines in patients' sera, we found an accumulation of IL-8 in all stages of disease. T-cell proliferation assays revealed that MDSC critically contribute to suppressed antigen-specific T-cell reactivity and thus might explain the frequently observed transient effects of immunotherapeutic strategies in melanoma patients.


Assuntos
Leucócitos Mononucleares/química , Melanoma/imunologia , Melanoma/patologia , Células Mieloides/imunologia , Neoplasias Cutâneas/imunologia , Neoplasias Cutâneas/patologia , Antígeno CD11b/análise , Estudos de Casos e Controles , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Técnicas de Cocultura , Progressão da Doença , Antígenos HLA-DR/análise , Humanos , Interleucina-8/sangue , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/imunologia , Receptores de Lipopolissacarídeos/análise , Ativação Linfocitária/imunologia , Contagem de Linfócitos , Células Mieloides/química , Estadiamento de Neoplasias , Receptores de Antígenos de Linfócitos T , Lectina 3 Semelhante a Ig de Ligação ao Ácido Siálico/análise , Linfócitos T Reguladores/imunologia , Toxoide Tetânico/farmacologia , Carga Tumoral , Evasão Tumoral
2.
PLoS One ; 8(12): e83384, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24376696

RESUMO

Regulatory T cells (Treg) play a pivotal role in the immune system since they inhibit the T cell response. It is well known that cyclophosphamide applied at low dose is able to stimulate the immune response while high dose cyclophosphamide exerts inhibitory activity. Data obtained in mice indicate that cyclophosphamide provokes a reduction in the number of Treg and impairs their suppressive activity, resulting in immune stimulation. Here, we addressed the question of the sensitivity of human Treg to cyclophosphamide, comparing Treg with cytotoxic T cells (CTL) and T helper cells (Th). We show that Treg are more sensitive than CTL and Th to mafosfamide, which is an active derivative of cyclophosphamide, which does not need metabolic activation. The high sensitivity of Treg was due to the induction of apoptosis. Treg compared to CTL and Th were not more sensitive to the alkylating drugs temozolomide and nimustine and also not to mitomycin C, indicating a specific Treg response to mafosfamide. The high sensitivity of Treg to mafosfamide resulted not only in enhanced cell death, but also in impaired Treg function as demonstrated by a decline in the suppressor activity of Treg in a co-culture model with Th and Helios positive Treg. Treatment of Treg with mafosfamide gave rise to a high level of DNA crosslinks, which were not repaired to the same extent as observed in Th and CTL. Also, Treg showed a low level of γH2AX foci up to 6 h and a high level 24 h after treatment, indicating alterations in the DNA damage response. Overall, this is the first demonstration that human Treg are, in comparison with Th and CTL, hypersensitive to cyclophosphamide, which is presumably due to a DNA repair defect.


Assuntos
Ciclofosfamida/análogos & derivados , Imunossupressores/farmacologia , Subunidade alfa de Receptor de Interleucina-2/metabolismo , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/imunologia , Apoptose/efeitos dos fármacos , Ciclofosfamida/farmacologia , Relação Dose-Resposta a Droga , Humanos , Necrose/induzido quimicamente , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/metabolismo
3.
Cancer Res ; 73(18): 5647-56, 2013 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-23878189

RESUMO

IFN-α is an antineoplastic agent in the treatment of several solid and hematologic malignancies that exerts strong immune- and autoimmune-stimulating activity. However, the mechanisms of immune activation by IFN-α remain incompletely understood, particularly with regard to CD4(+)CD25(high)Foxp(+) regulatory T cells (Treg). Here, we show that IFN-α deactivates the suppressive function of human Treg by downregulating their intracellular cAMP level. IFN-α-mediated Treg inactivation increased CD4(+) effector T-cell activation and natural killer cell tumor cytotoxicity. Mechanistically, repression of cAMP in Treg was caused by IFN-α-induced MAP-ERK kinase (MEK)/extracellular signal-regulated kinase (ERK)-mediated phosphodiesterase 4 (PDE4) activation and accompanied by downregulation of IFN receptor (IFNAR)-2 and negative regulation of T-cell receptor signaling. IFN-α did not affect the anergic state, cytokine production, Foxp3 expression, or methylation state of the Treg-specific demethylated region (TSDR) within the FOXP3 locus associated with a stable imprinted phenotype of human Treg. Abrogated protection by IFN-α-treated Treg in a humanized mouse model of xenogeneic graft-versus-host disease confirmed IFN-α-dependent regulation of Treg activity in vivo. Collectively, the present study unravels Treg inactivation as a novel IFN-α activity that provides a conceivable explanation for the immune-promoting effect and induction of autoimmunity by IFN-α treatment in patients with cancer and suggests IFN-α for concomitant Treg blockade in the context of therapeutic vaccination against tumor antigens.


Assuntos
Autoimunidade/efeitos dos fármacos , AMP Cíclico/antagonistas & inibidores , Doença Enxerto-Hospedeiro/imunologia , Interferon-alfa/farmacologia , Células Matadoras Naturais/imunologia , Subpopulações de Linfócitos T/imunologia , Linfócitos T Reguladores/efeitos dos fármacos , Animais , Células Cultivadas , AMP Cíclico/metabolismo , Proteínas de Ligação a DNA/fisiologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Doença Enxerto-Hospedeiro/metabolismo , Doença Enxerto-Hospedeiro/patologia , Humanos , Subunidade alfa de Receptor de Interleucina-2/metabolismo , Células Matadoras Naturais/metabolismo , Células Matadoras Naturais/patologia , Ativação Linfocitária , Camundongos , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Fosforilação , Fatores de Transcrição STAT/metabolismo , Subpopulações de Linfócitos T/metabolismo , Subpopulações de Linfócitos T/patologia , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo
4.
Macromol Biosci ; 12(12): 1637-47, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23042770

RESUMO

Nanoparticles (NP) represent a promising tool for biomedical applications. Here, sulfonate- and phosphonate-functionalized polystyrene NP are analyzed for their interaction with human monocyte-derived dendritic cells (DC). Immature dendritic cells (iDC) display a higher time- and dose-dependent uptake of functionalized polystyrene NP compared to mature dendritic cells (mDC). Notably, NP induce an enhanced maturation of iDC but not of mDC (upregulation of stimulatory molecules and cytokines). NP-triggered maturation results in a significantly enhanced T cell stimulatory capacity (increased CD4(+) T cell proliferation and IFN-γ production), indicating a shift to a pronounced Th1 response. Immunomodulatory properties of NP may be a useful strategy for strengthening the efficacy of NP-based approaches in immunotherapy.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Células Dendríticas/imunologia , Fatores Imunológicos/química , Ativação Linfocitária/efeitos dos fármacos , Nanopartículas/química , Poliestirenos/química , Citocinas/imunologia , Células Dendríticas/efeitos dos fármacos , Humanos , Fatores Imunológicos/farmacologia , Interferon gama/imunologia , Microscopia Confocal , Organofosfonatos , Poliestirenos/farmacologia , Ácidos Sulfônicos
5.
PLoS One ; 6(7): e22763, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21818385

RESUMO

BACKGROUND: Administration of interferon-α (IFN-α) represents an approved adjuvant therapy as reported for malignancies like melanoma and several viral infections. In malignant diseases, tolerance processes are critically involved in tumor progression. In this study, the effect of IFN-α on tolerance induction by human tolerogenic dendritic cells (DC) was analyzed. We focussed on tolerogenic IL-10-modulated DC (IL-10 DC) that are known to induce anergic regulatory T cells (iTregs). METHODOLOGY/PRINCIPAL FINDINGS: IFN-α promoted an enhanced maturation of IL-10 DC as demonstrated by upregulation of the differentiation marker CD83 as well as costimulatory molecules. IFN-α treatment resulted in an increased capacity of DC to stimulate T cell activation compared to control tolerogenic DC. We observed a strengthened T cell proliferation and increased IFN-γ production of CD4(+) and CD8(+) T cells stimulated by IFN-α-DC, demonstrating a restoration of the immunogenic capacity of tolerogenic DC in the presence of IFN-α. Notably, restimulation experiments revealed that IFN-α treatment of tolerogenic DC abolished the induction of T cell anergy and suppressor function of iTregs. In contrast, IFN-α neither affected the priming of iTregs nor converted iTregs into effector T cells. CONCLUSIONS/SIGNIFICANCE: IFN-α inhibits the induction of T cell tolerance by reversing the tolerogenic function of human DC.


Assuntos
Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Tolerância Imunológica/imunologia , Interferon-alfa/farmacologia , Diferenciação Celular/imunologia , Anergia Clonal/efeitos dos fármacos , Células Dendríticas/citologia , Humanos , Tolerância Imunológica/efeitos dos fármacos , Imunização , Imunofenotipagem , Interleucina-10/farmacologia , Ativação Linfocitária/efeitos dos fármacos , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/imunologia
6.
J Immunol ; 184(11): 6025-34, 2010 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-20421643

RESUMO

Dendritic cells (DCs) are the most potent APCs of the immune system. Understanding the intercellular and intracellular signaling processes that lead to DC maturation is critical for determining how these cells initiate T cell-mediated immune processes. NO synthesized by the inducible NO synthase (iNOS) is important for the function of murine DCs. In our study, we investigated the regulation of the arginine/NO-system in human monocyte-derived DCs. Maturation of DCs induced by inflammatory cytokines (IL-1beta, TNF, IL-6, and PGE(2)) resulted in a pronounced expression of neuronal NOS (nNOS) but only minimal levels of iNOS and endothelial NOS were detected in human mature DCs. In addition, reporter cell assays revealed the production of NO by mature DCs. Specific inhibitors of NOS (N-nitro-L-arginine methyl ester) or of the NO target guanylyl cyclase (H-(1,2,4)-oxadiazolo [4,3-a] quinoxalin-1-one) prevented DC maturation (shown by decreased expression of MHC class II, costimulatory and CD83 molecules and reduced IL-12 production) and preserved an immature phenotype, indicating an autocrine effect of nNOS-derived NO on human DC maturation. Notably, inhibitor-treated DCs were incapable of inducing efficient T cell responses after primary culture and generated an anergic T cell phenotype. In conclusion, our results suggest that, in the human system, nNOS-, but not iNOS-derived NO, plays an important regulatory role for the maturation of DCs and, thus, the induction of pronounced T cell responses.


Assuntos
Diferenciação Celular/imunologia , Células Dendríticas/citologia , Óxido Nítrico Sintase Tipo I/imunologia , Linhagem Celular , Separação Celular , Citocinas/biossíntese , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Humanos , Immunoblotting , Ativação Linfocitária/imunologia , Óxido Nítrico/biossíntese , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo I/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
7.
Int J Cancer ; 123(2): 312-321, 2008 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-18425820

RESUMO

Tumor cells express the glycolytic regulator pyruvate kinase subtype M2 (M2-PK), which can occur in a tetrameric form with high affinity to its substrate phosphoenolpyruvate (PEP) and a dimeric form with a low PEP affinity. The transition between both conformations contributes to the control of glycolysis and is important for tumor cell proliferation and survival. Here we targeted M2-PK by synthetic peptide aptamers, which specifically bind to M2-PK and shift the isoenzyme into its low affinity dimeric conformation. The aptamer-induced dimerization and inactivation of M2-PK led to a significant decrease in the PK mass-action ratio as well as ATP:ADP ratio in the target cells. Furthermore, the expression of M2-PK-binding peptide aptamers moderately reduced the growth of immortalized NIH3T3 cell populations by decelerating cell proliferation, but without affecting apoptotic cell death. Moreover, the M2-PK-binding peptide aptamers also reduced the proliferation rate of human U-2 OS osteosarcoma cells. In the present study, we developed the first specific inhibitors of the pyruvate kinase isoenzyme type M2 and present evidence that these inhibitors moderately decelerate tumor cell proliferation.


Assuntos
Antineoplásicos/farmacologia , Aptâmeros de Peptídeos/farmacologia , Osteossarcoma/tratamento farmacológico , Inibidores de Proteínas Quinases/farmacologia , Piruvato Quinase/antagonistas & inibidores , Difosfato de Adenosina/metabolismo , Trifosfato de Adenosina/metabolismo , Sequência de Aminoácidos , Animais , Antineoplásicos/metabolismo , Apoptose/efeitos dos fármacos , Aptâmeros de Peptídeos/metabolismo , Western Blotting , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Glicólise , Humanos , Focalização Isoelétrica , Isomerismo , Camundongos , Dados de Sequência Molecular , Células NIH 3T3 , Osteossarcoma/metabolismo , Plasmídeos , Inibidores de Proteínas Quinases/metabolismo , Estrutura Quaternária de Proteína/efeitos dos fármacos , Piruvato Quinase/metabolismo , Ensaio Tumoral de Célula-Tronco
8.
Cancer Res ; 66(6): 3024-33, 2006 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-16540651

RESUMO

Insulin-like growth factor binding protein-3 (IGFBP-3), the product of a tumor suppressor target gene, can modulate cell proliferation and apoptosis by IGF-I-dependent and IGF-I-independent mechanisms. IGFBP-3 controls the bioavailability of IGFs in the extracellular environment and is known to be subject to degradation by various extracellular proteases. Although nuclear localization and functions of IGFBP-3 have been described in the past, we show as the novel features of this study that the abundance of nuclear IGFBP-3 is directly regulated by ubiquitin/proteasome-dependent proteolysis. We show that IGFBP-3 degradation depends on an active ubiquitin-E1 ligase, specific 26S proteasome inhibitors can efficiently stabilize nuclear IGFBP-3, and the metabolic half-life of nuclear IGFBP-3 is strongly reduced relative to cytoplasmic IGFBP-3. Nuclear IGFBP-3 is highly polyubiquitinated at multiple lysine residues in its conserved COOH-terminal domain and stabilized through mutation of two COOH-terminal lysine residues. Moreover, we show that IGFBP-3, if ectopically expressed in the nucleus, can induce apoptotic cell death. These results suggest that ubiquitin/proteasome-mediated proteolysis of IGFBP-3 may contribute to down-regulation of apoptosis.


Assuntos
Apoptose/fisiologia , Neoplasias Ósseas/metabolismo , Proteína 3 de Ligação a Fator de Crescimento Semelhante à Insulina/metabolismo , Osteossarcoma/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Ubiquitina/metabolismo , Neoplasias Ósseas/enzimologia , Neoplasias Ósseas/patologia , Linhagem Celular Tumoral , Núcleo Celular/enzimologia , Núcleo Celular/metabolismo , Citosol/enzimologia , Citosol/metabolismo , Humanos , Osteossarcoma/enzimologia , Osteossarcoma/patologia , Enzimas Ativadoras de Ubiquitina/metabolismo
9.
Br J Haematol ; 132(5): 615-22, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16445836

RESUMO

Natural products are still an untapped source of promising lead compounds for the generation of antineoplastic drugs. Here, we investigated for the first time the antiproliferative and apoptotic effects of highly purified oxindole alkaloids, namely isopteropodine (A1), pteropodine (A2), isomitraphylline (A3), uncarine F (A4) and mitraphylline (A5) obtained from Uncaria tomentosa, a South American Rubiaceae, on human lymphoblastic leukaemia T cells (CCRF-CEM-C7H2). Four of the five tested alkaloids inhibited proliferation of acute lymphoblastic leukaemia cells. Furthermore, the antiproliferative effect of the most potent alkaloids pteropodine (A2) and uncarine F (A4) correlated with induction of apoptosis. After 48 h, 100 micromol/l A2 or A4 increased apoptotic cells by 57%. CEM-C7H2 sublines with tetracycline-regulated expression of bcl-2, p16ink4A or constitutively expressing the cowpox virus protein crm-A were used for further studies of the apoptosis-inducing properties of these alkaloids. Neither overexpression of bcl-2 or crm-A nor cell-cycle arrest in G0/G1 phase by tetracycline-regulated expression of p16INK4A could prevent alkaloid-induced apoptosis. Our results show the strong apoptotic effects of pteropodine and uncarine F on acute leukaemic lymphoblasts and recommend the alkaloids for further studies in xenograft models.


Assuntos
Alcaloides/uso terapêutico , Unha-de-Gato , Fitoterapia/métodos , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico , Apoptose/efeitos dos fármacos , Western Blotting/métodos , Caspase 9 , Caspases/análise , Proliferação de Células/efeitos dos fármacos , Colágeno Tipo XI/análise , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Fase G1 , Humanos , Alcaloides Indólicos/uso terapêutico , Indóis/uso terapêutico , Oxindóis , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Serpinas/metabolismo , Compostos de Espiro/uso terapêutico , Linfócitos T/metabolismo , Células Tumorais Cultivadas , Proteínas Virais/metabolismo
10.
J Gen Virol ; 86(Pt 12): 3235-3241, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16298968

RESUMO

E7 proteins are major oncoproteins of high-risk human papillomaviruses (HPVs), which play a key role in cervical carcinogenesis. These proteins have been shown to immortalize primary human cells. Due to the absence of antibodies with suitable sensitivity and specificity, little is known about expression of the E7 oncoproteins in naturally infected tissues. Recently, high-level expression of the E7 protein of HPV-16, the most prevalent oncogenic HPV type, was demonstrated in cervical carcinomas by immunohistochemistry; however, approximately 15 additional high-risk HPV types are known to be associated with cervical carcinoma. It is unknown whether the E7 oncoproteins of HPV-18 and -45, the second and third most prevalent HPV types, are expressed in cervical cancers. Using antibodies against HPV-18 and -45 E7 proteins, it is shown here for the first time that the HPV-18 and -45 E7 proteins can be detected in cervical carcinoma biopsies. Together with anti-HPV-16 E7 antibodies, this could create the possibility of detecting E7 oncoproteins in approximately 80 % of all cervical cancers.


Assuntos
Proteínas Oncogênicas Virais/análise , Papillomaviridae/genética , Neoplasias do Colo do Útero/virologia , Anticorpos Antivirais , Biópsia , Western Blotting , Proteínas de Ligação a DNA/análise , Proteínas de Ligação a DNA/imunologia , Feminino , Papillomavirus Humano 18/genética , Humanos , Imuno-Histoquímica , Microscopia Confocal , Microscopia de Fluorescência , Proteínas Oncogênicas Virais/imunologia
11.
Stem Cells ; 21(3): 266-71, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12743321

RESUMO

In phase I and II clinical trials, the deoxycytidine analogue 2',2' difluorodeoxycytidine (dFdC, gemcitabine) has shown promising antitumor activity in leukemia as well as in solid tumors. Preclinical and clinical studies of gemcitabine suggested that myelosuppression was the dose-limiting toxicity. The present investigations were designed to test the effect of continuously administered gemcitabine on the in vitro clonal growth of normal CD34(+) cells isolated from peripheral blood and the promyelocytic cell line, HL-60. For this purpose, CD34(+) and HL-60 cells were cultured in methylcellulose in the continuous presence of 0.1-16 nM of gemcitabine. The results show a dose-dependent inhibition of colony growth of normal as well as leukemic cells. However, HL-60 cells were up to 12-fold more sensitive towards gemcitabine than normal progenitors. For rescue experiments, the natural pyrimidine deoxycytidine (dCyd) was added to CD34(+) and HL-60 cells simultaneously or with delay. Coadministration of 1mM dCyd to separate cultures resulted in complete restoration of colony formation capacity of CD34(+) and HL-60 cells. Delayed addition of 1 mM dCyd after 48 and 72 hours recovered up to 90% and 40%, respectively, of stem cell proliferation, whereas HL-60 cells remained substantially inhibited (4.5% +/- 3.5% versus 0%). Delayed addition after 48 and 72 hours protected about 80% and 50%, respectively, of myelopoietic and erythropoetic colony formation, whereas colony formation obtained from HL-60 cells remained significantly inhibited (9.6% +/- 4.17% versus 0%). These in vitro data suggest that there is a marked difference in the susceptibility of leukemic and normal CD34(+) cells to gemcitabine and that delayed administration of dCyd may further reduce the bone marrow cytotoxicity of gemcitabine without impairing its antitumor effect.


Assuntos
Antígenos CD34/biossíntese , Antineoplásicos/antagonistas & inibidores , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacologia , Desoxicitidina/toxicidade , Células-Tronco Hematopoéticas/efeitos dos fármacos , Leucemia/tratamento farmacológico , Linfócitos T/efeitos dos fármacos , Antígenos CD34/imunologia , Antineoplásicos/toxicidade , Células da Medula Óssea/citologia , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/metabolismo , Divisão Celular/efeitos dos fármacos , Divisão Celular/fisiologia , Técnicas de Cocultura , Citotoxinas/antagonistas & inibidores , Citotoxinas/farmacologia , Desoxicitidina/antagonistas & inibidores , Relação Dose-Resposta a Droga , Esquema de Medicação , Interações Medicamentosas/fisiologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/imunologia , Quimioterapia Combinada , Células HL-60 , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Humanos , Imunossupressores/antagonistas & inibidores , Imunossupressores/toxicidade , Leucemia/imunologia , Leucemia/fisiopatologia , Linfócitos T/imunologia , Gencitabina
12.
Immunobiology ; 207(2): 149-57, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12675273

RESUMO

The effect of 2',2'-difluorodeoxycytidine (dFdC) on in vitro human lymphocyte response was assessed in comparison with that of its major metabolite 2',2'-difluorodeoxyuridine (dFdU). Peripheral blood mononuclear cells (PBMNC) from healthy human volunteers were used for assay of mixed lymphocyte reaction (MLR), blastogenesis and colony forming by PHA. Both substances inhibited mitogen and alloantigen responses of PBMNC in a dose-dependent manner, but dFdU was up to 10,000-fold less potent than its parent compound dFdC. The data indicate that activation by alloantigen is more sensitive to the action of dFdU and dFdC than the response to PHA. Thus, dFdU inhibits MLR-induced response at significantly lower doses than PHA-induced proliferation (IC50 +/- SD, 23.55 +/- 8 microM versus 133.2 +/- 12 microM) (p = 0.0003). dFdC also proved to be about 12.3-fold more potent against alloantigen response compared to PHA-induced proliferation of PBMNC (IC50 +/- SD, 2.28 +/- 0.5 nM versus 28.1 +/- 0.5 nM) (p = 0.0001). To get an insight into the toxic profile of dFdU and dFdC, both substances were additionally tested on the in vitro clonal growth of CD34+ cells. Cells were cultured in methylcellulose in the continuous presence of dFdU and dFdC in doses up to 640 microM and 16 nM, respectively. The results show a marked inhibition of erythroid (BFU-E) and myeloid progenitors (CFU-GM) in a dose-dependent manner, but BFU-E was more sensitive to the action of dFdU and dFdC than CFU-GM (p=0.0001). Compared to T-lymphocytes, however, similar or even higher doses of dFdU and dFdC were required for complete inhibition of colony formation obtained from CD34+ cells. To test the role of deoxycytidine kinase (dCK) in the metabolism of dFdU in comparison to that of dFdC, reversal studies with deoxycytidine (dCyd), the natural substrate of dCK, were performed on dFdU- and dFdC-treated HL-60 cells. The data show that relatively low concentrations of dCyd (10 microM) were sufficient to protect HL-60 cells from cytotoxicity of lethal doses of dFdU (160 microM), whereas 100-fold higher concentrations of deoxycytidine (dCyd) (1 mM) were required for a complete reversal of dFdC-mediated toxicity. This suggests that activation of dFdU is due to its phosphorylation by dCK, but dFdU has low affinity to dCK. These effects of dFdU and dFdC in relation to T-lymphocytes and CD34+ cells suggest their possible use as immunosuppressive agents.


Assuntos
Desoxicitidina/análogos & derivados , Desoxicitidina/farmacologia , Floxuridina/análogos & derivados , Floxuridina/farmacologia , Imunossupressores/farmacologia , Leucócitos Mononucleares/efeitos dos fármacos , Corticosteroides/farmacologia , Antígenos CD34/metabolismo , Desoxicitidina/metabolismo , Desoxicitidina Quinase/metabolismo , Relação Dose-Resposta a Droga , Quimioterapia Combinada , Células HL-60 , Humanos , Leucócitos Mononucleares/imunologia , Ativação Linfocitária/efeitos dos fármacos , Teste de Cultura Mista de Linfócitos , Fito-Hemaglutininas/farmacologia , Células-Tronco/efeitos dos fármacos , Gencitabina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...